alpha-synuclein and Nerve-Degeneration

alpha-synuclein has been researched along with Nerve-Degeneration* in 411 studies

Reviews

83 review(s) available for alpha-synuclein and Nerve-Degeneration

ArticleYear
Proposal for a Biologic Staging System of Parkinson's Disease.
    Journal of Parkinson's disease, 2023, Volume: 13, Issue:3

    The Parkinson's disease (PD) research field has seen the advent of several promising biomarkers and a deeper understanding of the clinical features of the disease from the earliest stages of pathology to manifest disease. Despite progress, a biologically based PD staging system does not exist. Such staging would be a useful framework within which to model the disease, develop and validate biomarkers, guide therapeutic development, and inform clinical trials design. We propose that the presence of aggregated neuronal α-synuclein, dopaminergic neuron dysfunction/degeneration, and clinical signs and symptoms identifies a group of individuals that have Lewy body pathology, which in early stages manifests with what is now referred to as prodromal non-motor features and later stages with the manifestations of PD and related Lewy body diseases as defined by clinical diagnostic criteria. Based on the state of the field, we herein propose a definition and staging of PD based on biology. We present the biologic basis for such a staging system and review key assumptions and evidence that support the proposed approach. We identify gaps in knowledge and delineate crucial research priorities that will inform the ultimate integrated biologic staging system for PD.

    Topics: alpha-Synuclein; Biological Products; Biomarkers; Humans; Lewy Bodies; Lewy Body Disease; Nerve Degeneration; Parkinson Disease; Prodromal Symptoms

2023
Neuropathology of incidental Lewy body & prodromal Parkinson's disease.
    Molecular neurodegeneration, 2023, 05-12, Volume: 18, Issue:1

    Parkinson's disease (PD) is a progressive neurodegenerative disorder associated with a loss of dopaminergic (DA) neurons. Despite symptomatic therapies, there is currently no disease-modifying treatment to halt neuronal loss in PD. A major hurdle for developing and testing such curative therapies results from the fact that most DA neurons are already lost at the time of the clinical diagnosis, rendering them inaccessible to therapy. Understanding the early pathological changes that precede Lewy body pathology (LBP) and cell loss in PD will likely support the identification of novel diagnostic and therapeutic strategies and help to differentiate LBP-dependent and -independent alterations. Several previous studies identified such specific molecular and cellular changes that occur prior to the appearance of Lewy bodies (LBs) in DA neurons, but a concise map of such early disease events is currently missing.. Here, we conducted a literature review to identify and discuss the results of previous studies that investigated cases with incidental Lewy body disease (iLBD), a presumed pathological precursor of PD.. Collectively, our review demonstrates numerous cellular and molecular neuropathological changes occurring prior to the appearance of LBs in DA neurons.. Our review provides the reader with a summary of early pathological events in PD that may support the identification of novel therapeutic and diagnostic targets and aid to the development of disease-modifying strategies in PD.

    Topics: alpha-Synuclein; Humans; Lewy Bodies; Lewy Body Disease; Nerve Degeneration; Neuropathology; Parkinson Disease

2023
Glymphatic System Dysfunction and Sleep Disturbance May Contribute to the Pathogenesis and Progression of Parkinson's Disease.
    International journal of molecular sciences, 2022, Oct-26, Volume: 23, Issue:21

    Parkinson's disease (PD) is a multisystem alpha-synucleinopathic neurodegenerative disease and the most prevalent neurodegenerative disorder after Alzheimer's disease with a high incidence rate in the elderly population. PD is highly multifactorial in etiology and has complex and wide-ranging pathogenic mechanisms. Environmental exposures and genetic predisposition are prominent risk factors. However, current evidence suggests that an intimate link may exist between the risk factor of sleep disturbance and PD pathogenesis. PD is characterized by the pathological hallmarks of alpha-synuclein aggregations and dopaminergic neuron degeneration in the substantia nigra. The loss of dopamine-producing neurons results in both motor and non-motor symptoms, most commonly, bradykinesia, tremor, rigidity, psychiatric disorders, sleep disorders and gastrointestinal problems. Factors that may exacerbate alpha-synuclein accumulation and dopamine neuron loss include neuroinflammation and glymphatic system impairment. Extracellular alpha-synuclein can induce an inflammatory response which can lead to neural cell death and inhibition of neurogenesis. The glymphatic system functions most optimally to remove extracellular brain solutes during sleep and therefore sleep disruption may be a crucial progression factor as well as a risk factor. This literature review interprets and analyses data from experimental and epidemiological studies to determine the recent advances in establishing a relationship between glymphatic system dysfunction, sleep disturbance, and PD pathogenesis and progression. This review addresses current limitations surrounding the ability to affirm a causal link between improved glymphatic clearance by increased sleep quality in PD prevention and management. Furthermore, this review proposes potential therapeutic approaches that could utilize the protective mechanism of sleep, to promote glymphatic clearance that therefore may reduce disease progression as well as symptom severity in PD patients.

    Topics: Aged; alpha-Synuclein; Dopamine; Glymphatic System; Humans; Nerve Degeneration; Neurodegenerative Diseases; Parkinson Disease; Sleep; Sleep Wake Disorders

2022
Pathogenesis of α-Synuclein in Parkinson's Disease: From a Neuron-Glia Crosstalk Perspective.
    International journal of molecular sciences, 2022, Nov-25, Volume: 23, Issue:23

    Parkinson's disease (PD) is a progressive neurodegenerative disorder. The classical behavioral defects of PD patients involve motor symptoms such as bradykinesia, tremor, and rigidity, as well as non-motor symptoms such as anosmia, depression, and cognitive impairment. Pathologically, the progressive loss of dopaminergic (DA) neurons in the substantia nigra (SN) and the accumulation of α-synuclein (α-syn)-composed Lewy bodies (LBs) and Lewy neurites (LNs) are key hallmarks. Glia are more than mere bystanders that simply support neurons, they actively contribute to almost every aspect of neuronal development and function; glial dysregulation has been implicated in a series of neurodegenerative diseases including PD. Importantly, amounting evidence has added glial activation and neuroinflammation as new features of PD onset and progression. Thus, gaining a better understanding of glia, especially neuron-glia crosstalk, will not only provide insight into brain physiology events but also advance our knowledge of PD pathologies. This review addresses the current understanding of α-syn pathogenesis in PD, with a focus on neuron-glia crosstalk. Particularly, the transmission of α-syn between neurons and glia, α-syn-induced glial activation, and feedbacks of glial activation on DA neuron degeneration are thoroughly discussed. In addition, α-syn aggregation, iron deposition, and glial activation in regulating DA neuron ferroptosis in PD are covered. Lastly, we summarize the preclinical and clinical therapies, especially targeting glia, in PD treatments.

    Topics: alpha-Synuclein; Dopaminergic Neurons; Humans; Lewy Bodies; Nerve Degeneration; Parkinson Disease; Substantia Nigra

2022
A historical review of multiple system atrophy with a critical appraisal of cellular and animal models.
    Journal of neural transmission (Vienna, Austria : 1996), 2021, Volume: 128, Issue:10

    Multiple system atrophy (MSA) is a progressive neurodegenerative disorder characterized by striatonigral degeneration (SND), olivopontocerebellar atrophy (OPCA), and dysautonomia with cerebellar ataxia or parkinsonian motor features. Isolated autonomic dysfunction with predominant genitourinary dysfunction and orthostatic hypotension and REM sleep behavior disorder are common characteristics of a prodromal phase, which may occur years prior to motor-symptom onset. MSA is a unique synucleinopathy, in which alpha-synuclein (aSyn) accumulates and forms insoluble inclusions in the cytoplasm of oligodendrocytes, termed glial cytoplasmic inclusions (GCIs). The origin of, and precise mechanism by which aSyn accumulates in MSA are unknown, and, therefore, disease-modifying therapies to halt or slow the progression of MSA are currently unavailable. For these reasons, much focus in the field is concerned with deciphering the complex neuropathological mechanisms by which MSA begins and progresses through the course of the disease. This review focuses on the history, etiopathogenesis, neuropathology, as well as cell and animal models of MSA.

    Topics: alpha-Synuclein; Animals; Inclusion Bodies; Models, Animal; Multiple System Atrophy; Nerve Degeneration

2021
Unzipping the Secrets of Amyloid Disassembly by the Human Disaggregase.
    Cells, 2021, 10-14, Volume: 10, Issue:10

    Neurodegenerative diseases (NDs) are increasingly positioned as leading causes of global deaths. The accelerated aging of the population and its strong relationship with neurodegeneration forecast these pathologies as a huge global health problem in the upcoming years. In this scenario, there is an urgent need for understanding the basic molecular mechanisms associated with such diseases. A major molecular hallmark of most NDs is the accumulation of insoluble and toxic protein aggregates, known as amyloids, in extracellular or intracellular deposits. Here, we review the current knowledge on how molecular chaperones, and more specifically a ternary protein complex referred to as the human disaggregase, deals with amyloids. This machinery, composed of the constitutive Hsp70 (Hsc70), the class B J-protein DnaJB1 and the nucleotide exchange factor Apg2 (Hsp110), disassembles amyloids of α-synuclein implicated in Parkinson's disease as well as of other disease-associated proteins such as tau and huntingtin. We highlight recent studies that have led to the dissection of the mechanism used by this chaperone system to perform its disaggregase activity. We also discuss whether this chaperone-mediated disassembly mechanism could be used to solubilize other amyloidogenic substrates. Finally, we evaluate the implications of the chaperone system in amyloid clearance and associated toxicity, which could be critical for the development of new therapies.

    Topics: alpha-Synuclein; Amyloid; Humans; Models, Biological; Molecular Chaperones; Nerve Degeneration; Protein Aggregates

2021
Mechanisms of Neurodegeneration in Various Forms of Parkinsonism-Similarities and Differences.
    Cells, 2021, 03-16, Volume: 10, Issue:3

    Parkinson's disease (PD), dementia with Lewy body (DLB), progressive supranuclear palsy (PSP), corticobasal degeneration (CBD) and multiple system atrophy (MSA) belong to a group of neurodegenerative diseases called parkinsonian syndromes. They share several clinical, neuropathological and genetic features. Neurodegenerative diseases are characterized by the progressive dysfunction of specific populations of neurons, determining clinical presentation. Neuronal loss is associated with extra- and intracellular accumulation of misfolded proteins. The parkinsonian diseases affect distinct areas of the brain. PD and MSA belong to a group of synucleinopathies that are characterized by the presence of fibrillary aggregates of α-synuclein protein in the cytoplasm of selected populations of neurons and glial cells. PSP is a tauopathy associated with the pathological aggregation of the microtubule associated tau protein. Although PD is common in the world's aging population and has been extensively studied, the exact mechanisms of the neurodegeneration are still not fully understood. Growing evidence indicates that parkinsonian disorders to some extent share a genetic background, with two key components identified so far: the microtubule associated tau protein gene (

    Topics: alpha-Synuclein; Animals; Brain; Genetic Predisposition to Disease; Humans; Mitochondria; Mitophagy; Nerve Degeneration; Neurons; Oxidative Stress; Parkinsonian Disorders; Phenotype; Risk Factors; tau Proteins

2021
The Contribution of Microglia to Neuroinflammation in Parkinson's Disease.
    International journal of molecular sciences, 2021, Apr-28, Volume: 22, Issue:9

    With the world's population ageing, the incidence of Parkinson's disease (PD) is on the rise. In recent years, inflammatory processes have emerged as prominent contributors to the pathology of PD. There is great evidence that microglia have a significant neuroprotective role, and that impaired and over activated microglial phenotypes are present in brains of PD patients. Thereby, PD progression is potentially driven by a vicious cycle between dying neurons and microglia through the instigation of oxidative stress, mitophagy and autophagy dysfunctions, a-synuclein accumulation, and pro-inflammatory cytokine release. Hence, investigating the involvement of microglia is of great importance for future research and treatment of PD. The purpose of this review is to highlight recent findings concerning the microglia-neuronal interplay in PD with a focus on human postmortem immunohistochemistry and single-cell studies, their relation to animal and iPSC-derived models, newly emerging technologies, and the resulting potential of new anti-inflammatory therapies for PD.

    Topics: alpha-Synuclein; Animals; Brain; Cytokines; Disease Models, Animal; Dopamine; Dopaminergic Neurons; Humans; Inflammation; Microglia; Nerve Degeneration; Neuroimmunomodulation; Neurons; Neuroprotection; Oxidative Stress; Parkinson Disease

2021
The Intersection of Parkinson's Disease, Viral Infections, and COVID-19.
    Molecular neurobiology, 2021, Volume: 58, Issue:9

    The Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), the causative agent of human COVID-19, not only causes flu-like symptoms and gut microbiome complications but a large number of infected individuals also experience a host of neurological symptoms including loss of smell and taste, seizures, difficulty concentrating, decreased alertness, and brain inflammation. Although SARS-CoV-2 infections are not more prevalent in Parkinson's disease patients, a higher mortality rate has been reported not only associated with older age and longer disease duration, but also through several mechanisms, such as interactions with the brain dopaminergic system and through systemic inflammatory responses. Indeed, a number of the neurological symptoms seen in COVID-19 patients, as well as the alterations in the gut microbiome, are also prevalent in patients with Parkinson's disease. Furthermore, biochemical pathways such as oxidative stress, inflammation, and protein aggregation have shared commonalities between Parkinson's disease and COVID-19 disease progression. In this review, we describe and compare the numerous similarities and intersections between neurodegeneration in Parkinson's disease and RNA viral infections, emphasizing the current SARS-CoV-2 global health crisis.

    Topics: Aged; alpha-Synuclein; Cognition Disorders; COVID-19; Cytokines; Diet; Disease Progression; Dysbiosis; Gastrointestinal Microbiome; Humans; Inflammation; Metals, Heavy; Models, Neurological; Nerve Degeneration; Olfactory Bulb; Oxidative Stress; Parkinson Disease; Practice Guidelines as Topic; Protein Aggregation, Pathological; Reactive Oxygen Species; RNA Virus Infections; SARS-CoV-2; Sensation Disorders

2021
Alpha-Synuclein and Mitochondrial Dysfunction in Parkinson's Disease: The Emerging Role of VDAC.
    Biomolecules, 2021, 05-11, Volume: 11, Issue:5

    Alpha-Synuclein (αSyn) is a protein whose function is still debated, as well as its role in modulation of mitochondrial function in both physiological and pathological conditions. Mitochondrial porins or Voltage-Dependent Anion Channel (VDAC) proteins are the main gates for ADP/ATP and various substrates towards the organelle. Furthermore, they act as a mitochondrial hub for many cytosolic proteins, including αSyn. This review analyzes the main aspects of αSyn-mitochondria interaction, focusing on the role of VDAC and its emerging involvement in the pathological processes.

    Topics: alpha-Synuclein; Animals; Cytosol; Humans; Mitochondria; Nerve Degeneration; Parkinson Disease; Voltage-Dependent Anion Channels

2021
The Catecholaldehyde Hypothesis for the Pathogenesis of Catecholaminergic Neurodegeneration: What We Know and What We Do Not Know.
    International journal of molecular sciences, 2021, Jun-01, Volume: 22, Issue:11

    3,4-Dihydroxyphenylacetaldehyde (DOPAL) is the focus of the catecholaldehyde hypothesis for the pathogenesis of Parkinson's disease and other Lewy body diseases. The catecholaldehyde is produced via oxidative deamination catalyzed by monoamine oxidase (MAO) acting on cytoplasmic dopamine. DOPAL is autotoxic, in that it can harm the same cells in which it is produced. Normally, DOPAL is detoxified by aldehyde dehydrogenase (ALDH)-mediated conversion to 3,4-dihydroxyphenylacetic acid (DOPAC), which rapidly exits the neurons. Genetic, environmental, or drug-induced manipulations of ALDH that build up DOPAL promote catecholaminergic neurodegeneration. A concept derived from the catecholaldehyde hypothesis imputes deleterious interactions between DOPAL and the protein alpha-synuclein (αS), a major component of Lewy bodies. DOPAL potently oligomerizes αS, and αS oligomers impede vesicular and mitochondrial functions, shifting the fate of cytoplasmic dopamine toward the MAO-catalyzed formation of DOPAL-destabilizing vicious cycles. Direct and indirect effects of DOPAL and of DOPAL-induced misfolded proteins could "freeze" intraneuronal reactions, plasticity of which is required for neuronal homeostasis. The extent to which DOPAL toxicity is mediated by interactions with αS, and vice versa, is poorly understood. Because of numerous secondary effects such as augmented spontaneous oxidation of dopamine by MAO inhibition, there has been insufficient testing of the catecholaldehyde hypothesis in animal models. The clinical pathophysiological significance of genetics, emotional stress, environmental agents, and interactions with numerous proteins relevant to the catecholaldehyde hypothesis are matters for future research. The imposing complexity of intraneuronal catecholamine metabolism seems to require a computational modeling approach to elucidate clinical pathogenetic mechanisms and devise pathophysiology-based, individualized treatments.

    Topics: Aldehyde Dehydrogenase; Aldehydes; alpha-Synuclein; Animals; Catechols; Dopamine; Humans; Monoamine Oxidase; Monoamine Oxidase Inhibitors; Nerve Degeneration; Neurons; Oxidation-Reduction; Parkinson Disease; PC12 Cells; Rats

2021
PIAS2-mediated blockade of IFN-β signaling: a basis for sporadic Parkinson disease dementia.
    Molecular psychiatry, 2021, Volume: 26, Issue:10

    Topics: alpha-Synuclein; Animals; Dementia; Dopaminergic Neurons; Humans; Interferon-beta; Mice; Mice, Knockout; Nerve Degeneration; Parkinson Disease; Protein Inhibitors of Activated STAT; Signal Transduction

2021
Parkinson's Disease-Related Genes and Lipid Alteration.
    International journal of molecular sciences, 2021, Jul-16, Volume: 22, Issue:14

    Parkinson's disease (PD) is a complex and progressive neurodegenerative disorder with a prevalence of approximately 0.5-1% among those aged 65-70 years. Although most of its clinical manifestations are due to a loss of dopaminergic neurons, the PD etiology is largely unknown. PD is caused by a combination of genetic and environmental factors, and the exact interplay between genes and the environment is still debated. Several biological processes have been implicated in PD, including mitochondrial or lysosomal dysfunctions, alteration in protein clearance, and neuroinflammation, but a common molecular mechanism connecting the different cellular alterations remains incompletely understood. Accumulating evidence underlines a significant role of lipids in the pathological pathways leading to PD. Beside the well-described lipid alteration in idiopathic PD, this review summarizes the several lipid alterations observed in experimental models expressing PD-related genes and suggests a possible scenario in relationship to the molecular mechanisms of neuronal toxicity. PD could be considered a lipid-induced proteinopathy, where alteration in lipid composition or metabolism could induce protein alteration-for instance, alpha-synuclein accumulation-and finally neuronal death.

    Topics: alpha-Synuclein; Dopaminergic Neurons; Glucosylceramidase; Group VI Phospholipases A2; Humans; Leucine-Rich Repeat Serine-Threonine Protein Kinase-2; Lipid Metabolism; Lipids; Lysosomes; Mitochondria; Nerve Degeneration; Parkinson Disease; Protein Kinases; Ubiquitin-Protein Ligases

2021
Alpha-Synuclein and LRRK2 in Synaptic Autophagy: Linking Early Dysfunction to Late-Stage Pathology in Parkinson's Disease.
    Cells, 2020, 04-30, Volume: 9, Issue:5

    The lack of effective disease-modifying strategies is the major unmet clinical need in Parkinson´s disease. Several experimental approaches have attempted to validate cellular targets and processes. Of these, autophagy has received considerable attention in the last 20 years due to its involvement in the clearance of pathologic protein aggregates and maintenance of neuronal homeostasis. However, this strategy mainly addresses a very late stage of the disease, when neuropathology and neurodegeneration have likely "tipped over the edge" and disease modification is extremely difficult. Very recently, autophagy has been demonstrated to modulate synaptic activity, a process distinct from its catabolic function. Abnormalities in synaptic transmission are an early event in neurodegeneration with Leucine-Rich Repeat Kinase 2 (LRRK2) and alpha-synuclein strongly implicated. In this review, we analyzed these processes separately and then discussed the unification of these biomolecular fields with the aim of reconstructing a potential "molecular timeline" of disease onset and progression. We postulate that the elucidation of these pathogenic mechanisms will form a critical basis for the design of novel, effective disease-modifying therapies that could be applied early in the disease process.

    Topics: alpha-Synuclein; Autophagy; Disease Progression; Humans; Leucine-Rich Repeat Serine-Threonine Protein Kinase-2; Nerve Degeneration; Neurons; Parkinson Disease; Synapses

2020
Neurodegeneration and Inflammation-An Interesting Interplay in Parkinson's Disease.
    International journal of molecular sciences, 2020, Nov-10, Volume: 21, Issue:22

    Parkinson's disease (PD) is a neurodegenerative disorder, caused by, so far, unknown pathogenetic mechanisms. There is no doubt that pro-inflammatory immune-mediated mechanisms are pivotal to the pathogenicity and progression of the disease. In this review, we highlight the binary role of microglia activation in the pathophysiology of the disorder, both neuroprotective and neuromodulatory. We present how the expression of several cytokines implicated in dopaminergic neurons (DA) degeneration could be used as biomarkers for PD. Viral infections have been studied and correlated to the disease progression, usually operating as trigger factors for the inflammatory process. The gut-brain axis and the possible contribution of the peripheral bowel inflammation to neuronal death, mainly dopaminergic neurons, seems to be a main contributor of brain neuroinflammation. The role of the immune system has also been analyzed implicating a-synuclein in the activation of innate and adaptive immunity. We also discuss therapeutic approaches concerning PD and neuroinflammation, which have been studied in experimental and in vitro models and data stemming from epidemiological studies.

    Topics: alpha-Synuclein; Animals; Autoimmunity; Biomarkers; Cytokines; Dopaminergic Neurons; Humans; Inflammation; Microglia; Nerve Degeneration; Parkinson Disease; Parkinsonian Disorders; Signal Transduction; Virus Diseases

2020
Neuron-Astrocyte Interactions in Parkinson's Disease.
    Cells, 2020, 12-07, Volume: 9, Issue:12

    Parkinson's disease (PD) is the second most common neurodegenerative disease. PD patients exhibit motor symptoms such as akinesia/bradykinesia, tremor, rigidity, and postural instability due to a loss of nigrostriatal dopaminergic neurons. Although the pathogenesis in sporadic PD remains unknown, there is a consensus on the involvement of non-neuronal cells in the progression of PD pathology. Astrocytes are the most numerous glial cells in the central nervous system. Normally, astrocytes protect neurons by releasing neurotrophic factors, producing antioxidants, and disposing of neuronal waste products. However, in pathological situations, astrocytes are known to produce inflammatory cytokines. In addition, various studies have reported that astrocyte dysfunction also leads to neurodegeneration in PD. In this article, we summarize the interaction of astrocytes and dopaminergic neurons, review the pathogenic role of astrocytes in PD, and discuss therapeutic strategies for the prevention of dopaminergic neurodegeneration. This review highlights neuron-astrocyte interaction as a target for the development of disease-modifying drugs for PD in the future.

    Topics: alpha-Synuclein; Animals; Antioxidants; Astrocytes; Disease Progression; Dopamine; Dopaminergic Neurons; Humans; Inflammation; Mitochondria; Nerve Degeneration; Neuroglia; Neurons; Neuroprotection; Oxidative Stress; Parkinson Disease; Signal Transduction

2020
The unlikely partnership between LRRK2 and α-synuclein in Parkinson's disease.
    The European journal of neuroscience, 2019, Volume: 49, Issue:3

    Our understanding of the mechanisms underlying Parkinson's disease, the once archetypical nongenetic neurogenerative disorder, has dramatically increased with the identification of α-synuclein and LRRK2 pathogenic mutations. While α-synuclein protein composes the aggregates that can spread through much of the brain in disease, LRRK2 encodes a multidomain dual-enzyme distinct from any other protein linked to neurodegeneration. In this review, we discuss emergent datasets from multiple model systems that suggest these unlikely partners do interact in important ways in disease, both within cells that express both LRRK2 and α-synuclein as well as through more indirect pathways that might involve neuroinflammation. Although the link between LRRK2 and disease can be understood in part through LRRK2 kinase activity (phosphotransferase activity), α-synuclein toxicity is multilayered and plausibly interacts with LRRK2 kinase activity in several ways. We discuss common protein interactors like 14-3-3s that may regulate α-synuclein and LRRK2 in disease. Finally, we examine cellular pathways and outcomes common to both mutant α-synuclein expression and LRRK2 activity and points of intersection. Understanding the interplay between these two unlikely partners in disease may provide new therapeutic avenues for PD.

    Topics: alpha-Synuclein; Animals; Brain; Humans; Leucine-Rich Repeat Serine-Threonine Protein Kinase-2; Mutation; Nerve Degeneration; Neurons; Parkinson Disease

2019
The usual suspects, dopamine and alpha-synuclein, conspire to cause neurodegeneration.
    Movement disorders : official journal of the Movement Disorder Society, 2019, Volume: 34, Issue:2

    Parkinson's disease (PD) is primarily a movement disorder driven by the loss of dopamine-producing neurons in the substantia nigra (SN). Early identification of the oxidative properties of dopamine implicated it as a potential source of oxidative stress in PD, yet few studies have investigated dopamine neurotoxicity in vivo. The discovery of PD-causing mutations in α-synuclein and the presence of aggregated α-synuclein in the hallmark Lewy body pathology of PD revealed another important player. Despite extensive efforts, the precise role of α-synuclein aggregation in neurodegeneration remains unclear. We recently manipulated both dopamine levels and α-synuclein expression in aged mice and found that only the combination of these 2 factors caused progressive neurodegeneration of the SN and an associated motor deficit. Dopamine modified α-synuclein aggregation in the SN, resulting in greater abundance of α-synuclein oligomers and unique dopamine-induced oligomeric conformations. Furthermore, disruption of the dopamine-α-synuclein interaction rescued dopaminergic neurons from degeneration in transgenic Caenorhabditis elegans models. In this Perspective, we discuss these findings in the context of known α-synuclein and dopamine biology, review the evidence for α-synuclein oligomer toxicity and potential mechanisms, and discuss therapeutic implications. © 2019 International Parkinson and Movement Disorder Society.

    Topics: alpha-Synuclein; Animals; Dopamine; Dopaminergic Neurons; Humans; Nerve Degeneration; Oxidative Stress; Parkinsonian Disorders

2019
Lipid and immune abnormalities causing age-dependent neurodegeneration and Parkinson's disease.
    Journal of neuroinflammation, 2019, Jul-22, Volume: 16, Issue:1

    This article describes pathogenic concepts and factors, in particular glycolipid abnormalities, that create cell dysfunction and synaptic loss in neurodegenerative diseases. By phenocopying lysosomal storage disorders, such as Gaucher disease and related disorders, age- and dose-dependent changes in glycolipid cell metabolism can lead to Parkinson's disease and related dementias. Recent results show that perturbation of sphingolipid metabolism can precede or is a part of abnormal protein handling in both genetic and idiopathic Parkinson's disease and Lewy body dementia. In aging and genetic predisposition with lipid disturbance, α-synuclein's normal vesicular and synaptic role may be detrimentally shifted toward accommodating and binding such lipids. Specific neuronal glycolipid, protein, and vesicular interactions create potential pathophysiology that is amplified by astroglial and microglial immune mechanisms resulting in neurodegeneration. This perspective provides a new logic for therapeutic interventions that do not focus on protein aggregation, but rather provides a guide to the complex biology and the common sequence of events that lead to age-dependent neurodegenerative disorders.

    Topics: alpha-Synuclein; Animals; Brain; Humans; Inflammation; Nerve Degeneration; Neurons; Parkinson Disease; tau Proteins

2019
Neurodegeneration and the ordered assembly of α-synuclein.
    Cell and tissue research, 2018, Volume: 373, Issue:1

    In 2017, it was 200 years since James Parkinson published 'An Essay on the Shaking Palsy' and 20 years since α-synuclein aggregation came to the fore. In 1998, multiple system atrophy joined Parkinson's disease and dementia with Lewy bodies as the third major synucleinopathy. Here, we describe the work that led to the identification of α-synuclein in Lewy bodies, Lewy neurites and Papp-Lantos bodies. We also review some of the findings reported since 1997.

    Topics: alpha-Synuclein; Amino Acid Sequence; Animals; Humans; Lewy Bodies; Nerve Degeneration; Protein Aggregates

2018
A Focus on the Beneficial Effects of Alpha Synuclein and a Re-Appraisal of Synucleinopathies.
    Current protein & peptide science, 2018, Volume: 19, Issue:6

    Alpha synuclein (α-syn) belongs to a class of proteins which are commonly considered to play a detrimental role in neuronal survival. This assumption is based on the occurrence of a severe neuronal degeneration in patients carrying a multiplication of the α-syn gene (SNCA) and in a variety of experimental models, where overexpression of α-syn leads to cell death and neurological impairment. In these conditions, a higher amount of normally structured α-syn produces a damage, which is even worse compared with that produced by α-syn owning an abnormal structure (as occurring following point gene mutations). In line with this, knocking out the expression of α-syn is reported to protect from specific neurotoxins such as 1-methyl, 4-phenyl 1,2,3,6-tetrahydropyridine (MPTP). In the present review we briefly discuss these well-known detrimental effects but we focus on findings showing that, in specific conditions α-syn is beneficial for cell survival. This occurs during methamphetamine intoxication which is counteracted by endogenous α-syn. Similarly, the dysfunction of the chaperone cysteine-string protein- alpha leads to cell pathology which is counteracted by over-expressing α-syn. In line with this, an increased expression of α-syn protects against oxidative damage produced by dopamine. Remarkably, when the lack of α-syn is combined with a depletion of β- and γ- synucleins, alterations in brain structure and function occur. This review tries to balance the evidence showing a beneficial effect with the bulk of data reporting a detrimental effect of endogenous α-syn. The specific role of α-syn as a chaperone protein is discussed to explain such a dual effect.

    Topics: alpha-Synuclein; Animals; Apoptosis; Brain; Gene Expression; Humans; Nerve Degeneration; Neurons; Neuroprotection; Oxidative Stress; Signal Transduction

2018
Modeling Parkinson's Disease in C. elegans.
    Journal of Parkinson's disease, 2018, Volume: 8, Issue:1

    Parkinson's disease (PD) is an adult onset neurodegenerative disease that is characterized by selective degeneration of neurons primarily in the substantia nigra. At present, the pathogenesis of PD is incompletely understood and there are no neuroprotective treatments available. Accurate animal models of PD provide the opportunity to elucidate disease mechanisms and identify therapeutic targets. This review focuses on C. elegans models of PD, including both genetic and toxicant models. This microscopic worm offers several advantages for the study of PD including ease of genetic manipulation, ability to complete experiments rapidly, low cost, and ability to perform large scale screens for disease modifiers. A number of C. elegans models of PD have been generated including transgenic worms that express α-synuclein or LRRK2, and worms with deletions in PRKN/pdr-1, PINK1/pink-1, DJ-1/djr-1.1/djr-1.2 and ATP13A2/catp-6. These worms have been shown to exhibit multiple phenotypic deficits including the loss of dopamine neurons, disruption of dopamine-dependent behaviors, increased sensitivity to stress, age-dependent aggregation, and deficits in movement. As a result, these phenotypes can be used as outcome measures to gain insight into disease pathogenesis and to identify disease modifiers. In this way, C. elegans can be used as an experimental tool to elucidate mechanisms involved in PD and to find novel therapeutic targets that can subsequently be validated in other models.

    Topics: alpha-Synuclein; Animals; Animals, Genetically Modified; Antiparkinson Agents; Caenorhabditis elegans; Caenorhabditis elegans Proteins; Disease Models, Animal; Dopaminergic Neurons; Drug Evaluation, Preclinical; Feeding Behavior; Gene-Environment Interaction; Genes, Reporter; Humans; Mitochondria; Movement Disorders; Nerve Degeneration; Neurotoxins; Parkinsonian Disorders; Phenotype; Protein Aggregation, Pathological; Recombinant Fusion Proteins; RNA Interference; Species Specificity

2018
The dopamine transporter: An unrecognized nexus for dysfunctional peripheral immunity and signaling in Parkinson's Disease.
    Brain, behavior, and immunity, 2018, Volume: 70

    The second-most common neurodegenerative disease, Parkinson's Disease (PD) has three hallmarks: dysfunctional dopamine transmission due, at least in part, to dopamine neuron degeneration; intracellular inclusions of α-synuclein aggregates; and neuroinflammation. The origin and interplay of these features remains a puzzle, as does the underlying mechanism of PD pathogenesis and progression. When viewed in the context of neuroimmunology, dopamine also plays a role in regulating peripheral immune cells. Intriguingly, plasma dopamine levels are altered in PD, suggesting collateral dysregulation of peripheral dopamine transmission. The dopamine transporter (DAT), the main regulator of dopaminergic tone in the CNS, is known to exist in lymphocytes and monocytes/macrophages, but little is known about peripheral DAT biology or how DAT regulates the dopaminergic tone, much less how peripheral DAT alters immune function. Our review is guided by the hypothesis that dysfunctional peripheral dopamine signaling might be linked to the dysfunctional immune responses in PD and thereby suggests a potential bidirectional communication between central and peripheral dopamine systems. This review seeks to foster new perspectives concerning PD pathogenesis and progression.

    Topics: alpha-Synuclein; Dopamine; Dopamine Plasma Membrane Transport Proteins; Dopaminergic Neurons; Humans; Lymphocytes; Macrophages; Monocytes; Nerve Degeneration; Neurodegenerative Diseases; Parkinson Disease; Signal Transduction

2018
Evidence for dopaminergic axonal degeneration as an early pathological process in Parkinson's disease.
    Parkinsonism & related disorders, 2018, Volume: 56

    Parkinson's disease is a common neurodegenerative disorder presenting with a variety of motor and non-motor symptoms. The motor symptoms manifest as a result of the progressive degeneration of midbrain dopaminergic neurons. The axons of these neurons project to the striatum as the nigrostriatal pathway, which is a crucial part of the basal ganglia circuitry controlling movement. In addition to the neuronal degeneration, abnormal intraneuronal α-synuclein protein inclusions called Lewy bodies and Lewy neurites increase in number and spread throughout the nervous system as the disease progresses. While the loss of midbrain dopaminergic neurons is well-established as being central to motor symptoms, there is an increasing focus on the timing of nigrostriatal degeneration, with preclinical evidence suggesting that early axonal degeneration may play a key role in the early stages of Parkinson's disease. Here we review recent evidence for early midbrain dopaminergic axonal degeneration in patients with Parkinson's disease, and explore the potential role of α-synuclein accumulation in this process, with a focus on studies in human populations at the imaging, post-mortem, cellular and molecular levels. Finally, we discuss the implications of this for neurotrophic factor therapies for Parkinson's disease.

    Topics: alpha-Synuclein; Animals; Axons; Dopaminergic Neurons; Early Diagnosis; Humans; Nerve Degeneration; Parkinson Disease

2018
Interaction of LRRK2 and α-Synuclein in Parkinson's Disease.
    Advances in neurobiology, 2017, Volume: 14

    Parkinson's disease (PD) is a progressively debilitating neurodegenerative syndrome. It is best described as a movement disorder characterized by motor dysfunctions, progressive degeneration of dopaminergic neurons of the substantia nigra pars compacta, and abnormal intraneuronal protein aggregates, named Lewy bodies and Lewy neurites. Nevertheless, knowledge of the molecular events leading to this pathophysiology is incomplete. To date, only mutations in the α-synuclein and LRRK2-encoding genes have been associated with typical findings of clinical and pathologic PD. LRRK2 appears to have a central role in the pathogenesis of PD as it is associated with α-synuclein pathology and other proteins implicated in neurodegeneration. Thus, LRRK2 dysfunction may influence the accumulation of α-synuclein and its pathology through diverse pathomechanisms altering cellular functions and signaling pathways, including immune system, autophagy, vesicle trafficking, and retromer complex modulation. Consequently, development of novel LRRK2 inhibitors can be justified to treat the neurodegeneration associated with abnormal α-synuclein accumulation.

    Topics: alpha-Synuclein; Humans; Leucine-Rich Repeat Serine-Threonine Protein Kinase-2; Mutation; Nerve Degeneration; Parkinson Disease

2017
The implication of neuronimmunoendocrine (NIE) modulatory network in the pathophysiologic process of Parkinson's disease.
    Cellular and molecular life sciences : CMLS, 2017, Volume: 74, Issue:20

    Parkinson's disease (PD) is a progressive neurodegenerative disorder implicitly marked by the substantia nigra dopaminergic neuron degeneration and explicitly characterized by the motor and non-motor symptom complexes. Apart from the nigrostriatal dopamine depletion, the immune and endocrine study findings are also frequently reported, which, in fact, have helped to broaden the symptom spectrum and better explain the pathogenesis and progression of PD. Nevertheless, based on the neural, immune, and endocrine findings presented above, it is still difficult to fully recapitulate the pathophysiologic process of PD. Therefore, here, in this review, we have proposed the neuroimmunoendocrine (NIE) modulatory network in PD, aiming to achieve a more comprehensive interpretation of the pathogenesis and progression of this disease. As a matter of fact, in addition to the classical motor symptoms, NIE modulatory network can also underlie the non-motor symptoms such as gastrointestinal, neuropsychiatric, circadian rhythm, and sleep disorders in PD. Moreover, the dopamine (DA)-melatonin imbalance in the retino-diencephalic/mesencephalic-pineal axis also provides an alternative explanation for the motor complications in the process of DA replacement therapy. In conclusion, the NIE network can be expected to deepen our understanding and facilitate the multi-dimensional management and therapy of PD in future clinical practice.

    Topics: alpha-Synuclein; Animals; Circadian Clocks; Dopamine; Dopaminergic Neurons; Genetic Predisposition to Disease; Humans; Hypothalamus; Inflammation; Melatonin; Nerve Degeneration; Parkinson Disease; Receptors, Cytoplasmic and Nuclear; Receptors, Dopamine; Weight Loss

2017
Protein aggregation and neurodegeneration in prototypical neurodegenerative diseases: Examples of amyloidopathies, tauopathies and synucleinopathies.
    Progress in neurobiology, 2017, Volume: 155

    Alzheimer's and Parkinson's diseases are the most prevalent neurodegenerative diseases that generate important health-related direct and indirect socio-economic costs. They are characterized by severe neuronal losses in several disease-specific brain regions associated with deposits of aggregated proteins. In Alzheimer's disease, β-amyloid peptide-containing plaques and intraneuronal neurofibrillary tangles composed of hyperphosphorylated microtubule-associated protein tau are the two main neuropathological lesions, while Parkinson's disease is defined by the presence of Lewy Bodies that are intraneuronal proteinaceous cytoplasmic inclusions. α-Synuclein has been identified as a major protein component of Lewy Bodies and heavily implicated in the pathogenesis of Parkinson's disease. In the past few years, evidence has emerged to explain how these aggregate-prone proteins can undergo spontaneous self-aggregation, propagate from cell to cell, and mediate neurotoxicity. Current research now indicates that oligomeric forms are probably the toxic species. This article discusses recent progress in the understanding of the pathogenesis of these diseases, with a focus on the underlying mechanisms of protein aggregation, and emphasizes the pathophysiological molecular mechanisms leading to cellular toxicity. Finally, we present the putative direct link between β-amyloid peptide and tau in causing toxicity in Alzheimer's disease as well as α-synuclein in Parkinson's disease, along with some of the most promising therapeutic strategies currently in development for those incurable neurodegenerative disorders.

    Topics: alpha-Synuclein; Alzheimer Disease; Animals; Humans; Nerve Degeneration; Neurodegenerative Diseases; Parkinson Disease; Protein Aggregation, Pathological; Tauopathies

2017
α-Synuclein-carrying extracellular vesicles in Parkinson's disease: deadly transmitters.
    Acta neurologica Belgica, 2017, Volume: 117, Issue:1

    Exosomes play a key role in delivery of various biological material and complex signals from one cell to another at long distances. These small extracellular vehicles are involved in mediating multiple physiological and pathogenic processes. In neurodegenerative diseases such as Parkinson's disease (PD), exosomes contribute to disease propagation through transferring misfolded proteins from affected cells to normal cells. In PD, progressive degeneration of neurons arises from the extensive accumulation of toxic forms of α-synuclein in the cytoplasm. α-Synuclein could exist in several forms, some of which (i.e., oligomeric and polymeric forms) are cytotoxic. Neuron-derived exosomes were found to transfer α-synuclein toxic forms between neuronal and non-neuronal cells (such as astrocytes and microglia) thereby contributing to PD spreading. Deposition of α-synuclein in glial cells induces inflammation that could be further propagated to other glial cells and neurons. Neuroinflammation promotes degeneration of neurons and aggravates the pathogenesis of PD.

    Topics: alpha-Synuclein; Animals; Exosomes; Humans; Nerve Degeneration; Parkinson Disease

2017
Glia and alpha-synuclein in neurodegeneration: A complex interaction.
    Neurobiology of disease, 2016, Volume: 85

    α-Synucleinopathies (ASP) comprise adult-onset, progressive neurodegenerative disorders such as Parkinson's disease (PD), dementia with Lewy bodies (DLB) and multiple system atrophy (MSA) that are characterized by α-synuclein (AS) aggregates in neurons or glia. PD and DLB feature neuronal AS-positive inclusions termed Lewy bodies (LB) whereas glial cytoplasmic inclusions (GCIs, Papp-Lantos bodies) are recognized as the defining hallmark of MSA. Furthermore, AS-positive cytoplasmic aggregates may also be seen in astroglial cells of PD/DLB and MSA brains. The glial AS-inclusions appear to trigger reduced trophic support resulting in neuronal loss. Moreover, microgliosis and astrogliosis can be found throughout the neurodegenerative brain and both are key players in the initiation and progression of ASP. In this review, we will highlight AS-dependent alterations of glial function and their impact on neuronal vulnerability thereby providing a detailed summary on the multifaceted role of glia in ASP.

    Topics: alpha-Synuclein; Animals; Humans; Nerve Degeneration; Neurodegenerative Diseases; Neuroglia

2016
α-Synuclein in Extracellular Vesicles: Functional Implications and Diagnostic Opportunities.
    Cellular and molecular neurobiology, 2016, Volume: 36, Issue:3

    Fibrillar inclusions of intraneuronal α-synuclein can be detected in certain brain areas from patients with Parkinson's disease (PD) and other disorders with Lewy body pathology. These insoluble protein aggregates do not themselves appear to have a prominent neurotoxic effect, whereas various α-synuclein oligomers appear harmful. Although it is incompletely known how the prefibrillar species may be pathogenic, they have been detected both within and on the outside of exosomes and other extracellular vesicles (EVs), suggesting that such structures may mediate toxic α-synuclein propagation between neurons. Vesicular transfer of α-synuclein may thereby contribute to the hierarchical spreading of pathology seen in the PD brain. Although the regulation of α-synuclein release via EVs is not understood, data suggest that it may involve other PD-related molecules, such as LRRK2 and ATP13A2. Moreover, new evidence indicates that CNS-derived EVs in plasma have the potential to serve as biomarkers for diagnostic purposes. In a recent study, levels of α-synuclein were found to be increased in L1CAM-positive vesicles isolated from plasma of PD patients compared to healthy controls, and follow-up studies will reveal whether α-synuclein in EVs could be developed as a future disease biomarker. Preferentially, toxic prefibrillar α-synuclein oligomers should then be targeted as a biomarker-as evidence suggests that they reflect the disease process more closely than total α-synuclein content. In such studies, it will be essential to adopt stringent EV isolation protocols in order to avoid contamination from the abundant pool of free plasma α-synuclein in different aggregational states.

    Topics: alpha-Synuclein; Animals; Central Nervous System; Extracellular Vesicles; Humans; Nerve Degeneration; Parkinson Disease; Protein Multimerization

2016
Pathogenic mechanisms of neurodegeneration in Parkinson disease.
    Neurologic clinics, 2015, Volume: 33, Issue:1

    The last 2 decades represent a period of unparalleled advancement in the understanding of the pathogenesis of Parkinson disease (PD). The discovery of several forms of familial parkinsonism with mendelian inheritance has elucidated insights into the mechanisms underlying the degeneration of dopaminergic neurons of the substantia nigra that histologically characterize PD. α-Synuclein, the principal component of Lewy bodies, remains the presumed pathogen at the heart of the current model; however, concurrently, a diverse range of other mechanisms have been implicated. The creation of a coherent disease model will be crucial to the development of effective disease modifying therapies for sporadic PD.

    Topics: alpha-Synuclein; Glucosylceramidase; Humans; Leucine-Rich Repeat Serine-Threonine Protein Kinase-2; Mutation; Nerve Degeneration; Parkinson Disease; Protein Serine-Threonine Kinases; Substantia Nigra

2015
Fluid biomarkers in multiple system atrophy: A review of the MSA Biomarker Initiative.
    Neurobiology of disease, 2015, Volume: 80

    Despite growing research efforts, no reliable biomarker currently exists for the diagnosis and prognosis of multiple system atrophy (MSA). Such biomarkers are urgently needed to improve diagnostic accuracy, prognostic guidance and also to serve as efficacy measures or surrogates of target engagement for future clinical trials. We here review candidate fluid biomarkers for MSA and provide considerations for further developments and harmonization of standard operating procedures. A PubMed search was performed until April 24, 2015 to review the literature with regard to candidate blood and cerebrospinal fluid (CSF) biomarkers for MSA. Abstracts of 1760 studies were retrieved and screened for eligibility. The final list included 60 studies assessing fluid biomarkers in patients with MSA. Most studies have focused on alpha-synuclein, markers of axonal degeneration or catecholamines. Their results suggest that combining several CSF fluid biomarkers may be more successful than using single markers, at least for the diagnosis. Currently, the clinically most useful markers may comprise a combination of the light chain of neurofilament (which is consistently elevated in MSA compared to controls and Parkinson's disease), metabolites of the catecholamine pathway and proteins such as α-synuclein, DJ-1 and total-tau. Beyond future efforts in biomarker discovery, the harmonization of standard operating procedures will be crucial for future success.

    Topics: alpha-Synuclein; Animals; Biomarkers; Brain; Catecholamines; Humans; Intermediate Filaments; Multiple System Atrophy; Nerve Degeneration

2015
[The role of alpha-synuclein in Parkinson's disease].
    Neuropsychopharmacologia Hungarica : a Magyar Pszichofarmakologiai Egyesulet lapja = official journal of the Hungarian Association of Psychopharmacology, 2014, Volume: 16, Issue:2

    α-synuclein, a small protein (140 amino acids) encoded by the SNCA gene is the best known isoform of the synuclein protein family. Though its physiological role is still not fully clarified, there is growing experimental evidence for a causal role of α-synuclein in the so-called conformational-neurodegenerative diseases. Conformational changes in the structure of the native soluble protein form insoluble neurotoxic aggregates and finally contribute to the formation of inclusion Lewy-bodies and Lewy-neurites. Neurodegeneration first hits the olfactory system, the peripheral autonomic nervous system, the enteric nervous system and the dorsal vagal motoneurons. The middle stage of the disease hits the dopaminergic neurons of the substantia nigra; and the neocortex is affected only in the late stage of the disease. This precise order of neurodegeneration is not always valid, but increases the likelihood that Lewy-bodies and neurodegenaration spread to intact areas in a prion-like way. Prions are infectious proteins which do not contain nucleic acids and cause diseases because they form toxic aggregates and filaments by misfolding in a β-sheet-rich conformation. The misfolded protein behaves like a template inducing conformational change in the wild type proteins causing cross-reaction and leading to neurodegeneration. Later, the defective proteins may infect healthy nerve cells, thus neurodegeneration is extended. Growing experimental evidence shows that monomers and aggregates of α-synuclein are secreted via exocytosis from damaged nerve cells and taken up via endocytosis by healthy nerve cells furnishing evidence for the prion-like role of α-synuclein.

    Topics: alpha-Synuclein; Animals; Antiparkinson Agents; Endocytosis; Exocytosis; Humans; Lewy Bodies; Nerve Degeneration; Neuroprotective Agents; Parkinson Disease; Prions; Protein Structure, Secondary; Selegiline

2014
The paths to neurodegeneration in genetic Parkinson's disease.
    CNS & neurological disorders drug targets, 2014, Volume: 13, Issue:9

    Parkinson's disease (PD) is a neurodegenerative disorder, which results from the loss of specific population of neurons, namely the pigmented dopamine secreting neurons of the substnatia nigra pars compatica (SNPc) of midbrain. The exact cause leading to nigrostriatal cell death is not yet known. In recent years, accumulating evidence from the identified molecular events in familial forms of PD contributed much to unraveling the mechanisms by which dopaminergic neurons die in PD and which hopefully would lead to the development of therapeutic interventions. Several major disease causing pathways were identified so far. These are possibly interconnected and some genes share a common pathway e.g., (i) defects in ubiquitin-proteasome pathway and protein misfolding and aggregation caused by α-synuclein and Parkin gene defects; (ii) defects in mitochondrial morphology and function in PINK1/Parkin and DJ-1 mutations; (iii) increased susceptibility to cellular oxidative stress which appear to underlie defects in α-synuclein, Parkin and DJ-1 genes. The aim of this review is to shed light on the molecular mechanisms by which mutations in familial-linked genes cause PD.

    Topics: alpha-Synuclein; Humans; Intracellular Signaling Peptides and Proteins; Mutation; Nerve Degeneration; Oncogene Proteins; Parkinson Disease; Protein Deglycase DJ-1; Signal Transduction; Ubiquitin-Protein Ligases

2014
Alpha-synuclein and tau: teammates in neurodegeneration?
    Molecular neurodegeneration, 2014, Oct-29, Volume: 9

    The accumulation of α-synuclein aggregates is the hallmark of Parkinson's disease, and more generally of synucleinopathies. The accumulation of tau aggregates however is classically found in the brains of patients with dementia, and this type of neuropathological feature specifically defines the tauopathies. Nevertheless, in numerous cases α-synuclein positive inclusions are also described in tauopathies and vice versa, suggesting a co-existence or crosstalk of these proteinopathies. Interestingly, α-synuclein and tau share striking common characteristics suggesting that they may work in concord. Tau and α-synuclein are both partially unfolded proteins that can form toxic oligomers and abnormal intracellular aggregates under pathological conditions. Furthermore, mutations in either are responsible for severe dominant familial neurodegeneration. Moreover, tau and α-synuclein appear to promote the fibrillization and solubility of each other in vitro and in vivo. This suggests that interactions between tau and α-synuclein form a deleterious feed-forward loop essential for the development and spreading of neurodegeneration. Here, we review the recent literature with respect to elucidating the possible links between α-synuclein and tau.

    Topics: alpha-Synuclein; Animals; Humans; Nerve Degeneration; Neurodegenerative Diseases; tau Proteins

2014
Oxidative and nitrative alpha-synuclein modifications and proteostatic stress: implications for disease mechanisms and interventions in synucleinopathies.
    Journal of neurochemistry, 2013, Volume: 125, Issue:4

    Alpha-synuclein (ASYN) is a major constituent of the typical protein aggregates observed in several neurodegenerative diseases that are collectively referred to as synucleinopathies. A causal involvement of ASYN in the initiation and progression of neurological diseases is suggested by observations indicating that single-point (e.g., A30P, A53T) or multiplication mutations of the gene encoding for ASYN cause early onset forms of Parkinson's disease (PD). The relative regional specificity of ASYN pathology is still a riddle that cannot be simply explained by its expression pattern. Also, transgenic over-expression of ASYN in mice does not recapitulate the typical dopaminergic neuronal death observed in PD. Thus, additional factors must contribute to ASYN-related toxicity. For instance, synucleinopathies are usually associated with inflammation and elevated levels of oxidative stress in affected brain areas. In turn, these conditions favor oxidative modifications of ASYN. Among these modifications, nitration of tyrosine residues, formation of covalent ASYN dimers, as well as methionine sulfoxidations are prominent examples that are observed in post-mortem PD brain sections. Oxidative modifications can affect ASYN aggregation, as well as its binding to biological membranes. This would affect neurotransmitter recycling, mitochondrial function and dynamics (fission/fusion), ASYN's degradation within a cell and, possibly, the transfer of modified ASYN to adjacent cells. Here, we propose a model on how covalent modifications of ASYN link energy stress, altered proteostasis, and oxidative stress, three major pathogenic processes involved in PD progression. Moreover, we hypothesize that ASYN may act physiologically as a catalytically regenerated scavenger of oxidants in healthy cells, thus performing an important protective role prior to the onset of disease or during aging.

    Topics: alpha-Synuclein; Humans; Nerve Degeneration; Nitric Oxide; Oxidative Stress; Parkinson Disease; Peroxynitrous Acid

2013
Dysfunction of the autophagy/lysosomal degradation pathway is a shared feature of the genetic synucleinopathies.
    FASEB journal : official publication of the Federation of American Societies for Experimental Biology, 2013, Volume: 27, Issue:9

    The past decade has witnessed huge advances in our understanding of the genetics underlying Parkinson's disease. Identifying commonalities in the biological function of genes linked to Parkinson's provides an opportunity to elucidate pathways that lead to neuronal degeneration and eventually to disease. We propose that the genetic forms of Parkinson's disease largely associated with α-synuclein-positive neuropathology (SNCA, LRRK2, and GBA) are brought together by involvement in the autophagy/lysosomal pathway and that this represents a unifying pathway to disease in these cases.

    Topics: alpha-Synuclein; Animals; Autophagy; Humans; Lysosomes; Models, Biological; Nerve Degeneration; Parkinson Disease

2013
Neuronal vulnerability, pathogenesis, and Parkinson's disease.
    Movement disorders : official journal of the Movement Disorder Society, 2013, Volume: 28, Issue:1

    Although there have been significant advances, pathogenesis in Parkinson's disease (PD) is still poorly understood. Potential clues about pathogenesis that have not been systematically pursued are suggested by the restricted pattern of neuronal pathology in the disease. In addition to dopaminergic neurons in the substantia nigra pars compacta (SNc), a significant number of other central and peripheral neuronal populations exhibit Lewy pathology (LP), phenotypic dysregulation, or frank degeneration in PD patients. Drawing on this literature, there appear to be a small number of risk factors contributing to vulnerability. These include autonomous activity, broad action potentials, low intrinsic calcium-buffering capacity, poorly myelinated long highly branched axons and terminal fields, and use of a monoamine neurotransmitter, often with the catecholamine-derived neuromelanin pigment. Of these phenotypic traits, only the physiological ones appear to provide a reachable therapeutic target at present.

    Topics: alpha-Synuclein; Humans; Nerve Degeneration; Nervous System; Neurons; Parkinson Disease

2013
α-Synuclein oligomers and clinical implications for Parkinson disease.
    Annals of neurology, 2013, Volume: 73, Issue:2

    Protein aggregation within the central nervous system has been recognized as a defining feature of neurodegenerative diseases since the early 20th century. Since that time, there has been a growing list of neurodegenerative disorders, including Parkinson disease, which are characterized by inclusions of specific pathogenic proteins. This has led to the long-held dogma that these characteristic protein inclusions, which are composed of large insoluble fibrillar protein aggregates and visible by light microscopy, are responsible for cell death in these diseases. However, the correlation between protein inclusion formation and cytotoxicity is inconsistent, suggesting that another form of the pathogenic proteins may be contributing to neurodegeneration. There is emerging evidence implicating soluble oligomers, smaller protein aggregates not detectable by conventional microscopy, as potential culprits in the pathogenesis of neurodegenerative diseases. The protein α-synuclein is well recognized to contribute to the pathogenesis of Parkinson disease and is the major component of Lewy bodies and Lewy neurites. However, α-synuclein also forms oligomeric species, with certain conformations being toxic to cells. The mechanisms by which these α-synuclein oligomers cause cell death are being actively investigated, as they may provide new strategies for diagnosis and treatment of Parkinson disease and related disorders. Here we review the possible role of α-synuclein oligomers in cell death in Parkinson disease and discuss the potential clinical implications.

    Topics: alpha-Synuclein; Animals; Humans; Inclusion Bodies; Nerve Degeneration; Oligonucleotides; Parkinson Disease

2013
The role of Ser129 phosphorylation of α-synuclein in neurodegeneration of Parkinson's disease: a review of in vivo models.
    Reviews in the neurosciences, 2013, Volume: 24, Issue:2

    Parkinson's disease is the most common neurodegenerative movement disorder. The motor impairments of Parkinson's disease are caused by the loss of dopaminergic neurons in the substantia nigra and associated with the appearance of fibrillar aggregates of α-synuclein (α-syn) called Lewy bodies. Approximately 90% of α-syn deposited in Lewy bodies is phosphorylated at serine 129 (Ser129). In contrast, only 4% or less of total α-syn is phosphorylated at this residue in the normal brain. This suggests that the accumulation of Ser129-phosphorylated α-syn leads to the formation of Lewy bodies and dopaminergic neurodegeneration in Parkinson's disease. Our laboratory and others have performed experiments using in vivo models of Parkinson's disease to elucidate the role of increased Ser129 phosphorylation in α-syn neurotoxicity. However, there has been a lack of consistency among these models. In this review, we summarize the main findings regarding the relationship between Ser129 phosphorylation and α-syn neurotoxicity, and examine the differences among models. We further discuss the role of Ser129 phosphorylation in α-syn aggregation and the future directions to test the potential of Ser129 phosphorylation as a therapeutic target for slowing the progression of Parkinson's disease.

    Topics: alpha-Synuclein; Animals; Disease Models, Animal; Humans; Nerve Degeneration; Parkinson Disease; Phosphorylation; Serine

2013
Animal models of multiple system atrophy.
    Neuroscience, 2012, Jun-01, Volume: 211

    Multiple system atrophy (MSA) is a sporadic adult-onset neurodegenerative disorder clinically characterized by a variable combination of dysautonomia, levodopa-unresponsive parkinsonian and cerebellar symptoms. Neurodegeneration in MSA occurs in the substantia nigra, putamen, inferior olive, pontine and brainstem nuclei, as well as intermediolateral cell column of the spinal cord. MSA is recognized as a synucleinopathy due to the accumulation of insoluble alpha-synuclein in oligodendroglial cytoplasmic inclusions. Several animal models have been developed in order to reproduce various clinical and pathological features of MSA. Using "double toxin-double lesion" or "single toxin-double lesion", neurotoxin-based models were designed in rats, mice and non-human primates to reproduce the neuropathology of MSA in the nigrostriatal system while gene-based models were developed in mice to reproduce the accumulation of insoluble alpha-synuclein in oligodendrocytes. Both approaches have then been merged to create optimized, dual-hit models. This review describes the different animal models of MSA, their respective advantages and limitations and their usefulness to decipher the pathophysiology of MSA then to define efficient symptomatic and disease-modifying therapies. This article is part of a Special Issue entitled: Neuroscience Disease Models.

    Topics: alpha-Synuclein; Animals; Corpus Striatum; Disease Models, Animal; Multiple System Atrophy; Nerve Degeneration; Neurotoxins; Substantia Nigra

2012
α-Synuclein synaptic pathology and its implications in the development of novel therapeutic approaches to cure Parkinson's disease.
    Brain research, 2012, Jan-13, Volume: 1432

    Parkinson's disease (PD) is characterized by a progressive loss of dopamine (DA) neurons of the nigrostriatal system and by the presence of Lewy bodies (LB), proteinaceous inclusions mainly composed of filamentous α-synuclein aggregates. Alpha-synuclein is a natively unfolded protein which plays a central role in the control of dopaminergic neuronal functions and which is thought to be critically implicated in PD pathophysiology. Indeed, besides the fact that α-synuclein is the main protein component of LB, genetic studies showed that mutations and multiplications of the α-synuclein gene are responsible for the onset of familial forms of PD. A large body of evidence indicates that α-synuclein pathology at dopaminergic synapses may underlie the onset of neuronal cell dysfunction and degeneration in the PD brain. Thus, since the available therapeutic approaches to cure this disease are still limited, we hypothesized that the analysis of the α-synuclein synaptic proteome/lipidome may represent a tool to identify novel potential therapeutic targets to cure this disorder. We thus performed a critical review of studies describing α-synuclein pathophysiology at synaptic sites in experimental models of PD and in this paper we outline the most relevant findings regarding the specific modulatory effects exerted by α-synuclein in the control of synaptic functions in physiological and pathological conditions. The conclusions of these studies allow to single out novel potential therapeutic targets among the α-synuclein synaptic partners. These targets may be considered for the development of new pharmacological and gene-based strategies to cure PD.

    Topics: alpha-Synuclein; Animals; Antiparkinson Agents; Humans; Nerve Degeneration; Parkinson Disease; Synapses

2012
Synaptic protein alterations in Parkinson's disease.
    Molecular neurobiology, 2012, Volume: 45, Issue:1

    Alterations occur within distal neuronal compartments, including axons and synapses, during the course of neurodegenerative diseases such as Parkinson's disease (PD). These changes could hold important implications for the functioning of neural networks, especially since research studies have shown a loss of dendritic spines locating to medium spiny projection neurons and impaired axonal transport in PD-affected brains. However, despite ever-increasing awareness of the vulnerability of synapses and axons, inadequate understanding of the independent mechanisms regulating non-somatic neurodegeneration prevails. This has resulted in limited therapeutic strategies capable of targeting these distinct cellular compartments. Deregulated protein synthesis, folding and degrading proteins, and protein quality-control systems have repeatedly been linked with morphological and functional alterations of synapses in the PD-affected brains. Here, we review current understanding concerning the proteins involved in structural and functional changes that affect synaptic contact-points in PD. The collection of studies discussed emphasizes the need for developing therapeutics aimed at deregulated protein synthesis and degradation pathways operating at axonal and dendritic synapses for preserving "normal" circuitry and function, for as long as possible.

    Topics: alpha-Synuclein; Animals; Humans; Nerve Degeneration; Nerve Tissue Proteins; Parkinson Disease; Synapses; Synaptic Membranes; Synaptic Transmission

2012
Synaptic dysfunction in Parkinson's disease.
    Advances in experimental medicine and biology, 2012, Volume: 970

    Activity-dependent modifications in synaptic efficacy, such as long-term depression (LTD) and long-term potentiation (LTP), represent key cellular substrates for adaptive motor control and procedural memory. The impairment of these two forms of synaptic plasticity in the nucleus striatum could account for the onset and the progression of motor and cognitive symptoms of Parkinson's disease (PD), characterized by the massive degeneration of dopaminergic neurons. In fact, both LTD and LTP are peculiarly controlled and modulated by dopaminergic transmission coming from nigrostriatal terminals. Changes in corticostriatal and nigrostriatal neuronal excitability may influence profoundly the threshold for the induction of synaptic plasticity, and changes in striatal synaptic transmission efficacy are supposed to play a role in the occurrence of PD symptoms. Understanding of these maladaptive forms of synaptic plasticity has mostly come from the analysis of experimental animal models of PD. A series of cellular and synaptic alterations occur in the striatum of experimental parkinsonism in response to the massive dopaminergic loss. In particular, dysfunctions in trafficking and subunit composition of glutamatergic NMDA receptors on striatal efferent neurons contribute to the clinical features of the experimental parkinsonism. Interestingly, it has become increasingly evident that in striatal spiny neurons, the correct assembly of NMDA receptor complex at the postsynaptic site is a major player in early phases of PD, and it is sensitive to distinct degrees of DA denervation. The molecular defects at the basis of PD progression may be not confined just at the postsynaptic neuron: accumulating evidences have recently shown that the genes linked to PD play a critical role at the presynaptic site. DA release into the synaptic cleft relies on a proper presynaptic vesicular transport; impairment of SV trafficking, modification of DA flow, and altered presynaptic plasticity have been described in several PD animal models. Furthermore, an impaired DA turnover has been described in presymptomatic PD patients. Thus, given the pathological events occurring precociously at the synapses of PD patients, post- and presynaptic sites may represent an adequate target for early therapeutic intervention.

    Topics: alpha-Synuclein; Animals; Corpus Striatum; Disease Models, Animal; Dopamine; Dopaminergic Neurons; Humans; Leucine-Rich Repeat Serine-Threonine Protein Kinase-2; Long-Term Potentiation; Mice; Mutation; Nerve Degeneration; Neuronal Plasticity; Parkinson Disease; Protein Serine-Threonine Kinases; Receptors, N-Methyl-D-Aspartate; Synapses; Synaptic Transmission; Transport Vesicles

2012
The molecular basis of dopaminergic brain imaging in Parkinson's disease.
    The quarterly journal of nuclear medicine and molecular imaging : official publication of the Italian Association of Nuclear Medicine (AIMN) [and] the International Association of Radiopharmacology (IAR), [and] Section of the Society of..., 2012, Volume: 56, Issue:1

    The central role of dopamine neuronal loss in Parkinson's disease provides a clear pathologic framework and rationale for imaging the system both to interrogate dynamic pathophysiologic changes as well as to aid in diagnosis and clinical management. Recent post mortem studies of Parkinson's brain provide a much fuller depiction of the inexorable and progressive topology of pathophysiologic changes, including brain alpha-synuclein deposition. This informs PET and SPECT evaluations for testing hypotheses regarding the course of degeneration in longitudinal studies of Parkinson's disease patients. Recent work has underscored the subtlety of change in the dopaminergic neuronal system and its neural connections as a function of disease status and treatment. The interplay between other neurochemical brain systems and dopamine elucidates potential new targets for therapeutic intervention across the stages of the disease.

    Topics: alpha-Synuclein; Brain; Dopaminergic Neurons; Humans; Nerve Degeneration; Parkinson Disease; Positron-Emission Tomography; Tomography, Emission-Computed, Single-Photon

2012
Spinal cord and parkinsonism: neuromorphological evidences in humans and experimental studies.
    Journal of chemical neuroanatomy, 2011, Volume: 42, Issue:4

    The involvement of the spinal cord in parkinsonism is becoming more and more evident based on human autopsies and on experimental models, obtained using specific neurotoxins or genetic manipulations. Besides Parkinson disease, other degenerative disorders characterized by parkinsonism, involve the spinal cord, and multiple neurotransmitters, apart dopamine, are altered in parkinsonism, also in their spinal projections. In the present review we discuss spinal cord pathology of different genetic or toxic experimental models of parkinsonism, as well as the neuropathological reports from autoptic cases of sporadic Parkinson disease and of other neurodegenerative conditions, overlapping with parkinsonism. Furthermore, anatomical distribution of alpha-synuclein in the spinal cord and coeruleo-spinal projections are reviewed, at the light of their possible involvement in spinal neurons degeneration. All these evidences call for an anatomical stemmed novel approach to understand specific features of parkinsonism, which might be due to such an involvement of the spinal cord. Moreover they suggest a common neurodegenerative process, underlying distinct neurodegenerative disorders, to which spinal neurons could be the more sensible.

    Topics: alpha-Synuclein; Animals; Disease Models, Animal; Humans; Nerve Degeneration; Neurons; Parkinsonian Disorders; Spinal Cord

2011
Genetic LRRK2 models of Parkinson's disease: Dissecting the pathogenic pathway and exploring clinical applications.
    Movement disorders : official journal of the Movement Disorder Society, 2011, Volume: 26, Issue:8

    Dominantly inherited mutations in leucine-rich repeat kinase 2 are the most common cause of familial Parkinson's disease. Understanding leucine-rich repeat kinase 2 biology and pathophysiology is central to the elucidation of Parkinson's disease etiology and development of disease intervention. Recently, a number of genetic mouse models of leucine-rich repeat kinase 2 have been reported utilizing different genetic approaches. Some similarities in Parkinson's disease-related pathology emerge in these genetic models despite lack of substantial neuropathology and clinical syndromes of Parkinson's disease. The systematic characterization of these models has begun to shed light on leucine-rich repeat kinase 2 biology and pathophysiology and is expected to offer the identification and validation of drug targets. In this review, we summarize the progress of genetic leucine-rich repeat kinase 2 mouse models and discuss their utility in understanding much needed knowledge regarding early-stage (presymptomatic) disease progression, identifying drug targets, and exploring the potential to aid compound screening focused on inhibitors of kinase activity of leucine-rich repeat kinase 2.

    Topics: alpha-Synuclein; Animals; Antiparasitic Agents; Disease Models, Animal; Gene Expression Regulation; Humans; Leucine-Rich Repeat Serine-Threonine Protein Kinase-2; Mice; Mice, Transgenic; Mutation; Nerve Degeneration; Neuroglia; Parkinson Disease; Protein Serine-Threonine Kinases; tau Proteins

2011
Drug targets from genetics: α-synuclein.
    CNS & neurological disorders drug targets, 2011, Sep-01, Volume: 10, Issue:6

    One of the critical issues in Parkinson disease (PD) research is the identity of the specific toxic, pathogenic moiety. In PD, mutations in α-synuclein (αsyn) or multiplication of the SNCA gene encoding αsyn, result in a phenotype of cellular inclusions, cell death, and brain dysfunction. While the historical point of view has been that the macroscopic aggregates containing αsyn are the toxic species, in the last several years evidence has emerged that suggests instead that smaller soluble species--likely oligomers containing misfolded αsyn--are actually the toxic moiety and that the fibrillar inclusions may even be a cellular detoxification pathway and less harmful. If soluble misfolded species of αsyn are the toxic moieties, then cellular mechanisms that degrade misfolded αsyn would be neuroprotective and a rational target for drug development. In this review we will discuss the fundamental mechanisms underlying αsyn toxicity including oligomer formation, oxidative stress, and degradation pathways and consider rational therapeutic strategies that may have the potential to prevent or halt αsyn induced pathogenesis in PD.

    Topics: alpha-Synuclein; Animals; Antioxidants; Humans; Molecular Targeted Therapy; Nerve Degeneration; Oxidative Stress; Parkinson Disease

2011
Caenorhabditis elegans as an experimental tool for the study of complex neurological diseases: Parkinson's disease, Alzheimer's disease and autism spectrum disorder.
    Invertebrate neuroscience : IN, 2011, Volume: 11, Issue:2

    The nematode Caenorhabditis elegans has a very well-defined and genetically tractable nervous system which offers an effective model to explore basic mechanistic pathways that might be underpin complex human neurological diseases. Here, the role C. elegans is playing in understanding two neurodegenerative conditions, Parkinson's and Alzheimer's disease (AD), and a complex neurological condition, autism, is used as an exemplar of the utility of this model system. C. elegans is an imperfect model of Parkinson's disease because it lacks orthologues of the human disease-related genes PARK1 and LRRK2 which are linked to the autosomal dominant form of this disease. Despite this fact, the nematode is a good model because it allows transgenic expression of these human genes and the study of the impact on dopaminergic neurons in several genetic backgrounds and environmental conditions. For AD, C. elegans has orthologues of the amyloid precursor protein and both human presenilins, PS1 and PS2. In addition, many of the neurotoxic properties linked with Aβ amyloid and tau peptides can be studied in the nematode. Autism spectrum disorder is a complex neurodevelopmental disorder characterised by impairments in human social interaction, difficulties in communication, and restrictive and repetitive behaviours. Establishing C. elegans as a model for this complex behavioural disorder is difficult; however, abnormalities in neuronal synaptic communication are implicated in the aetiology of the disorder. Numerous studies have associated autism with mutations in several genes involved in excitatory and inhibitory synapses in the mammalian brain, including neuroligin, neurexin and shank, for which there are C. elegans orthologues. Thus, several molecular pathways and behavioural phenotypes in C. elegans have been related to autism. In general, the nematode offers a series of advantages that combined with knowledge from other animal models and human research, provides a powerful complementary experimental approach for understanding the molecular mechanisms and underlying aetiology of complex neurological diseases.

    Topics: alpha-Synuclein; Alzheimer Disease; Amyloid beta-Protein Precursor; Animals; Animals, Genetically Modified; Caenorhabditis elegans; Caenorhabditis elegans Proteins; Child; Child Development Disorders, Pervasive; Disease Models, Animal; Dopaminergic Neurons; Humans; Leucine-Rich Repeat Serine-Threonine Protein Kinase-2; Nerve Degeneration; Parkinson Disease; Presenilins; Protein Serine-Threonine Kinases; Synapses

2011
Hsp70 molecular chaperones and Parkinson's disease.
    Biopolymers, 2010, Volume: 93, Issue:3

    Because over expression of Hsp70 molecular chaperones suppresses the toxicity of aberrantly folded proteins that occur in Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis, and various polyQ-diseases (Huntington's disease and ataxias), Hsp70 is garnering attention as a possible therapeutic agent for these various diseases. Here, I review progress in this fascinating field of molecular chaperones and neurodegeneration and describe our current understanding of the mechanisms by which Hsp70 protects cells from the PD-related protein called alpha-synuclein (alpha-syn).

    Topics: alpha-Synuclein; Amyloid; Animals; Autophagy; Biomarkers; HSP70 Heat-Shock Proteins; Humans; Models, Molecular; Molecular Chaperones; Nerve Degeneration; Parkinson Disease; Prokaryotic Cells; Protein Conformation

2010
Ageing, neurodegeneration and Parkinson's disease.
    Age and ageing, 2010, Volume: 39, Issue:2

    Age is the largest risk factor for the development and progression of Parkinson's disease (PD). Ageing affects many cellular processes that predispose to neurodegeneration, and age-related changes in cellular function predispose to the pathogenesis of PD. The accumulation of age-related somatic damage combined with a failure of compensatory mechanisms may lead to an acceleration of PD with age. The formation of Lewy bodies may represent a marker for protective mechanisms against age-related dysfunction and degeneration of the nervous system. Mild parkinsonian signs may be present in older people, which are associated with reduced function. These may be due to age-related decline in dopaminergic activity, incidental Lewy body disease, degenerative pathologies (early PD and Alzheimer's disease) or vascular pathology. Ageing may affect the clinical presentation of PD with altered drug side effects, increased risk of developing dementia and an increased likelihood of admission to a nursing home. Progression of PD, including the development of dementia, and hallucinations is related to the age of the patient rather than the age of disease onset. PD may reflect a failure of the normal cellular compensatory mechanisms in vulnerable brain regions, and this vulnerability is increased by ageing. PD is one of the best examples of an age-related disease.

    Topics: Age Factors; Aging; alpha-Synuclein; Disease Progression; Female; Humans; Lewy Bodies; Male; Nerve Degeneration; Parkinson Disease; Risk Factors

2010
[Mechanism of neuronal degeneration of multiple system atrophy].
    Brain and nerve = Shinkei kenkyu no shinpo, 2009, Volume: 61, Issue:9

    Multiple system atrophy (MSA) is a sporadic neurodegenerative disorder that encompasses olivopontocerebellar atrophy (OPCA), striatonigral degeneration (SND) and Shy-Drager syndrome (SDS). The histopathological hallmarks are alpha-synuclein (AS) positive glial cytoplasmic inclusions (GCIs) in oligodendroglias. AS aggregation is also found in glial nuclear inclusions (GNIs), neuronal cytoplasmic inclusions (NCIs), neuronal nuclear inclusions (NNIs) and dystrophic neurites. Reviewing the pathological features in 102 MSA cases revealed that the, OPCA-type was relatively more frequent and SND-type was less frequent in Japanese MSA cases. The frequency of the SND-type is relatively high in Western countries. This different in the dominant type suggests that the phenotypic patterns of MSA may vary with the race. In early stages of MSA, in addition to GCIs, NNIs, NCIs, and diffuse homogenous stain of AS in neuronal nuclei and cytoplasm were observed in various vulnerable lesions including the pontine nuclei, putamen, substantia nigra, locus ceruleus, inferior olivary nucleus, intermediolateral column of the thoracic cord, lower motor neurons, and cortical pyramidal neurons. These findings indicated that the primary nonfibrillar and fibrillar AS aggregation also occurred in neurons. Therefore, both the direct involvement of neurons themselves and the oligodendroglia-myelin-axon mechanism may synergistically accelerate the degenerative process of MSA.

    Topics: alpha-Synuclein; Humans; Inclusion Bodies; Multiple System Atrophy; Nerve Degeneration; Oligodendroglia

2009
Alpha-synuclein and autophagy as common steps in neurodegeneration.
    Parkinsonism & related disorders, 2008, Volume: 14 Suppl 2

    The major component of Lewy bodies in Parkinson's disease (PD) is alpha-synuclein, which is considered as a substrate of the ubiquitin-proteasome (UP) system, although autophagy seems to be equally involved. Here we discuss the co-existence of alpha-synuclein and proteins belonging to the UP system within autophagic granules, further developing as neuronal inclusions. We hypothesize that, following slight insults, both UP and autophagy are induced; if toxic stimuli are prolonged, these pathways are overwhelmed and cell death occurs. We then indicate a protective role of autophagy in PD and suggest it as a therapeutic target to slow down the progression of the disease.

    Topics: alpha-Synuclein; Animals; Autophagy; Humans; Nerve Degeneration

2008
Research in motion: the enigma of Parkinson's disease pathology spread.
    Nature reviews. Neuroscience, 2008, Volume: 9, Issue:10

    Neuropathological changes in Parkinson's disease progress slowly and spread according to a characteristic pattern. Recent papers have shed light on this progression of pathology by examining the fate of neurons grafted into the brains of patients with Parkinson's disease. Two of these studies demonstrate that grafted healthy neurons can gradually develop the same pathology as host neurons in the diseased brains. According to these studies, implanted neurons developed alpha-synuclein- and ubiquitin-positive Lewy bodies more than a decade after transplantation. We discuss the possible underlying mechanisms and their implications for how pathology spreads in Parkinson's disease.

    Topics: alpha-Synuclein; Brain; Brain Tissue Transplantation; Disease Progression; Graft Survival; Humans; Lewy Bodies; Nerve Degeneration; Oxidative Stress; Parkinson Disease

2008
[Clinical and pathological study on early diagnosis of Parkinson's disease and dementia with Lewy bodies].
    Rinsho shinkeigaku = Clinical neurology, 2008, Volume: 48, Issue:11

    Cardiac uptake of meta-iodobenzylguanidine (MIBG) is specifically reduced in Lewy body disease (LBD). To see pathological basis of the reduced cardiac uptake of MIBG in LBD, we immunohistichemically examined cardiac tissues from patients with LBD, related movement disorders and Alzheimer's disease (AD). In LBD, cardiac sympathetic denervation occurs, which accounts for the reduced cardiac uptake of MIBG. Patients with LBD have Lewy bodies (LBs) in the nervous system, whereas patients with the other neurodegenerative parkinsonism, parkin-associated Parkinson's disease (PD) and AD and have no LBs. Therefore, cardiac sympathetic denervation is closely related to the presence of LBs in a wide range of neurodegenerative processes. We further investigate how a-synuclein aggregates are involved in degeneration of the cardiac sympathetic nerve in PD. Accumulation of alpha-synuclein aggregates in the distal axons of the cardiac sympathetic nervous system precedes that of neuronal somata or neurites in the paravertebral sympathetic ganglia and that it heralds centripetal degeneration of the cardiac sympathetic nerve in PD. This chronological and dynamic relationship between alpha-synuclein aggregates and degeneration of the cardiac sympathetic nervous system may represent the pathological mechanism underlying a common degenerative process in PD.

    Topics: 3-Iodobenzylguanidine; alpha-Synuclein; Biomarkers; Diagnosis, Differential; Early Diagnosis; Heart; Humans; Iodine Radioisotopes; Lewy Body Disease; Nerve Degeneration; Parkinson Disease; Radionuclide Imaging; Radiopharmaceuticals; Sympathetic Nervous System

2008
[Lewy body formation in Parkinson's disease: neurodegeneration or neuroprotection?].
    Rinsho shinkeigaku = Clinical neurology, 2008, Volume: 48, Issue:11

    The histological hallmark of Parkinson's disease (PD) is the presence of fibrillar aggregates called Lewy bodies (LBs). LB formation has been considered to be a marker for neuronal degeneration, because neuronal loss is found in the predilection sites for LBs. To date, more than 70 molecules have been identified in LBs, in which alpha-synuclein is a major constituent of LB fibrils. Alpha-synuclein immunohistochemistry reveals that diffuse cytoplasmic staining develops into pale bodies via compaction, and that LBs arise from the peripheral portion of pale bodies. Abnormal accumulation of alpha-synuclein (diffuse cytoplasmic staining, pale bodies and LBs) is found in 10% of pigmented neurons in the substantia nigra and more than 50% of those in the locus ceruleus in PD. Recent studies have suggested that oligomers and protofibrils of alpha-synuclein are cytotoxic, and that pale bodies and LBs may represent a cytoprotective mechanism in PD.

    Topics: alpha-Synuclein; Cytoplasm; Humans; Lewy Bodies; Locus Coeruleus; Nerve Degeneration; Neurons; Parkinson Disease; Substantia Nigra

2008
The role of autophagy-lysosome pathway in neurodegeneration associated with Parkinson's disease.
    Brain : a journal of neurology, 2008, Volume: 131, Issue:Pt 8

    The ubiquitin-proteasome system (UPS) and autophagy-lysosome pathway (ALP) are the two most important mechanisms that normally repair or remove abnormal proteins. Alterations in the function of these systems to degrade misfolded and aggregated proteins are being increasingly recognized as playing a pivotal role in the pathogenesis of many neurodegenerative disorders such as Parkinson's disease. Dysfunction of the UPS has been already strongly implicated in the pathogenesis of this disease and, more recently, growing interest has been shown in identifying the role of ALP in neurodegeneration. Mutations of alpha-synuclein and the increase of intracellular concentrations of non-mutant alpha-synuclein have been associated with Parkinson's disease phenotype. The demonstration that alpha-synuclein is degraded by both proteasome and autophagy indicates a possible linkage between the dysfunction of the UPS or ALP and the occurrence of this disorder. The fact that mutant alpha-synucleins inhibit ALP functioning by tightly binding to the receptor on the lysosomal membrane for autophagy pathway further supports the assumption that impairment of the ALP may be related to the development of Parkinson's disease. In this review, we summarize the recent findings related to this topic and discuss the unique role of the ALP in this neurogenerative disorder and the putative therapeutic potential through ALP enhancement.

    Topics: alpha-Synuclein; Autophagy; Humans; Lysosomes; Mutation; Nerve Degeneration; Nerve Tissue Proteins; Parkinson Disease; Protein Kinases; Signal Transduction; TOR Serine-Threonine Kinases; Ubiquitin

2008
Protein folding diseases and neurodegeneration: lessons learned from yeast.
    Biochimica et biophysica acta, 2008, Volume: 1783, Issue:7

    Budding yeast Saccharomyces cerevisiae has proven to be a valuable model organism for studying fundamental cellular processes across the eukaryotic kingdom including man. In this respect, complementation assays, in which the yeast protein is replaced by a homologous protein from another organism, have been very instructive. A newer trend is to use the yeast cell factory as a toolbox to understand cellular processes controlled by proteins for which the yeast lacks functional counterparts. An increasing number of studies have indicated that S. cerevisiae is a suitable model system to decipher molecular mechanisms involved in a variety of neurodegenerative disorders caused by aberrant protein folding. Here we review the current knowledge gained by the use of so-called humanized yeasts in the field of Huntington's, Parkinson's and Alzheimer's diseases.

    Topics: alpha-Synuclein; Alzheimer Disease; Amyloid beta-Peptides; Apoptosis; Apoptosis Regulatory Proteins; Heat-Shock Proteins; Humans; Huntingtin Protein; Huntington Disease; Models, Biological; Nerve Degeneration; Nerve Tissue Proteins; Nuclear Proteins; Parkinson Disease; Peptides; Protein Folding; Saccharomyces cerevisiae; tau Proteins; Yeasts

2008
Genetic models of Parkinson's disease: mechanisms and therapies.
    SEB experimental biology series, 2008, Volume: 60

    Topics: alpha-Synuclein; Animals; Animals, Genetically Modified; Disease Models, Animal; Drosophila melanogaster; Drosophila Proteins; Humans; Models, Animal; Nerve Degeneration; Parkinson Disease

2008
[Clinical and pathological study on early diagnosis of Parkinson's disease and dementia with Lewy bodies].
    Rinsho shinkeigaku = Clinical neurology, 2008, Volume: 48, Issue:1

    [123I] Meta-iodobenzylguanidine (MIBG) myocardial scintigraphy has been used to evaluate postganglionic cardiac sympathetic innervation in heart diseases and some neurological disorders. To see clinical usefulness of MIBG myocardial scintigraphy to differentiate Parkinson's disease (PD) and dementia with Lewy bodies (DLB) from related movement disorders and Alzheimer disease (AD), we performed MIBG myocardial scintigraphy in patients with these disorders. Cardiac uptake of MIBG is specifically reduced in PD and DLB, and this imaging approach is a sensitive diagnostic tool that possibly differentiates PD and DLB from related movement disorders and AD. To see pathological basis of the reduced cardiac uptake of MIBG in Lewy body disease, we immunohistochemically examined cardiac tissues from patients with PD, DLB, related movement disorders and AD using antibodies against tyrosine hydroxylase (TH) and phosphorylated neurofilament (NF). Not only TH- but also NF-immunoreactive (ir) axons in the epicardial nerve fascicles were markedly decreased in Lewy body disease, namely cardiac sympathetic denervation, which accounts for the reduced cardiac uptake of MIBG in Lewy body disease. Patients with PD and DLB have Lewy bodies (LBs) in the nervous system, whereas patients with multiple system atrophy (MSA), progressive supranuclear palsy, corticobasal degeneration, parkin-associated PD and AD have no LBs in the nervous system. Even in patients with MSA, cardiac sympathetic denervation was associated with the presence of LBs. Therefore, cardiac sympathetic denervation is closely related to the presence of LBs in a wide range of neurodegenerative processes. Taken together, we conclude that the reduced cardiac uptake of MIBG is a potential biomarker for the presence of LBs. Because alpha-synuclein is one of the key molecules in the pathogenesis of PD, we further investigate how alpha-synuclein aggregates are involved in degeneration of the cardiac sympathetic nerve in PD. We immunohistochemically examined cardiac tissues from patients with incidental Lewy body disease (ILBD) and PD using antibodies against TH and phosphorylated alpha-synuclein. We found that (1) alpha-synuclein aggregates in the epicardial nerve fascicles, namely the distal axons of the cardiac sympathetic nerve, were much more abundant in ILBD with preserved TH-ir axons than in ILBD with decreased TH-ir axons and PD; (2) alpha-synuclein aggregates in the epicardial nerve fascicles were closely related

    Topics: 3-Iodobenzylguanidine; alpha-Synuclein; Diagnosis, Differential; Early Diagnosis; Heart; Humans; Iodine Radioisotopes; Lewy Body Disease; Myocardium; Nerve Degeneration; Parkinson Disease; Radionuclide Imaging; Radiopharmaceuticals; Sympathetic Nervous System

2008
Transgenic mice with human mutant genes causing Parkinson's disease and amyotrophic lateral sclerosis provide common insight into mechanisms of motor neuron selective vulnerability to degeneration.
    Reviews in the neurosciences, 2007, Volume: 18, Issue:2

    A variety of gene mutations can cause familial forms of Parkinson's disease (PD) or amyotrophic lateral sclerosis (ALS). Mutations in the synaptic protein alpha-synuclein (alpha-Syn) cause PD. Mutations in the antioxidant enzyme superoxide dismutase-1 (SOD1) cause ALS. The mechanisms of human mutant a-Syn and SOD1 toxicity to neurons are not known. Transgenic (tg) mice expressing human mutant alpha-Syn or SOD1 develop profound fatal neurologic disease characterized by progressive motor deficits, paralysis, and neurodegeneration. Ala-53-->Thr (A53T)-mutant alpha-Syn and Gly-93-->Ala (G93A)-mutant SOD1 tg mice develop prominent mitochondrial abnormalities. Interestingly, although nigral neurons in A53T mice are relatively preserved, spinal motor neurons (MNs) undergo profound degeneration. In A53T mice, mitochondria degenerate in neurons, and complex IV activity is reduced. Furthermore, mitochondria in neurons develop DNA breaks and have p53 targeted to the outer membrane. Nitrated a-Syn accumulates in degenerating MNs in A53T mice. mSOD1 mouse MNs accumulate mitochondria from the axon terminals and generate higher levels of reactive oxygen/nitrogen species than MNs in control mice. mSOD1 mouse MNs accumulate DNA single-strand breaks prior to double-strand breaks occurring in nuclear and mitochondrial DNA. Nitrated and aggregated cytochrome c oxidase subunit-I and nitrated SOD2 accumulate in mSOD1 mouse spinal cord. Mitochondria in mSOD1 mouse MNs accumulate NADPH diaphorase and inducible NOS (iNOS)-like immunoreactivity, and iNOS gene deletion significantly extends the lifespan of G93A-mSOD1 mice. Mitochondrial changes develop long before symptoms emerge. These experiments reveal that mitochondrial nitrative stress and perturbations in mitochondrial trafficking may be antecedents of neuronal cell death in animal models of PD and ALS.

    Topics: alpha-Synuclein; Amyotrophic Lateral Sclerosis; Animals; Cell Death; Humans; Mice; Mice, Transgenic; Mitochondria; Motor Neurons; Mutation; Nerve Degeneration; Parkinson Disease; Superoxide Dismutase; Superoxide Dismutase-1

2007
Multiple system atrophy: alpha-synuclein and neuronal degeneration.
    Neuropathology : official journal of the Japanese Society of Neuropathology, 2007, Volume: 27, Issue:5

    Multiple system atrophy (MSA) is a sporadic neurodegenerative disorder that encompasses olivopontocerebellar atrophy (OPCA), striatonigral degeneration (SND) and Shy-Drager syndrome (SDS). The histopathological hallmark is the formation of alpha-synuclein-positive glial cytoplasmic inclusions (GCIs) in oligodendroglia. alpha-synuclein aggregation is also found in glial nuclear inclusions, neuronal cytoplasmic inclusions (NCIs), neuronal nuclear inclusions (NNIs) and dystrophic neurites. We evaluated the pathological features of 102 MSA cases, and presented the pathological spectrum of MSA and initial features of alpha-synuclein accumulation. We found that 39% of the 102 cases showed equivalent SND and OPCA pathologies, 33% showed OPCA- and 22% showed SND-predominant pathology, whereas 6% showed extremely mild changes. Our pathological analysis indicated that OPCA-type was relatively more frequent and SND-type was less frequent in Japanese MSA cases, compared to the relatively high frequency of SND-type in Western countries, suggesting that different phenotypic patterns of MSA may exist between races. In the early stage, in addition to GCIs, NNIs and diffuse homogenous alpha-synuclein staining in neuronal nuclei and cytoplasm were observed in lesions in the pontine nuclei, putamen, substantia nigra, locus ceruleus, inferior olivary nucleus, intermediolateral column of thoracic spinal cord, lower motor neurons and cortical pyramidal neurons. A subgroup of MSA cases with severe temporal atrophy showed numerous NCIs, particularly in the limbic system. These findings suggest that primary non-fibrillar and fibrillar alpha-synuclein aggregation also occur in neurons. The oligo-myelin-axon-neuron complex mechanism, along with the direct involvement of neurons themselves, may synergistically accelerate the degenerative process of MSA.

    Topics: alpha-Synuclein; Autonomic Nervous System; Biomarkers; Brain; Cerebral Cortex; Humans; Motor Neurons; Multiple System Atrophy; Nerve Degeneration; Neurons

2007
The pathogenesis of cell death in Parkinson's disease--2007.
    Movement disorders : official journal of the Movement Disorder Society, 2007, Volume: 22 Suppl 17

    A number of factors have been implicated in the pathogenesis of cell death in Parkinson's disease (PD). These include oxidative stress, mitochondrial dysfunction, inflammation, excitotoxicity, and apoptosis. While the precise pathogenic mechanism leading to neurodegeneration in PD is not known, there is considerable evidence suggesting that cell death occurs by way of a signal-mediated apoptotic process. PD is also characterized by intracellular proteinaceous inclusions or Lewy bodies. Proteolytic stress arises as a consequence of the excessive production of misfolded proteins, which exceed the capacity of the ubiquitin-proteasome system to degrade them. Recent genetic and laboratory studies support the possible relevance of proteolytic stress to both familial and sporadic forms of PD. Postmortem studies have shown that in the SNc of sporadic PD patients there are reduced levels of the alpha subunit of the 20S proteasome and reduced proteolytic enzyme activities. A determination as to the precise cause of cell death in PD, and the identification of specific targets for the development of drugs that might modify disease progression is one of the most critical goals in PD research. It is anticipated that over the next few years there will be a flurry of scientific activity examining the mechanism of cell death and putative neuroprotective interventions.

    Topics: alpha-Synuclein; Cell Death; Disease Progression; Dopamine; Humans; Leucine-Rich Repeat Serine-Threonine Protein Kinase-2; Lewy Bodies; Nerve Degeneration; Oxidative Stress; Oxygen; Parkinson Disease; Point Mutation; Proteasome Endopeptidase Complex; Protein Serine-Threonine Kinases; Signal Transduction; Ubiquitin

2007
Hero versus antihero: the multiple roles of alpha-synuclein in neurodegeneration.
    Experimental neurology, 2006, Volume: 199, Issue:2

    Topics: alpha-Synuclein; Animals; Humans; Models, Biological; Nerve Degeneration; Neurodegenerative Diseases

2006
Regional alpha-synuclein aggregation, dopaminergic dysregulation, and the development of drug-related visual hallucinations in Parkinson's disease.
    The Journal of neuropsychiatry and clinical neurosciences, 2006,Spring, Volume: 18, Issue:2

    Visual hallucinations in Parkinson's disease are usually treatment-related and occur in at least 30% of patients. Although their clinical and epidemiological features have been extensively reviewed, their etiopathogenesis remains a matter of debate. Based on the current evidence available, this review suggests that regional neurodegeneration of the ventral dopaminergic pathway, as evident in the aggregation of the protein alpha-synuclein, is the main event linked to the development of visual hallucinations in Parkinson's disease. Denervation supersensitivity of dopaminergic receptors in ventral striatal and mesocorticolimbic areas as well as defective synaptic buffering ability due to the loss of dopaminergic presynaptic terminals and dopamine transporter may be among the key factors leading to visual hallucinations in Parkinson's disease.

    Topics: alpha-Synuclein; Antiparkinson Agents; Cerebral Cortex; Corpus Striatum; Dopamine; Dopamine Plasma Membrane Transport Proteins; Hallucinations; Humans; Mesencephalon; Nerve Degeneration; Neural Pathways; Parkinson Disease; Receptors, Dopamine; Receptors, Presynaptic

2006
Cellular pathology in multiple system atrophy.
    Neuropathology : official journal of the Japanese Society of Neuropathology, 2006, Volume: 26, Issue:4

    Multiple system atrophy (MSA) is a sporadic, adult-onset neurodegenerative disease, which is characterized by striatonigral degeneration, olivopontocerebellar atrophy, and preganglionic autonomic lesions in any combination. The histological hallmark is the presence of argyrophilic fibrillary inclusions in the oligodendrocytes, referred to as glial cytoplasmic inclusions (GCIs). Fibrillary inclusions are also found in the neuronal somata, axons, and nucleus. Neuronal cytoplasmic inclusions are frequently found in the pontine and inferior olivary nuclei. Since the discovery of alpha-synuclein as a major component of glial and neuronal inclusions in MSA, two neurodegenerative processes have been considered in this disease: one is due to the widespread occurrence of GCIs associated with oligodendroglia-myelin degeneration (oligodendrogliopathy) in the central nervous system, and the other is due to the filamentous aggregation of alpha-synuclein in the neurons in several brain regions. These two degenerative processes might synergistically cause neuronal depletion in MSA.

    Topics: alpha-Synuclein; Animals; Brain; Humans; Immunohistochemistry; Inclusion Bodies; Multiple System Atrophy; Nerve Degeneration; Neuroglia; Neurons

2006
Molecular mechanisms of nigral neurodegeneration in Park2 and regulation of parkin protein by other proteins.
    Journal of neural transmission. Supplementum, 2006, Issue:70

    Most of the patients with Parkinson's disease (PD) are sporadic. However, Since identification of monogenic forms of PD, the contribution of genetic factors to the pathogenesis of sporadic PD is proposed as one of major risk factors. Indeed, this is supported by the demonstration of the high concordance in twins, increased risk among relatives of PD patients in case control and family studies. Thus, the functional analysis for the gene products for familial PD provides us a good hint to elucidate the pathogenesis of nigral degeneration. For example, although alpha-synuclein is involved in a rare dominant form of familial PD with dopa responsive parkinsonian features, this molecule is a major component of and Lewy bodies (LBs). In contrast, Park2 (parkin-related disease) is the most frequent form among patients with young-onset PD. However, Park2 brains generally lack the formation of LBs. In the other word, parkin responsible for Park2 is essential for the formation of LBs. Thus, both alpha-synuclein and parkin are speculated to share a common pathway. Here, we reviewed the parkin function and molecular mechanisms of Park2.

    Topics: alpha-Synuclein; Gene Expression Regulation; Humans; Nerve Degeneration; Nerve Tissue Proteins; Parkinson Disease; Substantia Nigra; Ubiquitin-Protein Ligases

2006
Role of DJ-1 in Parkinson's disease.
    Journal of molecular neuroscience : MN, 2006, Volume: 29, Issue:3

    Parkinson's disease (PD), one of the most common neurodegenerative diseases, is a multifactorial disease caused by both genetic and environmental factors. Although most patients suffering from PD have a sporadic disease, several genetic causes have been identified in recent years, including alpha-synuclein, parkin, PINK1, dardarin (LRRK2), and DJ-1. DJ-1 deletions and point mutations have been found worldwide, and loss of functional protein was shown to cause autosomal recessive PD. Moreover, DJ-1 immunoreactive inclusions are found in other alpha-synucleopathies and tauopathies, indicating that different neurodegenerative diseases might share a common mechanism in which DJ-1 might play a key role. The function of DJ-1 is still unknown; however, it is associated with various cellular processes, including response to oxidative stress, cellular transformation, RNAbinding, androgen-receptor signaling, spermatogenesis, and fertilization. This article reviews the current knowledge on DJ-1, focusing on its importance in the pathogenesis of PD.

    Topics: alpha-Synuclein; Animals; Cell Transformation, Viral; Dopamine; Drosophila melanogaster; Drosophila Proteins; Genes, Recessive; Humans; Intracellular Signaling Peptides and Proteins; Mice; Mice, Knockout; Mutation; Nerve Degeneration; Nerve Tissue Proteins; Oncogene Proteins; Oxidative Stress; Parkinsonian Disorders; Peroxiredoxins; Protein Deglycase DJ-1; Tauopathies; Ubiquitin-Protein Ligases

2006
Nosology of Parkinson's disease: looking for the way out of a quagmire.
    Neuron, 2005, Aug-18, Volume: 47, Issue:4

    The discovery of SNCA mutations pathogenic for autosomal-dominant Lewy body Parkinson's disease (PD) in 1997 heralded a revolution in understanding the molecular and genetic basis of PD. Indeed, it now is clear that Lewy body PD is one of many neurodegenerative parkinsonian disorders that result from nigrostriatal degeneration caused by diverse mechanisms. However, to capitalize on these new insights and facilitate efforts to improve the diagnosis and therapy of neurodegenerative movement disorders, it is timely to define a nosology for these diseases that is based on their genetic and molecular underpinnings, as proposed here.

    Topics: alpha-Synuclein; Animals; Genetic Predisposition to Disease; Humans; Mutation; Nerve Degeneration; Nerve Tissue Proteins; Parkinson Disease; Substantia Nigra; Synucleins; Terminology as Topic

2005
Proteomic approach to studying Parkinson's disease.
    Molecular neurobiology, 2004, Volume: 29, Issue:3

    Parkinson's disease is a common age-related neurodegenerative disease characterized pathologically by a loss of dopaminergic neurons in the substantia nigra with resultant depletion of striatal dopamine and presence of Lewy bodies in the remaining neurons. The Lewy body contains numerous functional and structural proteins, including alpha-synuclein and ubiquitin; aggregation of alpha-synuclein is thought to be important in Lewy body formation as well as neurodegeneration, although the detailed mechanisms remain to be defined. Increasing evidence has suggested that mitochondrial dysfunction, increased oxidative stress, and dysfunction of the ubiquitin-proteasome system may be involved in alpha-synuclein aggregation, Lewy body formation, and neurodegeneration. However, how these processes are related to each other is not fully understood, given that there are Parkinsonian animal models as well as human diseases with significant nigral neurodegeneration regardless of whether Lewy bodies form or not. This review summarizes the current related research fields and proposes a proteomic approach to investigate the mechanisms that may dictate alpha-synuclein aggregation, Lewy body formation, and neurodegeneration.

    Topics: alpha-Synuclein; Animals; Humans; Lewy Bodies; Models, Animal; Nerve Degeneration; Nerve Tissue Proteins; Parkinson Disease; Proteomics; Substantia Nigra; Synucleins; Ubiquitin

2004
The role of alpha-synuclein in both neuroprotection and neurodegeneration.
    Annals of the New York Academy of Sciences, 2004, Volume: 1035

    Although alpha-synuclein is a central player in the pathophysiology of the dopaminergic neurodegeneration that occurs in Parkinson's disease (PD), emerging results suggest that the fundamental property of the wild-type form of this protein may be one of neuroprotection, as it can inhibit apoptosis in response to various pro-apoptotic stimuli. Such properties may be lost by its familial PD-linked mutations upon alterations in its expression levels or clearance (overexpression of the gene, reduced protein degradation) or following exposure to certain neurotoxins. Moreover, converging observations suggest that a primary function for alpha-synuclein in dopaminergic neurons may be the regulation of dopamine content and tone at the synapse. In this paper, we review how, indeed, alpha-synuclein regulates both the synthesis of dopamine, its storage into vesicles, its release in the synapse, and its re-uptake into the dopaminergic neurons. We also show how disruption of these events, and of the neuroprotective effects of alpha-synuclein, can initiate the observed neurotoxicity of alpha-synuclein in dopaminergic neurons and the genesis of the degenerative processes associated with PD.

    Topics: alpha-Synuclein; Animals; Dopamine; Humans; Models, Neurological; Nerve Degeneration; Neuroprotective Agents

2004
Proteolytic stress: a unifying concept for the etiopathogenesis of Parkinson's disease.
    Annals of neurology, 2003, Volume: 53 Suppl 3

    The etiopathogenesis of Parkinson's disease (PD) has been elusive. Recently, several lines of evidence have converged to suggest that defects in the ubiquitin-proteasome system and proteolytic stress underlie nigral pathology in both familial and sporadic forms of the illness. In support of this concept, mutations in alpha-synuclein that cause the protein to misfold and resist proteasomal degradation cause familial PD. Similarly, mutations in two enzymes involved in the normal function of the ubiquitin-proteasome system, parkin and ubiquitin C-terminal hydrolase L1, are also associated with hereditary PD. Furthermore, structural and function defects in 26/20S proteasomes with accumulation and aggregation of potentially cytotoxic abnormal proteins have been identified in the substantia nigra pars compacta of patients with sporadic PD. Thus, a defect in protein handling appears to be a common factor in sporadic and the various familial forms of PD. This hypothesis may also account for the vulnerability of the substantia nigra pars compacta in PD, why the disorder is age related, and the nature of the Lewy body. It has also facilitated the development of experimental models that recapitulate the behavioral and pathological features of PD, and hopefully will lead to the development of novel neuroprotective therapies for the disorder.

    Topics: alpha-Synuclein; Cysteine Endopeptidases; Humans; Lewy Bodies; Ligases; Multienzyme Complexes; Nerve Degeneration; Nerve Tissue Proteins; Parkinson Disease; Peptide Hydrolases; Point Mutation; Proteasome Endopeptidase Complex; Substantia Nigra; Synucleins; Thiolester Hydrolases; Ubiquitin Thiolesterase; Ubiquitin-Protein Ligases

2003
Modeling CNS neurodegeneration by overexpression of disease-causing proteins using viral vectors.
    Trends in neurosciences, 2003, Volume: 26, Issue:7

    Defective handling of proteins is a central feature of major neurodegenerative diseases. The discovery that neuronal dysfunction or degeneration can be caused by mutations in single cellular proteins has given new opportunities to model the underlying disease processes by genetic modification of cells in vitro or by generation of transgenic animals carrying the disease-causing gene. Recent developments in recombinant viral-vector technology have opened up an interesting alternative possibility, based on direct gene transfer to selected subregions or subsets of neurons in the brain. Using the highly efficient adeno-associated virus or lentivirus vectors, recent reports have shown that overexpression of mutated human huntingtin or alpha-synuclein in neurons in the striatum or substantia nigra induces progressive neuropathology and neurodegeneration, similar to that seen in Huntington's and Parkinson's diseases. Targeted overexpression of disease-causing genes by recombinant viral vectors provides a new and highly flexible approach for in vivo modeling of neurodegenerative diseases, not only in mice and rats but also in primates.

    Topics: alpha-Synuclein; Animals; Animals, Genetically Modified; Central Nervous System; Corpus Striatum; Dependovirus; Disease Models, Animal; Gene Transfer Techniques; Genetic Vectors; Huntingtin Protein; Huntington Disease; Lentivirus; Mutation; Nerve Degeneration; Nerve Tissue Proteins; Neurons; Nuclear Proteins; Parkinson Disease; Substantia Nigra; Synucleins

2003
What have we learnt from CDNA microarray gene expression studies about the role of iron in MPTP induced neurodegeneration and Parkinson's disease?
    Journal of neural transmission. Supplementum, 2003, Issue:65

    There have been numerous hypotheses concerning the etiology and mechanism of dorsal raphe dopaminergic neurodegeneration in Parkinson's disease and its animal models, MPTP (N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) and 6-hydroxydopamine. The advent of cDNA microarray gene expression where expression of thousands of genes can be globally assessed has indicated that mechanism of neurodegeneration by MPTP is a complex cascade of vicious circles. One of these is the alteration of genes associated with iron metabolism, a transitional metal closely associated with inducing the formation of reactive oxygen species and inducing oxidative stress. cDNA gene expression analyses support the established hypothesis of oxidative induced neurodegeneration involving iron deposition in substantia nigra pars compacta (SNPC) parkinsonian brains. The regulation of cellular iron metabolism has been further enhanced by the recent discovery of two iron regulatory proteins, IRP1 and IRP2 which control the level of iron with in the cell. When the cellular level of iron increases IRP2 is degraded by ubiquitination and no further iron accumulates. The reverse occurs when the level of iron is low within the cell. Knock-out IRP1 and IRP2 mice have shown that in latter mice brain iron accumulation precedes the neurodegeneration, ataxia and bradykinesia observed in these animals. Indeed MPTP treatment, which results in iron accumulation in SNCP, abolishes IRP2 with the concomitant increase in alpha-synuclein. Iron chelators such as R-apomorphine and EGCG, which protect against MPTP neurotoxicity, prevent the loss of IRP2 and the increase in alpha-synuclein. The presence of iron together with alpha-synuclein in SNPC may be detrimental for dopaminergic neurons. Since, iron has been shown to cause aggregation of alpha-synuclein to a neurotoxic agent. The use of iron chelators penetrating the blood brain barrier as neuroprotective drugs has been envisaged.

    Topics: 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine; alpha-Synuclein; Animals; Brain; Cell Death; Dopamine; Dopamine Agents; Humans; Iron; Iron Chelating Agents; Nerve Degeneration; Nerve Tissue Proteins; Neurons; Neuroprotective Agents; Oligonucleotide Array Sequence Analysis; Parkinson Disease; Substantia Nigra; Synucleins

2003
[Lewy bodies, a misleading marker for Parkinson's disease?].
    Bulletin de l'Academie nationale de medecine, 2003, Volume: 187, Issue:2

    The Lewy body, an eosinophilic inclusion around 10 microns in diameter, is localised in the neuronal perikaryon. Its dense core is surrounded by a clear halo, which is lacking in the so-called "cortical Lewy bodies". Numerous proteins have been identified in Lewy bodies, among which the three neurofilament isoforms, ubiquitin and proteasome subunits. More recently, alpha-synuclein--a pre-synaptic protein--has been found to be the essential constituent of the Lewy body. Alpha-synuclein antibody has greatly increased the sensitivity of the neuropathological examination: it has emphasized the frequency of "Lewy neurites" (accumulation of alpha--synuclein in neuronal processes) and has shown the importance of extra-nigral pathology. Lewy bodies and neurites are indeed to be found in many areas of the central and peripheral nervous system: stellate ganglia, cardiac and enteric plexus, pigmented nuclei of the brainstem, basal nucleus of Meynert, amygdala, limbic nuclei of the thalamus, parahippocampal and cingulate gyri, insula and isocortex. Lewy body diseases include at least three clinical syndromes: 1) idiopathic Parkinson disease in which the brainstem bears the brunt of the pathology 2) Parkinson disease dementia in which Lewy lesions are found in the brainstem and are also abundant in the isocortex. A large number of senile plaques is frequently associated. 3) In dementia with Lewy bodies, the same lesions are observed but the cognitive deficit occurs first or shortly (less than one year) after the motor symptoms.

    Topics: Aging; alpha-Synuclein; Alzheimer Disease; Brain Stem; Diagnosis, Differential; Humans; Lewy Bodies; Lewy Body Disease; Locus Coeruleus; Nerve Degeneration; Nerve Tissue Proteins; Parkinson Disease; Synucleins; Ubiquitin

2003
Neuritic alterations and neural system dysfunction in Alzheimer's disease and dementia with Lewy bodies.
    Neurochemical research, 2003, Volume: 28, Issue:11

    Alzheimer's disease (AD) and dementia with Lewy bodies (DLB) are neurodegenerative disorders that share progressive dementia as the common major clinical symptom. Damages to memory-related brain structures are the likely pathological correlate, and in both illnesses deposition of amyloidogenic proteins are present mainly within these limbic structures. Amyloid-beta-positive plaques and phospho-tau-positive neurofibrillary tangles are the main feature of AD and alpha-synuclein-positive Lewy bodies and Lewy neurites are found in DLB. Interestingly the associated proteins also interfere with synaptic function and synaptic plasticity. Here, we propose that the same neuronal circuits are disturbed within the hippocampal formation in AD and DLB and that in both diseases the associated proteins might lead to changes in synaptic plasticity and function. Thus both classic neuropathological changes and cellular dysfunctions might contribute to the cognitive impairments in AD and DLB.

    Topics: alpha-Synuclein; Alzheimer Disease; Amyloid beta-Peptides; Animals; Humans; Lewy Body Disease; Memory Disorders; Nerve Degeneration; Nerve Tissue Proteins; Neural Pathways; Neurites; Neuronal Plasticity; Synaptic Transmission; Synucleins

2003
Cycles of aberrant synaptic sprouting and neurodegeneration in Alzheimer's and dementia with Lewy bodies.
    Neurochemical research, 2003, Volume: 28, Issue:11

    Alzheimer's disease (AD) and dementia with Lewy bodies (DLB) are the most common neurodegenerative disorders affecting the elderly. The cognitive and motor deficits in these diseases are associated with the disruption of neuritic substructure, loss of synaptic contacts in selectively vulnerable circuitries, and aberrant sprouting. Where as in AD, accumulation of misfolded forms of Abeta triggers neurodegeneration, in DLB accumulation of alpha-synuclein might play a central role. The mechanisms by which oligomeric forms of these proteins might lead to cycles of synapse loss and aberrant sprouting are currently under investigation. Several possibilities are being considered, including mitochondrial damage, caspase activation, lysosomal leakage, fragmentation of the Golgi apparatus, interference with synaptic vesicle transport and function, and interference with gene transcription and signaling. Among them, recent lines of research support the possibility that alterations in signaling pathways such extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK) and p38 relevant to synaptic plasticity and cell survival might play a pivotal role. A wide range of cellular functions are affected by the accumulation of misfolded Abeta and alpha-synuclein; thus it is possible that a more fundamental cellular alteration may underlie the mechanisms of synaptic pathology in these disorders. Among them, one possibility is that scaffold proteins, such as caveolin and JNK-interacting protein (JIP), which are necessary to integrate signaling pathways, are affected, leading to cycles of synapse loss and aberrant sprouting. This is significant because both caveolar dysfunction and altered axonal plasticity might be universally important in the pathogenesis of various neurodegenerative disorders, and therefore these signaling pathways might be common therapeutic targets for these devastating diseases.

    Topics: alpha-Synuclein; Alzheimer Disease; Amyloid beta-Peptides; Animals; Humans; Lewy Body Disease; Memory Disorders; Nerve Degeneration; Nerve Tissue Proteins; Neural Pathways; Neurites; Neuronal Plasticity; Synaptic Transmission; Synucleins

2003
Molecular pathways of neurodegeneration in Parkinson's disease.
    Science (New York, N.Y.), 2003, Oct-31, Volume: 302, Issue:5646

    Parkinson's disease (PD) is a complex disorder with many different causes, yet they may intersect in common pathways, raising the possibility that neuroprotective agents may have broad applicability in the treatment of PD. Current evidence suggests that mitochondrial complex I inhibition may be the central cause of sporadic PD and that derangements in complex I cause alpha-synuclein aggregation, which contributes to the demise of dopamine neurons. Accumulation and aggregation of alpha-synuclein may further contribute to the death of dopamine neurons through impairments in protein handling and detoxification. Dysfunction of parkin (a ubiquitin E3 ligase) and DJ-1 could contribute to these deficits. Strategies aimed at restoring complex I activity, reducing oxidative stress and alpha-synuclein aggregation, and enhancing protein degradation may hold particular promise as powerful neuroprotective agents in the treatment of PD.

    Topics: alpha-Synuclein; Animals; Animals, Genetically Modified; Brain; Cysteine Endopeptidases; Dopamine; Electron Transport Complex I; Humans; Mitochondria; Multienzyme Complexes; Mutation; Nerve Degeneration; Nerve Tissue Proteins; Neurons; Oxidative Stress; Parkinson Disease; Parkinsonian Disorders; Proteasome Endopeptidase Complex; Synucleins; Ubiquitin; Ubiquitin-Protein Ligases

2003
Caught in the act: alpha-synuclein is the culprit in Parkinson's disease.
    Neuron, 2003, Oct-30, Volume: 40, Issue:3

    Previous reports on Parkinson's disease indicate that genetic mutations in alpha-synuclein result in the aberrant accumulation of this protein, causing toxic gain of function leading to the development of Parkinson's. A recent report on the Iowan kindred, an extended pedigree with an autosomal dominant form of this disease, provides new mechanistic insight into Parkinson's disease by showing that an elevation in wild-type alpha-synuclein protein is sufficient to develop the early-onset form of the disorder. This review discusses how insights gained from these studies of alpha-synuclein may direct future research into Parkinson's disease.

    Topics: alpha-Synuclein; Animals; Brain; Genes, Dominant; Humans; Lewy Bodies; Nerve Degeneration; Nerve Tissue Proteins; Parkinson Disease; Proteome; Synucleins

2003
Familial Parkinson's disease. Alpha-synuclein and parkin.
    Advances in neurology, 2001, Volume: 86

    We have reviewed recent progress in establishing the function of alpha-synuclein and parkin in relation to nigral degeneration in autosomal dominant and autosomal recessive PD. Mutations of alpha-synuclein (Ala53Thr and Ala30Pro) cause a form of autosomal dominant PD with early onset. Parkin is a novel protein expressed in the cytoplasm, including the terminal regions and Golgi apparatus. Mutations of parkin cause a form of autosomal recessive young-onset PD (ARJP). Both proteins appear to be associated with fast axonal transport. In addition, in sporadic PD, normal alpha-synuclein shows an increased tendency to self-aggregate. Thus, altered axonal transport of presynaptic proteins appears to play a crucial role in neurodegeneration in PD.

    Topics: alpha-Synuclein; Humans; Ligases; Nerve Degeneration; Nerve Tissue Proteins; Parkinsonian Disorders; Synucleins; Ubiquitin-Protein Ligases

2001
Involvement of alpha-synuclein in Parkinson's disease and other neurodegenerative disorders.
    Journal of neural transmission (Vienna, Austria : 1996), 2000, Volume: 107, Issue:1

    A major step in the elucidation of the pathogenesis of neurodegenerative disorders was the identification of a mutation in the alpha-synuclein gene in autosomal dominant Parkinson's disease (PD). Alpha-synuclein is the main component of Lewy bodies (LB), the neuropathological hallmark of PD. Moreover, a fragment of alpha-synuclein (NAC) is the second major component of amyloid plaques in Alzheimer's disease (AD). Recent studies of other neurodegenerative disorders such as dementia with LB (DLB), multiple system atrophy (MSA) and amyotrophic lateral sclerosis (ALS) also revealed intracellular accumulations of alpha-synuclein in affected brain regions. This may indicate that these disorders partially share common pathogenic mechanisms. Recent data provide first insights into the physiological function of alpha-synuclein and support the concept of an essential role of alpha-synuclein in neurodegeneration. Increasing knowledge on the pathogenic molecular mechanisms of neurodegeneration and of the pathophysiological function of alpha-synuclein in particular may influence future development of therapeutic strategies in neurodegenerative disorders.

    Topics: alpha-Synuclein; Alzheimer Disease; Humans; Nerve Degeneration; Nerve Tissue Proteins; Parkinson Disease; Synucleins

2000
The synucleins: a family of proteins involved in synaptic function, plasticity, neurodegeneration and disease.
    Trends in neurosciences, 1998, Volume: 21, Issue:6

    Synuclein proteins are produced, in vertebrates, by three genes. They share structural resemblance to apolipoproteins, but are abundant in the neuronal cytosol and present in enriched amounts at presynaptic terminals. Synucleins have been specifically implicated in three diseases:Alzheimer's (AD), Parkinson's (PD) and breast cancer. In AD, a peptide derived from alpha-synuclein forms an intrinsic component of plaque amyloid. In PD, an alpha-synuclein allele is genetically linked to several independent familial cases, and the protein appears to accumulate in Lewy bodies. In breast cancer, increased expression of gamma-synuclein correlates with disease progression. In songbirds, alpha-synuclein expression is correlated with plasticity in the developing song control system. Although the normal function of synucleins is unknown, a role in membrane plasticity seems likely.

    Topics: alpha-Synuclein; Animals; gamma-Synuclein; Humans; Molecular Sequence Data; Nerve Degeneration; Nerve Tissue Proteins; Nervous System Diseases; Neuronal Plasticity; Sequence Homology, Amino Acid; Synapses; Synucleins

1998
Genetic dissection of familial Parkinson's disease.
    Molecular medicine today, 1998, Volume: 4, Issue:10

    In the past few years, the genetic contribution to Parkinson's disease (PD) has gained major attention and has resulted in the identification of the first mutant gene, called alpha-synuclein, involved in the pathogenesis of autosomal-dominant PD. alpha-Synuclein is a major component of Lewy bodies, which are a neuropathological feature of PD. Furthermore, deletions in the parkin gene have been identified as the primary cause in rare forms of autosomal-recessive juvenile PD. The elucidation of polygenic changes in the dopamine pathway, mitochondrial dysfunction, and metabolism of xenobiotics is now technically possible by means of association and genotype studies. The increasing knowledge of the pathogenesis of PD at a molecular level will have important implications for the development of individual therapeutic strategies to prevent disease progression.

    Topics: alpha-Synuclein; Amino Acid Sequence; Animals; Forecasting; Genes, Dominant; Genes, Recessive; Humans; Molecular Sequence Data; Nerve Degeneration; Nerve Tissue Proteins; Parkinson Disease; Synucleins

1998

Other Studies

328 other study(ies) available for alpha-synuclein and Nerve-Degeneration

ArticleYear
Pathological α-synuclein accumulation, CSF metabolites changes and brain microstructures in cynomolgus monkeys treated with 6-hydroxydopamine.
    Neurotoxicology, 2023, Volume: 94

    The lack of evidence indicating the accumulation of phosphorylated α-synuclein (P-α-syn), a neuropathological hallmark of Parkinson disease (PD), limits the application of 6-OHDA animal models. In cynomolgus monkeys received unilateral 6-hydroxydopamine (6-OHDA) injection, we identified nigrostriatal dysfunction related behavioral defects, such as the increase of PD score, decrease of locomotor activities, and exhibition of typical rotations. We found the dopaminergic neurons were significantly reduced and had fragmented morphology in substantia nigra (SN). Furthermore, insoluble P-α-syn aggregates were observed. The P-α-syn aggregates were extracellular distributed and had typical morphology of inclusion. Immunofluorescence staining showed that the P-α-syn colocalized with ubiquitin (Ub) and p62. We also found there were more actived astrocytes and microglial in SN and striatum, reflecting neuroinflammations increase in nigrostriatal pathway. At last, to determine the long-term consequence of dopamine (DA) neuron loss induced by 6-OHDA injection, the changes of cerebrospinal fluid (CSF) neurotransmitters over time as well as the brain microstructure alternations were examined. The dopamine-related metabolites were decreased after 6-OHDA injection reflecting dopaminergic neuron loss. The levels of γ-aminobutyric acid (GABA) and acetylcholine (Ach) showed an increasing trend but not significant. By diffusion tensor Magnetic Resonance Imaging (MRI) image scans, the fractional anisotropy (FA) value in the ipsilateral SN and caudate was found to reduce, which indicated neural fiber injury. Therefore, these results suggested that α-syn pathology might participate in process of 6-OHDA injuring DA neurons, and may expand the application of 6-OHDA monkeys on investigations into the pathogenesis of PD.

    Topics: alpha-Synuclein; Animals; Brain; Disease Models, Animal; Dopamine; Dopaminergic Neurons; Macaca fascicularis; Nerve Degeneration; Oxidopamine; Parkinson Disease; Substantia Nigra

2023
Opposing effects of β-2 and β-1 adrenergic receptor signaling on neuroinflammation and dopaminergic neuron survival in α-synuclein-mediated neurotoxicity.
    Journal of neuroinflammation, 2023, Mar-02, Volume: 20, Issue:1

    Noradrenergic neurons in the locus coeruleus (LC) are the primary source of norepinephrine (NE) in the brain and degeneration of these neurons is reported in the early stages of Parkinson's disease (PD), even prior to dopaminergic neuron degeneration in the substantia nigra (SN), which is a hallmark of PD pathology. NE depletion is generally associated with increased PD pathology in neurotoxin-based PD models. The effect of NE depletion in other models of PD-like α-synuclein-based models is largely unexplored. In PD models and in human patients, β-adrenergic receptors' (AR) signaling is associated with a reduction of neuroinflammation and PD pathology. However, the effect of NE depletion in the brain and the extent of NE and β-ARs signaling involvement in neuroinflammation, and dopaminergic neuron survival is poorly understood.. Two mouse models of PD, a 6OHDA neurotoxin-based model and a human α-synuclein (hα-SYN) virus-based model of PD, were used. DSP-4 was used to deplete NE levels in the brain and its effect was confirmed by HPLC with electrochemical detection. A pharmacological approach was used to mechanistically understand the impact of DSP-4 in the hα-SYN model of PD using a norepinephrine transporter (NET) and a β-AR blocker. Epifluorescence and confocal imaging were used to study changes in microglia activation and T-cell infiltration after β1-AR and β2-AR agonist treatment in the hα-SYN virus-based model of PD.. Consistent with previous studies, we found that DSP-4 pretreatment increased dopaminergic neuron loss after 6OHDA injection. In contrast, DSP-4 pretreatment protected dopaminergic neurons after hα-SYN overexpression. DSP-4-mediated protection of dopaminergic neurons after hα-SYN overexpression was dependent on β-AR signaling since using a β-AR blocker prevented DSP-4-mediated dopaminergic neuron protection in this model of PD. Finally, we found that the β-2AR agonist, clenbuterol, reduced microglia activation, T-cell infiltration, and dopaminergic neuron degeneration, whereas xamoterol a β-1AR agonist showed increased neuroinflammation, blood brain barrier permeability (BBB), and dopaminergic neuron degeneration in the context of hα-SYN-mediated neurotoxicity.. Our data demonstrate that the effects of DSP-4 on dopaminergic neuron degeneration are model specific, and suggest that in the context of α-SYN-driven neuropathology, β2-AR specific agonists may have therapeutic benefit in PD.

    Topics: alpha-Synuclein; Animals; Dopaminergic Neurons; Humans; Mice; Nerve Degeneration; Neuroinflammatory Diseases; Neurotoxicity Syndromes; Neurotoxins; Parkinson Disease; Receptors, Adrenergic, beta-1; Receptors, Adrenergic, beta-2

2023
Rose essential oil diminishes dopaminergic neuron degenerations and reduces α-synuclein aggregation in Caenorhabditis elegans models of Parkinson's disease.
    Phytotherapy research : PTR, 2023, Volume: 37, Issue:7

    Parkinson's disease (P.D.) is the second most progressive neurodegenerative disorder in the elderly. Degeneration of dopaminergic (DA) neurons and α-synuclein (α-Syn) accumulated toxicity is the major contributor to this disease. At present, the disease has no effective treatment. Many recent studies focus on identifying novel therapeutics that provide benefits to stop the disease progression in P.D. patients. Screening novel and effective drugs in P.D. animal models is time- and cost-consuming. Rose Essential Oil (REO) extracted from Rosa Rugosa species (R. Setate × R. Rugosa). REO contains Citronellol, Geraniol, and Octadiene that possess anti-Aβ, anti-oxidative, and anti-depression-like properties, but no reports have defined the REO effect on P.D. yet. The present study examines the REO neuroprotective potential in transgenic Caenorhabditis elegans P.D. models. We observed that REO reduced α-Syn aggregations and diminished DA neuron degenerations induced by 6-OHDA, reduced food-sensing behavioural disabilities, and prolonged the lifespan of the nematode. Moreover, REO augmented the chymotrypsin-like proteasome and SOD-3 activities. Further, we observed the anti-oxidative role of REO by reducing internal cells ROS. Together, these findings supported REO as an anti-PD drug and may exert its effects by lowering oxidative stress via the anti-oxidative pathway.

    Topics: alpha-Synuclein; Animals; Animals, Genetically Modified; Caenorhabditis elegans; Disease Models, Animal; Dopaminergic Neurons; Nerve Degeneration; Oils, Volatile; Parkinson Disease; Rosa

2023
Cntnap4 partial deficiency exacerbates α-synuclein pathology through astrocyte-microglia C3-C3aR pathway.
    Cell death & disease, 2023, 04-22, Volume: 14, Issue:4

    Parkinson's disease (PD) is the most common progressive neurodegenerative movement disorder, which is characterized by dopaminergic (DA) neuron death and the aggregation of neurotoxic α-synuclein. Cntnap4, a risk gene of autism, has been implicated to participate in PD pathogenesis. Here we showed Cntnap4 lacking exacerbates α-synuclein pathology, nigrostriatal DA neuron degeneration and motor impairment, induced by injection of adeno-associated viral vector (AAV)-mediated human α-synuclein overexpression (AAV-hα-Syn). This scenario was further validated in A53T α-synuclein transgenic mice injected with AAV-Cntnap4 shRNA. Mechanistically, α-synuclein derived from damaged DA neuron stimulates astrocytes to release complement C3, activating microglial C3a receptor (C3aR), which in turn triggers microglia to secrete complement C1q and pro-inflammatory cytokines. Thus, the astrocyte-microglia crosstalk further drives DA neuron death and motor dysfunction in PD. Furthermore, we showed that in vivo depletion of microglia and microglial targeted delivery of a novel C3aR antagonist (SB290157) rescue the aggravated α-synuclein pathology resulting from Cntnap4 lacking. Together, our results indicate that Cntnap4 plays a key role in α-synuclein pathogenesis by regulating glial crosstalk and may be a potential target for PD treatment.

    Topics: alpha-Synuclein; Animals; Astrocytes; Complement C3; Dopaminergic Neurons; Ferroptosis; Humans; Inflammation; Male; Membrane Proteins; Mice; Mice, Inbred C57BL; Mice, Transgenic; Microglia; Mitochondria; Nerve Degeneration; Nerve Tissue Proteins; Parkinson Disease; Receptors, Complement

2023
Asymmetric Presynaptic Depletion of Dopamine Neurons in a
    International journal of molecular sciences, 2023, May-11, Volume: 24, Issue:10

    Parkinson's disease (PD) often displays a strong unilateral predominance in arising symptoms. PD is correlated with dopamine neuron (DAN) degeneration in the substantia nigra pars compacta (SNPC), and in many patients, DANs appear to be affected more severely on one hemisphere than the other. The reason for this asymmetric onset is far from being understood.

    Topics: alpha-Synuclein; Animals; Disease Models, Animal; Dopaminergic Neurons; Drosophila; Drosophila melanogaster; Humans; Nerve Degeneration; Parkinson Disease; Substantia Nigra

2023
Transcriptomic profiling of Parkinson's disease brains reveals disease stage specific gene expression changes.
    Acta neuropathologica, 2023, Volume: 146, Issue:2

    Parkinson´s disease (PD) is a progressive neurodegenerative disorder characterized by both motor and non-motor symptoms. Aggravation of symptoms is mirrored by accumulation of protein aggregates mainly composed by alpha-synuclein in different brain regions, called Lewy bodies (LB). Previous studies have identified several molecular mechanisms as autophagy and inflammation playing a role in PD pathogenesis. Increased insights into mechanisms involved in early disease stages and driving the progression of the LB pathology are required for the development of disease-modifying strategies. Here, we aimed to elucidate disease stage-specific transcriptomic changes in brain tissue of well-characterized PD and control donors. We collected frontal cortex samples from 84 donors and sequenced both the coding and non-coding RNAs. We categorized our samples into groups based on their degree of LB pathology aiming to recapitulate a central aspect of disease progression. Using an analytical pipeline that corrected for sex, age at death, RNA quality, cell composition and unknown sources of variation, we found major disease stage-specific transcriptomic changes. Gene expression changes were most pronounced in donors at the disease stage when microscopic LB changes first occur in the sampled brain region. Additionally, we identified disease stage-specific enrichment of brain specific pathways and immune mechanisms. On the contrary, we showed that mitochondrial mechanisms are enriched throughout the disease course. Our data-driven approach also suggests a role for several poorly characterized lncRNAs in disease development and progression of PD. Finally, by combining genetic and epigenetic information, we highlighted two genes (MAP4K4 and PHYHIP) as candidate genes for future functional studies. Together our results indicate that transcriptomic dysregulation and associated functional changes are highly disease stage-specific, which has major implications for the study of neurodegenerative disorders.

    Topics: alpha-Synuclein; Brain; Humans; Intracellular Signaling Peptides and Proteins; Lewy Bodies; Nerve Degeneration; Parkinson Disease; Protein Serine-Threonine Kinases; Transcriptome

2023
Feasibility of combining alpha-synuclein aggregation and 6-OHDA in embryonic midbrain culture for modeling dopamine neuron degeneration.
    Neuroscience letters, 2023, 11-01, Volume: 816

    Parkinson's disease (PD) is characterized by the loss of nigrostriatal dopamine (DA) neurons and the presence of alpha-synuclein (αSyn)-positive Lewy body (LB) pathology. In this study, we attempted to recapitulate both these features in a novel in vitro model for PD. To achieve this, we combined the αSyn pre-formed fibril (PFF)-seeded LB-like pathology with 6-hydroxydopamine (6-OHDA)-induced mitochondrial toxicity in mouse embryonic midbrain cultures. To pilot the model for therapeutics testing, we assessed the effects of cerebral dopamine neurotrophic factor (CDNF) on αSyn aggregation and neuron survival. PFF-seeded pathology did not lead to DA neuron loss even with the highest dose of PFFs. The combination of PFFs and 6-OHDA did not trigger additional neurodegeneration or LB-like pathology and instead presented DA neuron loss to a similar extent as with 6-OHDA only. CDNF did not affect the PFF-seeded αSyn pathology or the DA neuron survival in the combination model but showed a trend toward neuroprotection in the 6-OHDA-only cultures.

    Topics: alpha-Synuclein; Animals; Dopamine; Feasibility Studies; Mesencephalon; Mice; Nerve Degeneration; Oxidopamine; Parkinson Disease; Synucleinopathies

2023
NME1 Protects Against Neurotoxin-, α-Synuclein- and LRRK2-Induced Neurite Degeneration in Cell Models of Parkinson's Disease.
    Molecular neurobiology, 2022, Volume: 59, Issue:1

    Parkinson's disease (PD) is a neurodegenerative disease characterised by the progressive degeneration of midbrain dopaminergic neurons, coupled with the intracellular accumulation of α-synuclein. Axonal degeneration is a central part of the pathology of PD. While the majority of PD cases are sporadic, some are genetic; the G2019S mutation in leucine-rich repeat kinase 2 (LRRK2) is the most common genetic form. The application of neurotrophic factors to protect dopaminergic neurons is a proposed experimental therapy. One such neurotrophic factor is growth differentiation factor (GDF)5. GDF5 is a dopaminergic neurotrophic factor that has been shown to upregulate the expression of a protein called nucleoside diphosphate kinase A (NME1). However, whether NME1 is neuroprotective in cell models of axonal degeneration of relevance to PD is unknown. Here we show that treatment with NME1 can promote neurite growth in SH-SY5Y cells, and in cultured dopaminergic neurons treated with the neurotoxin 6-hydroxydopamine (6-OHDA). Similar effects of NME1 were found in SH-SY5Y cells and dopaminergic neurons overexpressing human wild-type α-synuclein, and in stable SH-SY5Y cell lines carrying the G2019S LRRK2 mutation. We found that the effects of NME1 require the RORα/ROR2 receptors. Furthermore, increased NF-κB-dependent transcription was partially required for the neurite growth-promoting effects of NME1. Finally, a combined bioinformatics and biochemical analysis of the mitochondrial oxygen consumption rate revealed that NME1 enhanced mitochondrial function, which is known to be impaired in PD. These data show that recombinant NME1 is worthy of further study as a potential therapeutic agent for axonal protection in PD.

    Topics: alpha-Synuclein; Cell Line, Tumor; Dopaminergic Neurons; Humans; Leucine-Rich Repeat Serine-Threonine Protein Kinase-2; Nerve Degeneration; Neurites; Neuronal Outgrowth; Neuroprotective Agents; NM23 Nucleoside Diphosphate Kinases

2022
The mitochondrial protein Sideroflexin 3 (SFXN3) influences neurodegeneration pathways in vivo.
    The FEBS journal, 2022, Volume: 289, Issue:13

    Synapses are a primary pathological target in neurodegenerative diseases. Identifying therapeutic targets at the synapse could delay progression of numerous conditions. The mitochondrial protein SFXN3 is a neuronally enriched protein expressed in synaptic terminals and regulated by key synaptic proteins, including α-synuclein. We first show that SFXN3 uses the carrier import pathway to insert into the inner mitochondrial membrane. Using high-resolution proteomics on Sfxn3-KO mice synapses, we then demonstrate that SFXN3 influences proteins and pathways associated with neurodegeneration and cell death (including CSPα and Caspase-3), as well as neurological conditions (including Parkinson's disease and Alzheimer's disease). Overexpression of SFXN3 orthologues in Drosophila models of Parkinson's disease significantly reduced dopaminergic neuron loss. In contrast, the loss of SFXN3 was insufficient to trigger neurodegeneration in mice, indicating an anti- rather than pro-neurodegeneration role for SFXN3. Taken together, these results suggest a potential role for SFXN3 in the regulation of neurodegeneration pathways.

    Topics: alpha-Synuclein; Animals; Cation Transport Proteins; Mice; Mitochondrial Membranes; Mitochondrial Proteins; Nerve Degeneration; Parkinson Disease; Synapses

2022
Interleukin-6 triggers toxic neuronal iron sequestration in response to pathological α-synuclein.
    Cell reports, 2022, 02-15, Volume: 38, Issue:7

    α-synuclein (α-syn) aggregation and accumulation drive neurodegeneration in Parkinson's disease (PD). The substantia nigra of patients with PD contains excess iron, yet the underlying mechanism accounting for this iron accumulation is unclear. Here, we show that misfolded α-syn activates microglia, which release interleukin 6 (IL-6). IL-6, via its trans-signaling pathway, induces changes in the neuronal iron transcriptome that promote ferrous iron uptake and decrease cellular iron export via a pathway we term the cellular iron sequestration response, or CISR. The brains of patients with PD exhibit molecular signatures of the IL-6-mediated CISR. Genetic deletion of IL-6, or treatment with the iron chelator deferiprone, reduces pathological α-syn toxicity in a mouse model of sporadic PD. These data suggest that IL-6-induced CISR leads to toxic neuronal iron accumulation, contributing to synuclein-induced neurodegeneration.

    Topics: alpha-Synuclein; Animals; Behavior, Animal; Disease Models, Animal; Dopaminergic Neurons; Female; Interleukin-6; Iron; Iron Chelating Agents; Male; Mice, Inbred C57BL; Mice, Transgenic; Nerve Degeneration; Neurons; Parkinson Disease; Signal Transduction; Substantia Nigra

2022
A Novel NOX Inhibitor Treatment Attenuates Parkinson's Disease-Related Pathology in Mouse Models.
    International journal of molecular sciences, 2022, Apr-12, Volume: 23, Issue:8

    Parkinson's disease (PD) is a progressive neurodegenerative motor disorder without an available therapeutic to halt the formation of Lewy bodies for preventing dopaminergic neuronal loss in the nigrostriatal pathway. Since oxidative-stress-mediated damage has been commonly reported as one of the main pathological mechanisms in PD, we assessed the efficacy of a novel NOX inhibitor from AptaBio Therapeutics (C-6) in dopaminergic cells and PD mouse models. The compound reduced the cytotoxicity and enhanced the cell viability at various concentrations against MPP+ and α-synuclein preformed fibrils (PFFs). Further, the levels of ROS and protein aggregation were significantly reduced at the optimal concentration (1 µM). Using two different mouse models, we gavaged C-6 at two different doses to the PD sign-displaying transgenic mice for 2 weeks and stereotaxically PFF-injected mice for 5 weeks. Our results demonstrated that both C-6-treated mouse models showed alleviated motor deficits in pole test, hindlimb clasping, crossbeam, rotarod, grooming, and nesting analyses. We also confirmed that the compound treatment reduced the levels of protein aggregation, along with phosphorylated-α-synuclein, in the striatum and ventral midbrain and further dopaminergic neuronal loss. Taken together, our results strongly suggest that NOX inhibition can be a potential therapeutic target for PD.

    Topics: alpha-Synuclein; Animals; Disease Models, Animal; Dopamine; Dopaminergic Neurons; Mice; Mice, Transgenic; Nerve Degeneration; Parkinson Disease; Protein Aggregates

2022
Alpha-Synuclein: The Spark That Flames Dopaminergic Neurons, In Vitro and In Vivo Evidence.
    International journal of molecular sciences, 2022, Aug-30, Volume: 23, Issue:17

    Mitochondria, α-syn fibrils and the endo-lysosomal system are key players in the pathophysiology of Parkinson's disease. The toxicity of α-syn is amplified by cell-to-cell transmission and aggregation of endogenous species in newly invaded neurons. Toxicity of α-syn PFF was investigated using primary cultures of dopaminergic neurons or on aged mice after infusion in the SNpc and combined with mild inhibition of GBA. In primary dopaminergic neurons, application of α-syn PFF induced a progressive cytotoxicity associated with mitochondrial dysfunction, oxidative stress, and accumulation of lysosomes suggesting that exogenous α-syn reached the lysosome (from the endosome). Counteracting the α-syn endocytosis with a clathrin inhibitor, dopaminergic neuron degeneration was prevented. In vivo, α-syn PFF induced progressive neurodegeneration of dopaminergic neurons associated with motor deficits. Histology revealed progressive aggregation of α-syn and microglial activation and accounted for the seeding role of α-syn, injection of which acted as a spark suggesting a triggering of cell-to-cell toxicity. We showed for the first time that a localized SNpc α-syn administration combined with a slight lysosomal deficiency and aging triggered a progressive lesion. The cellular and animal models described could help in the understanding of the human disease and might contribute to the development of new therapies.

    Topics: alpha-Synuclein; Animals; Dopaminergic Neurons; Humans; Lysosomes; Mice; Nerve Degeneration; Parkinson Disease

2022
Infections of
    Frontiers in cellular and infection microbiology, 2022, Volume: 12

    Cryptococcosis in the central nervous system (CNS) can present with motor declines described as Parkinsonism. Although several lines of evidence indicate that dopaminergic (DA) neuron degeneration and α-synuclein accumulation contribute to the hallmark of Parkinsonism and Parkinson's disease (PD), little is known about cryptococcal infections associated with neuronal degeneration. In this study, the effects of

    Topics: alpha-Synuclein; Animals; Animals, Genetically Modified; Caenorhabditis elegans; Cryptococcus; Disease Models, Animal; Dopamine; Dopaminergic Neurons; Nerve Degeneration

2022
Granulovacuolar degeneration bodies are independently induced by tau and α-synuclein pathology.
    Alzheimer's research & therapy, 2022, 12-14, Volume: 14, Issue:1

    Granulovacuolar degeneration bodies (GVBs) are intracellular vesicular structures that commonly accompany pathological tau accumulations in neurons of patients with tauopathies. Recently, we developed the first model for GVBs in primary neurons, that requires exogenous tau seeds to elicit tau aggregation. This model allowed the identification of GVBs as proteolytically active lysosomes induced by tau pathology. GVBs selectively accumulate cargo in a dense core, that shows differential and inconsistent immunopositivity for (phosphorylated) tau epitopes. Despite the strong evidence connecting GVBs to tau pathology, these structures have been reported in neurons without apparent pathology in brain tissue of tauopathy patients. Additionally, GVBs and putative GVBs have also been reported in the brain of patients with non-tau proteinopathies. Here, we investigated the connection between pathological protein assemblies and GVBs in more detail.. This study combined newly developed primary neuron models for tau and α-synuclein pathology with observations in human brain tissue from tauopathy and Parkinson's disease patients. Immunolabeling and imaging techniques were employed for extensive characterisation of pathological proteins and GVBs. Quantitative data were obtained by high-content automated microscopy as well as single-cell analysis of confocal images.. Employing a novel seed-independent neuronal tau/GVB model, we show that in the context of tauopathy, GVBs are inseparably associated with the presence of cytosolic pathological tau and that intracellular tau aggregation precedes GVB formation, strengthening the causal relationship between pathological accumulation of tau and GVBs. We also report that GVBs are inseparably associated with pathological tau at the single-cell level in the hippocampus of tauopathy patients. Paradoxically, we demonstrate the presence of GVBs in the substantia nigra of Parkinson's disease patients and in a primary neuron model for α-synuclein pathology. GVBs in this newly developed α-synuclein/GVB model are induced in the absence of cytosolic pathological tau accumulations. GVBs in the context of tau or α-synuclein pathology showed similar immunoreactivity for different phosphorylated tau epitopes. The phosphorylated tau immunoreactivity signature of GVBs is therefore independent of the presence of cytosolic tau pathology.. Our data identify the emergence of GVBs as a more generalised response to cytosolic protein pathology.

    Topics: alpha-Synuclein; Epitopes; Humans; Nerve Degeneration; Parkinson Disease; tau Proteins; Tauopathies

2022
Toll-like receptor 4 deficiency facilitates α-synuclein propagation and neurodegeneration in a mouse model of prodromal Parkinson's disease.
    Parkinsonism & related disorders, 2021, Volume: 91

    The evidence linking innate immunity mechanisms and neurodegenerative diseases is growing, but the specific mechanisms are incompletely understood. Experimental data suggest that microglial TLR4 mediates the uptake and clearance of α-synuclein also termed synucleinophagy. The accumulation of misfolded α-synuclein throughout the brain is central to Parkinson's disease (PD). The distribution and progression of the pathology is often attributed to the propagation of α-synuclein. Here, we apply a classical α-synuclein propagation model of prodromal PD in wild type and TLR4 deficient mice to study the role of TLR4 in the progression of the disease. Our data suggest that TLR4 deficiency facilitates the α-synuclein seed spreading associated with reduced lysosomal activity of microglia. Three months after seed inoculation, more pronounced proteinase K-resistant α-synuclein inclusion pathology is observed in mice with TLR4 deficiency. The facilitated propagation of α-synuclein is associated with early loss of dopamine transporter (DAT) signal in the striatum and loss of dopaminergic neurons in substantia nigra pars compacta of TLR4 deficient mice. These new results support TLR4 signaling as a putative target for disease modification to slow the progression of PD and related disorders.

    Topics: alpha-Synuclein; Animals; Brain; Corpus Striatum; Disease Models, Animal; Dopamine Plasma Membrane Transport Proteins; Dopaminergic Neurons; Lysosomes; Mice; Microglia; Nerve Degeneration; Parkinson Disease; Pars Compacta; Signal Transduction; Toll-Like Receptor 4

2021
Peiminine Reduces ARTS-Mediated Degradation of XIAP by Modulating the PINK1/Parkin Pathway to Ameliorate 6-Hydroxydopamine Toxicity and α-Synuclein Accumulation in Parkinson's Disease Models In Vivo and In Vitro.
    International journal of molecular sciences, 2021, Sep-23, Volume: 22, Issue:19

    Topics: alpha-Synuclein; Animals; Animals, Genetically Modified; Apoptosis Regulatory Proteins; Caenorhabditis elegans; Caenorhabditis elegans Proteins; Cevanes; Dopamine; Dopaminergic Neurons; Nerve Degeneration; Parkinson Disease; Proteasome Endopeptidase Complex; Protein Serine-Threonine Kinases; Substantia Nigra; Ubiquitin; Ubiquitin-Protein Ligases; X-Linked Inhibitor of Apoptosis Protein

2021
CD4
    Science (New York, N.Y.), 2021, Nov-12, Volume: 374, Issue:6569

    Recent studies indicate that the adaptive immune system plays a role in Lewy body dementia (LBD). However, the mechanism regulating T cell brain homing in LBD is unknown. Here, we observed T cells adjacent to Lewy bodies and dopaminergic neurons in postmortem LBD brains. Single-cell RNA sequencing of cerebrospinal fluid (CSF) identified up-regulated expression of

    Topics: alpha-Synuclein; Animals; Brain; CD4-Positive T-Lymphocytes; Cerebrospinal Fluid; Chemokine CXCL12; Female; Humans; Lewy Body Disease; Lymphocyte Activation; Male; Meninges; Mice; Mice, Inbred C57BL; Nerve Degeneration; Receptors, CXCR4; Signal Transduction; T-Lymphocyte Subsets; Th17 Cells; Up-Regulation

2021
Microglia-specific overexpression of α-synuclein leads to severe dopaminergic neurodegeneration by phagocytic exhaustion and oxidative toxicity.
    Nature communications, 2021, 10-29, Volume: 12, Issue:1

    Recent findings in human samples and animal models support the involvement of inflammation in the development of Parkinson's disease. Nevertheless, it is currently unknown whether microglial activation constitutes a primary event in neurodegeneration. We generated a new mouse model by lentiviral-mediated selective α-synuclein (αSYN) accumulation in microglial cells. Surprisingly, these mice developed progressive degeneration of dopaminergic (DA) neurons without endogenous αSYN aggregation. Transcriptomics and functional assessment revealed that αSYN-accumulating microglial cells developed a strong reactive state with phagocytic exhaustion and excessive production of oxidative and proinflammatory molecules. This inflammatory state created a molecular feed-forward vicious cycle between microglia and IFNγ-secreting immune cells infiltrating the brain parenchyma. Pharmacological inhibition of oxidative and nitrosative molecule production was sufficient to attenuate neurodegeneration. These results suggest that αSYN accumulation in microglia induces selective DA neuronal degeneration by promoting phagocytic exhaustion, an excessively toxic environment and the selective recruitment of peripheral immune cells.

    Topics: Adaptive Immunity; alpha-Synuclein; Animals; CX3C Chemokine Receptor 1; Dopaminergic Neurons; Encephalitis; Gene Expression; Immunity, Innate; Male; Mice, Inbred C57BL; Mice, Transgenic; Microglia; Nerve Degeneration; Nitric Oxide; Parkinson Disease; Phagocytosis; Reactive Oxygen Species; Substantia Nigra

2021
Peripheral inflammation exacerbates α-synuclein toxicity and neuropathology in Parkinson's models.
    Neuropathology and applied neurobiology, 2021, Volume: 47, Issue:1

    Parkinson's disease and related disorders are devastating neurodegenerative pathologies. Since α-synuclein was identified as a main component of Lewy bodies and neurites, efforts have been made to clarify the pathogenic mechanisms of α-synuclein's detrimental effects. α-synuclein oligomers are the most harmful species and may recruit and activate glial cells. Inflammation is emerging as a bridge between genetic susceptibility and environmental factors co-fostering Parkinson's disease. However, direct evidence linking inflammation to the harmful activities of α-synuclein oligomers or to the Parkinson's disease behavioural phenotype is lacking.. To clarify whether neuroinflammation influences Parkinson's disease pathogenesis, we developed: (i) a 'double-hit' approach in C57BL/6 naive mice where peripherally administered lipopolysaccharides were followed by intracerebroventricular injection of an inactive oligomer dose; (ii) a transgenic 'double-hit' model where lipopolysaccharides were given to A53T α-synuclein transgenic Parkinson's disease mice.. Lipopolysaccharides induced a long-lasting neuroinflammatory response which facilitated the detrimental cognitive activities of oligomers. LPS-activated microglia and astrocytes responded differently to the oligomers with microglia activating further and acquiring a pro-inflammatory M1 phenotype, while astrocytes atrophied. In the transgenic 'double-hit' A53T mouse model, lipopolysaccharides aggravated cognitive deficits and increased microgliosis. Again, astrocytes responded differently to the double challenge. These findings indicate that peripherally induced neuroinflammation potentiates the α-synuclein oligomer's actions and aggravates cognitive deficits in A53T mice.. The fine management of both peripheral and central inflammation may offer a promising therapeutic approach to prevent or slow down some behavioural aspects in α-synucleinopathies.

    Topics: alpha-Synuclein; Animals; Astrocytes; Disease Models, Animal; Inflammation; Mice, Inbred C57BL; Mice, Transgenic; Microglia; Nerve Degeneration; Nervous System Diseases; Parkinson Disease; Substantia Nigra

2021
A New Synuclein-Transgenic Mouse Model for Early Parkinson's Reveals Molecular Features of Preclinical Disease.
    Molecular neurobiology, 2021, Volume: 58, Issue:2

    Understanding Parkinson's disease (PD), in particular in its earliest phases, is important for diagnosis and treatment. However, human brain samples are collected post-mortem, reflecting mainly end-stage disease. Because brain samples of mouse models can be collected at any stage of the disease process, they are useful in investigating PD progression. Here, we compare ventral midbrain transcriptomics profiles from α-synuclein transgenic mice with a progressive, early PD-like striatal neurodegeneration across different ages using pathway, gene set, and network analysis methods. Our study uncovers statistically significant altered genes across ages and between genotypes with known, suspected, or unknown function in PD pathogenesis and key pathways associated with disease progression. Among those are genotype-dependent alterations associated with synaptic plasticity and neurotransmission, as well as mitochondria-related genes and dysregulation of lipid metabolism. Age-dependent changes were among others observed in neuronal and synaptic activity, calcium homeostasis, and membrane receptor signaling pathways, many of which linked to G-protein coupled receptors. Most importantly, most changes occurred before neurodegeneration was detected in this model, which points to a sequence of gene expression events that may be relevant for disease initiation and progression. It is tempting to speculate that molecular changes similar to those changes observed in our model happen in midbrain dopaminergic neurons before they start to degenerate. In other words, we believe we have uncovered molecular changes that accompany the progression from preclinical to early PD.

    Topics: Aging; alpha-Synuclein; Animals; Corpus Striatum; Disease Models, Animal; Female; Gene Expression Profiling; Gene Expression Regulation, Developmental; Gene Ontology; Gene Regulatory Networks; Genotype; Humans; Mice, Transgenic; Nerve Degeneration; Parkinson Disease; Substantia Nigra; Transgenes

2021
NLRP3 Inflammasome Inhibition Prevents α-Synuclein Pathology by Relieving Autophagy Dysfunction in Chronic MPTP-Treated NLRP3 Knockout Mice.
    Molecular neurobiology, 2021, Volume: 58, Issue:4

    Recent researches showed that nucleotide-binding domain and leucine-rich repeat protein 3 (NLRP3) inflammasome inhibition exerted dopaminergic neuroprotection in cellular or animal models of Parkinson's disease (PD). NLRP3 inflammasome has been proposed as a drug target for treatment of PD. However, the interplay between chronic NLRP3 inflammasome and progressive α-synuclein pathology keeps poorly understood. Moreover, the potential mechanism keeps unknown. In the present study, we investigate whether NLRP3 inflammasome inhibition prevents α-synuclein pathology by relieving autophagy dysfunction in the chronic 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP) mouse model of PD. NLRP3 knockout mice and their wild-type counterparts were treated with continuous MPTP administration via osmotic mini-pumps. Dopaminergic neuronal degeneration was assessed by western blotting and immunohistochemistry (IHC). The levels of dopamine and its metabolites were determined using high-performance liquid chromatography. NLRP3 inflammasome activation and autophagy biomarkers were assessed by western blot. The expressions of pro-inflammatory cytokines were measured by ELISA. The glial reaction and α-synuclein pathology were assessed by IHC and immunofluorescence. Our results show that NLRP3 inflammasome inhibition via NLRP3 knockout not only protects against nigral dopaminergic degeneration and striatal dopamine deletion but also prevents nigral pathological α-synuclein formation in PD mice. Furthermore, it significantly suppresses MPTP-induced glial reaction accompanied by the secretion of pro-inflammatory cytokines in the midbrain of mice. Most importantly, it relieves autophagy dysfunction in the midbrain of PD mice. Collectively, we demonstrate for the first time that improving autophagy function is involved in the preventive effect of NLRP3 inflammasome inhibition on α-synuclein pathology in PD.

    Topics: 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine; alpha-Synuclein; Animals; Autophagy; Corpus Striatum; Dopaminergic Neurons; Inflammasomes; Inflammation; Male; Mice, Inbred C57BL; Mice, Knockout; Nerve Degeneration; NLR Family, Pyrin Domain-Containing 3 Protein; Protein Aggregates; Substantia Nigra

2021
CD4 T cells mediate brain inflammation and neurodegeneration in a mouse model of Parkinson's disease.
    Brain : a journal of neurology, 2021, 08-17, Volume: 144, Issue:7

    α-Synuclein, a key pathological component of Parkinson's disease, has been implicated in the activation of the innate and adaptive immune system. This immune activation includes microgliosis, increased inflammatory cytokines, and the infiltration of T cells into the CNS. More recently, peripherally circulating CD4 and CD8 T cells derived from individuals with Parkinson's disease have been shown to produce Th1/Th2 cytokines in response to α-synuclein, suggesting there may be a chronic memory T cell response present in Parkinson's disease. To understand the potential effects of these α-syn associated T cell responses we used an α-synuclein overexpression mouse model, T cell-deficient mice, and a combination of immunohistochemistry and flow cytometry. In this study, we found that α-synuclein overexpression in the midbrain of mice leads to the upregulation of the major histocompatibility complex II (MHCII) protein on CNS myeloid cells as well as the infiltration of IFNγ producing CD4 and CD8 T cells into the CNS. Interestingly, genetic deletion of TCRβ or CD4, as well as the use of the immunosuppressive drug fingolimod, were able to reduce the CNS myeloid MHCII response to α-synuclein. Furthermore, we observed that CD4-deficient mice were protected from the dopaminergic cell loss observed due to α-syn overexpression. These results suggest that T cell responses associated with α-synuclein pathology may be damaging to key areas of the CNS in Parkinson's disease and that targeting these T cell responses could be an avenue for disease modifying treatments.

    Topics: alpha-Synuclein; Animals; CD4-Positive T-Lymphocytes; Disease Models, Animal; Encephalitis; Female; Male; Mice; Mice, Inbred C57BL; Nerve Degeneration; Parkinson Disease

2021
Quinacrine and Niclosamide Promote Neurite Growth in Midbrain Dopaminergic Neurons Through the Canonical BMP-Smad Pathway and Protect Against Neurotoxin and α-Synuclein-Induced Neurodegeneration.
    Molecular neurobiology, 2021, Volume: 58, Issue:7

    Parkinson's disease is a neurodegenerative disorder characterised by nigrostriatal dopaminergic degeneration, and intracellular α-synuclein aggregation. Current pharmacological treatments are solely symptomatic so there is a need to identify agents that can slow or stop dopaminergic degeneration. One proposed class of therapeutics are neurotrophic factors which promote the survival of nigrostriatal dopaminergic neurons. However, neurotrophic factors need to be delivered directly to the brain. An alternative approach may be to identify pharmacological agents which can reach the brain to stimulate neurotrophic factor expression and/or their signalling pathways in dopaminergic neurons. BMP2 is a neurotrophic factor that is expressed in the human substantia nigra; exogenous BMP2 administration protects against dopaminergic degeneration in in vitro models of PD. In this study, we investigated the neurotrophic potential of two FDA-approved drugs, quinacrine and niclosamide, that are modulators of BMP2 signalling. We report that quinacrine and niclosamide, like BMP2, significantly increased neurite length, as a readout of neurotrophic action, in SH-SY5Y cells and dopaminergic neurons in primary cultures of rat ventral mesencephalon. We also show that these effects of quinacrine and niclosamide require the activation of BMP-Smad signalling. Finally, we demonstrate that quinacrine and niclosamide are neuroprotective against degeneration induced by the neurotoxins, MPP

    Topics: alpha-Synuclein; Animals; Bone Morphogenetic Protein 2; Bone Morphogenetic Proteins; Cell Line, Tumor; Cells, Cultured; Dopaminergic Neurons; Dose-Response Relationship, Drug; Humans; Mesencephalon; Nerve Degeneration; Neurites; Neuroprotection; Neurotoxins; Niclosamide; Quinacrine; Rats; Smad Proteins

2021
α-Synuclein A53T Binds to Transcriptional Adapter 2-Alpha and Blocks Histone H3 Acetylation.
    International journal of molecular sciences, 2021, May-20, Volume: 22, Issue:10

    α-Synuclein (α-syn) is a hallmark amyloidogenic protein component of Lewy bodies in dopaminergic neurons affected by Parkinson's disease (PD). Despite the multi-faceted gene regulation of α-syn in the nucleus, the mechanism underlying α-syn crosstalk in chromatin remodeling in PD pathogenesis remains elusive. Here, we identified transcriptional adapter 2-alpha (TADA2a) as a novel binding partner of α-syn using the BioID system. TADA2a is a component of the p300/CBP-associated factor and is related to histone H3/H4 acetylation. We found that α-syn A53T was more preferentially localized in the nucleus than the α-syn wild-type (WT), leading to a stronger disturbance of TADA2a. Indeed, α-syn A53T significantly reduced the level of histone H3 acetylation in SH-SY5Y cells; its reduction was also evident in the striatum (STR) and substantia nigra (SN) of mice that were stereotaxically injected with α-syn preformed fibrils (PFFs). Interestingly, α-syn PFF injection resulted in a decrease in TADA2a in the STR and SN of α-syn PFF-injected mice. Furthermore, the levels of TADA2a and acetylated histone H3 were significantly decreased in the SN of patients with PD. Therefore, histone modification through α-syn A53T-TADA2a interaction may be associated with α-syn-mediated neurotoxicity in PD pathology.

    Topics: Acetylation; alpha-Synuclein; Animals; Cell Line, Tumor; Corpus Striatum; Disease Models, Animal; DNA-Binding Proteins; Dopaminergic Neurons; Histones; Humans; Lewy Bodies; Male; Mice; Mice, Inbred C57BL; Nerve Degeneration; Parkinson Disease; Substantia Nigra; Transcription Factors

2021
Diterpene glycosides from Holothuria scabra exert the α-synuclein degradation and neuroprotection against α-synuclein-Mediated neurodegeneration in C. elegans model.
    Journal of ethnopharmacology, 2021, Oct-28, Volume: 279

    Holothuria (Metriatyla) scabra Jaeger (H. scabra), sea cucumber, is the marine organism that has been used as traditional food and medicine to gain the health benefits since ancient time. Although our recent studies have shown that crude extracts from H. scabra exhibited neuroprotective effects against Parkinson's disease (PD), the underlying mechanisms and bioactive compounds are still unknown.. In the present study, we examined the efficacy of purified compounds from H. scabra and their underlying mechanism on α-synuclein degradation and neuroprotection against α-synuclein-mediated neurodegeneration in a transgenic Caenorhabditis elegans PD model.. The H. scabra compounds (HSEA-P1 and P2) were purified and examined for their toxicity and optimal dose-range by food-clearance and lifespan assays. The α-synuclein degradation and neuroprotection against α-synuclein-mediated neurodegeneration were determined using transgenic C. elegans model, Punc-54::α-syn and Pdat-1:: α-syn; Pdat-1::GFP, respectively, and then further investigated by determining the behavioral assays including locomotion rate, basal slowing rate, ethanol avoidance, and area-restricted searching. The underlying mechanisms related to autophagy were clarified by quantitative PCR and RNAi experiments.. Our results showed that HSEA-P1 and HSEA-P2 significantly diminished α-synuclein accumulation, improved motility deficits, and recovered the shortened lifespan. Moreover, HSEA-P1 and HSEA-P2 significantly protected dopaminergic neurons from α-synuclein toxicity and alleviated dopamine-associated behavioral deficits, i.e., basal slowing, ethanol avoidance, and area-restricted searching. HSEA-P1 and HSEA-P2 also up-regulated autophagy-related genes, including beclin-1/bec-1, lc-3/lgg-1, and atg-7/atg-7. RNA interference (RNAi) of these genes in transgenic α-synuclein worms confirmed that lc-3/lgg-1 and atg-7/atg-7 were required for α-synuclein degradation and DAergic neuroprotection activities of HSEA-P1 and HSEA-P2. NMR and mass spectrometry analysis revealed that the HSEA-P1 and HSEA-P2 contained diterpene glycosides.. These findings indicate that diterpene glycosides extracted from H. scabra decreases α-synuclein accumulation and protects α-synuclein-mediated DAergic neuronal loss and its toxicities via lgg-1 and atg-7.

    Topics: alpha-Synuclein; Animals; Animals, Genetically Modified; Autophagy; Behavior, Animal; Caenorhabditis elegans; Diterpenes; Dopaminergic Neurons; Glycosides; Holothuria; Locomotion; Nerve Degeneration; Neuroprotective Agents; Parkinsonian Disorders

2021
Mechanistic basis for receptor-mediated pathological α-synuclein fibril cell-to-cell transmission in Parkinson's disease.
    Proceedings of the National Academy of Sciences of the United States of America, 2021, 06-29, Volume: 118, Issue:26

    The spread of pathological α-synuclein (α-syn) is a crucial event in the progression of Parkinson's disease (PD). Cell surface receptors such as lymphocyte activation gene 3 (LAG3) and amyloid precursor-like protein 1 (APLP1) can preferentially bind α-syn in the amyloid over monomeric state to initiate cell-to-cell transmission. However, the molecular mechanism underlying this selective binding is unknown. Here, we perform an array of biophysical experiments and reveal that LAG3 D1 and APLP1 E1 domains commonly use an alkaline surface to bind the acidic C terminus, especially residues 118 to 140, of α-syn. The formation of amyloid fibrils not only can disrupt the intramolecular interactions between the C terminus and the amyloid-forming core of α-syn but can also condense the C terminus on fibril surface, which remarkably increase the binding affinity of α-syn to the receptors. Based on this mechanism, we find that phosphorylation at serine 129 (pS129), a hallmark modification of pathological α-syn, can further enhance the interaction between α-syn fibrils and the receptors. This finding is further confirmed by the higher efficiency of pS129 fibrils in cellular internalization, seeding, and inducing PD-like α-syn pathology in transgenic mice. Our work illuminates the mechanistic understanding on the spread of pathological α-syn and provides structural information for therapeutic targeting on the interaction of α-syn fibrils and receptors as a potential treatment for PD.

    Topics: alpha-Synuclein; Amyloid; Amyloid beta-Protein Precursor; Animals; Antigens, CD; Cell Line, Tumor; Endocytosis; Humans; Lymphocyte Activation Gene 3 Protein; Mice; Nerve Degeneration; Neurons; Parkinson Disease; Phosphorylation; Phosphoserine; Protein Binding; Static Electricity

2021
Neurorescue Effects of Frondoside A and Ginsenoside Rg3 in
    Molecules (Basel, Switzerland), 2021, Aug-10, Volume: 26, Issue:16

    Parkinson's disease (PD) is a currently incurable neurodegenerative disorder characterized by the loss of dopaminergic (DAergic) neurons in the substantia nigra pars compacta and α-synuclein aggregation. Accumulated evidence indicates that the saponins, especially from ginseng, have neuroprotective effects against neurodegenerative disorders. Interestingly, saponin can also be found in marine organisms such as the sea cucumber, but little is known about its effect in neurodegenerative disease, including PD. In this study, we investigated the anti-Parkinson effects of frondoside A (FA) from

    Topics: alpha-Synuclein; Animals; Animals, Genetically Modified; Apoptosis; Caenorhabditis elegans; Caenorhabditis elegans Proteins; Disease Models, Animal; Dopaminergic Neurons; Gene Expression Regulation; Ginsenosides; Glycosides; Longevity; Nerve Degeneration; Oxidopamine; Parkinson Disease; Proteolysis; Triterpenes

2021
Alpha-synuclein oligomerization and dopaminergic degeneration occur synchronously in the brain and colon of MPTP-intoxicated parkinsonian monkeys.
    Neuroscience letters, 2020, 01-18, Volume: 716

    Dopaminergic (DAergic) degeneration and abnormal α-synuclein (α-syn) expression, phosphorylation and aggregation are observed in both the nigrostriatal system (NSS) and enteric nervous system (ENS) of patients with Parkinson's disease (PD). Whether these alterations in α-syn and DAergic neurons occur synchronously in the two nervous systems or follow a process that spreads from the gut to the brain remains a subject of debate. Here, in MPTP-intoxicated cynomolgus monkeys, we showed a parallel DAergic degeneration in the colon as well as in the substantia nigra and striatum (SN/STR), as indicated by reduced expression of tyrosine hydroxylase (TH) and dopamine transporter (DAT). In addition, we observed a simultaneous increase in the concentrations of total, phosphorylated, and oligomeric α-syn in the colon and SN/STR. Moreover, we identified that the above changes in α-syn were associated with an increase in the expression of polo-like kinase 2 (PLK2), an enzyme that promotes α-syn phosphorylation, and a decrease in the activity of protein phosphatase 2A (PP2A), an enzyme that facilitates α-syn dephosphorylation. Because the colonic ENS can be readily analyzed using routine biopsies, the shared pathological features between the colonic ENS and the brain NSS found in this study provide useful information for assessing and understanding the neuropathology in PD patients using colonic biopsies.

    Topics: alpha-Synuclein; Animals; Brain; Dopaminergic Neurons; Enteric Nervous System; Macaca fascicularis; Male; Nerve Degeneration; Parkinsonian Disorders

2020
High-salt diet does not boost neuroinflammation and neurodegeneration in a model of α-synucleinopathy.
    Journal of neuroinflammation, 2020, Jan-24, Volume: 17, Issue:1

    Pre-clinical studies in models of multiple sclerosis and other inflammatory disorders suggest that high-salt diet may induce activation of the immune system and potentiate inflammation. However, high-salt diet constitutes a common non-pharmacological intervention to treat autonomic problems in synucleinopathies such as Parkinson's disease and multiple system atrophy. Since neuroinflammation plays an important pathogenic role in these neurodegenerative disorders, we asked here whether high-salt diet may aggravate the disease phenotype in a transgenic model of multiple system atrophy.. Nine-month-old PLP-hαSyn and matched wildtype mice received normal or high-salt diet for a period of 3 months. Behavioral, histological, and molecular analyses were performed to evaluate the effect of high-salt diet on motor decline, neuroinflammation, neurodegeneration, and α-synuclein accumulation in these mice.. Brain subregion-specific molecular and histological analyses showed no deleterious effects of high-salt diet on the level of microglial activation. Moreover, neuroinflammation-related cytokines and chemokines, T cell recruitment or astrogliosis were unaffected by high-salt diet exposure. Behavioral testing showed no effect of diet on motor decline. High-salt diet was not related to the deterioration of neurodegeneration or α-synuclein accumulation in PLP-hαSyn mice.. Here, we demonstrate that high-salt diet does not aggravate neuroinflammation and neurodegeneration in PLP-hαSyn mice. Our findings discard a deleterious pro-neuroinflammatory effect of high-salt diet in multiple system atrophy.

    Topics: alpha-Synuclein; Animals; Brain; Disease Models, Animal; Humans; Inflammation; Mice; Mice, Inbred C57BL; Mice, Transgenic; Microglia; Multiple System Atrophy; Nerve Degeneration; Neurons; Sodium Chloride, Dietary

2020
Alpha-Synuclein deficiency ameliorates chronic methamphetamine induced neurodegeneration in mice.
    Toxicology, 2020, 05-30, Volume: 438

    The α-Synuclein (α-syn) and tau have synergistic effects on neurodegenerative diseases induced by environmental factors or genetic mutation. Thus, we investigated the role of α-syn and tau in neurodegeneration induced by chronic methamphetamine (METH) exposure (1.0∼20.0 mg/kg/d body weight, for 14 consecutive days). Here, we present a mice model with evidences of α-syn and tau participating in toxicology in chronic METH. METH increased α-syn level in the stratum oriens, pyramidal layer, stratum radiatum and stratum moleculare of hippocampal CA1, CA2 and CA3, polymorph layer of hippocampal dentate gyrus (DG), and substantia nigra (SN). The subcellular locations of the upregulated α-syn were mainly found in mitochondria and axons. The METH upregulated α-syn may directly induce mitochondrial damage, myelin sheath destruction, and synaptic failure. Also, the excess α-syn might indirectly promote tau phosphorylation through tau kinase GSK3β and CDK5, leading to microtubule depolymerization and eventually fusion deficit of autophagosome and lysosome. In the in vitro experiment, the autophagic vacuoles failed to fuse with the lysosome. The neuropathology induced by both the direct and indirect effects of α-syn could be alleviated by α-syn knockout. Taking together, these results indicate that the α-syn mediates the neurodegenerative process induced by chronic METH and that reducing α-syn might be a potential approach to protect the toxic effects of METH and also be, to a broader view, of therapeutic value in neurodegenerative diseases.

    Topics: alpha-Synuclein; Animals; Autophagy; Axons; Cells, Cultured; Cyclin-Dependent Kinase 5; Disease Models, Animal; Glycogen Synthase Kinase 3 beta; Hippocampus; Lysosomes; Male; Methamphetamine; Mice, Inbred C57BL; Mice, Knockout; Mitochondria; Nerve Degeneration; Neurons; Neurotoxicity Syndromes; Phosphorylation; Substantia Nigra; tau Proteins; Time Factors

2020
Unilateral intranigral administration of β-sitosterol β-D-glucoside triggers pathological α-synuclein spreading and bilateral nigrostriatal dopaminergic neurodegeneration in the rat.
    Acta neuropathologica communications, 2020, 04-22, Volume: 8, Issue:1

    The spreading and accumulation of α-synuclein and dopaminergic neurodegeneration, two hallmarks of Parkinson's disease (PD), have been faithfully reproduced in rodent brains by chronic, oral administration of β-sitosterol β-D-glucoside (BSSG). We investigated whether a single injection of BSSG (6 μg BSSG/μL DMSO) in the left substantia nigra of Wistar rats causes the same effects. Mock DMSO injections and untreated rats formed control groups. We performed immunostainings against the pathological α-synuclein, the dopaminergic marker tyrosine hydroxylase (TH), the neuroskeleton marker β-III tubulin, the neurotensin receptor type 1 (NTSR1) as non-dopaminergic phenotype marker and Fluro-Jade C (F-J C) label for neurodegeneration. Using β-galactosidase (β-Gal) assay and active caspase-3 immunostaining, we assessed cell death mechanisms. Golgi-Cox staining was used to measure the density and types of dendritic spines of striatal medium spiny neurons. Motor and non-motor alterations were also evaluated. The study period comprised 15 to 120 days after the lesion. In the injured substantia nigra, BSSG caused a progressive α-synuclein aggregation and dopaminergic neurodegeneration caused by senescence and apoptosis. The α-synuclein immunoreactivity was also present within microglia cells. Decreased density of dopaminergic fibers and dendritic spines also occurred in the striatum. Remarkably, all the histopathological changes also appeared on the contralateral nigrostriatal system, and α-synuclein aggregates were present in other brain regions. Motor and non-motor behavioral alterations were progressive. Our data show that the stereotaxic BSSG administration reproduces PD α-synucleinopathy phenotype in the rat. This approach will aid in identifying the spread mechanism of α-synuclein pathology and validate anti-synucleinopathy therapies.

    Topics: alpha-Synuclein; Animals; Disease Models, Animal; Dopaminergic Neurons; Injections, Intraventricular; Nerve Degeneration; Parkinson Disease; Rats; Rats, Wistar; Sitosterols; Substantia Nigra

2020
Impact of α-synuclein pathology on transplanted hESC-derived dopaminergic neurons in a humanized α-synuclein rat model of PD.
    Proceedings of the National Academy of Sciences of the United States of America, 2020, 06-30, Volume: 117, Issue:26

    Preclinical assessment of the therapeutic potential of dopamine (DA) neuron replacement in Parkinson's disease (PD) has primarily been performed in the 6-hydroxydopamine toxin model. While this is a good model to assess graft function, it does not reflect the pathological features or progressive nature of the disease. In this study, we establish a humanized transplantation model of PD that better recapitulates the main disease features, obtained by coinjection of preformed human α-synuclein (α-syn) fibrils and adeno-associated virus (AAV) expressing human wild-type α-syn unilaterally into the rat substantia nigra (SN). This model gives rise to DA neuron dysfunction and progressive loss of DA neurons from the SN and terminals in the striatum, accompanied by extensive α-syn pathology and a prominent inflammatory response, making it an interesting and relevant model in which to examine long-term function and integrity of transplanted neurons in a PD-like brain. We transplanted DA neurons derived from human embryonic stem cells (hESCs) into the striatum and assessed their survival, growth, and function over 6 to 18 wk. We show that the transplanted cells, even in the presence of ongoing pathology, are capable of innervating the DA-depleted striatum. However, on closer examination of the grafts, we found evidence of α-syn pathology in the form of inclusions of phosphorylated α-syn in a small fraction of the grafted DA neurons, indicating host-to-graft transfer of α-syn pathology, a phenomenon that has previously been observed in PD patients receiving fetal tissue grafts but has not been possible to demonstrate and study in toxin-based animal models.

    Topics: alpha-Synuclein; Animals; Cell Survival; Dopaminergic Neurons; Down-Regulation; Embryonic Stem Cells; Female; Humans; Inflammation; Nerve Degeneration; Rats; Rats, Sprague-Dawley; Stem Cell Transplantation; Substantia Nigra; Synucleinopathies

2020
Minocycline inhibition of microglial rescues nigrostriatal dopaminergic neurodegeneration caused by mutant alpha-synuclein overexpression.
    Aging, 2020, 07-24, Volume: 12, Issue:14

    Studies indicate that mutant α-synuclein (mαSyn) is involved in the pathogenesis of Parkinson's disease (PD). The mαSyn expression leads to the loss of dopaminergic neurons in the substantia nigra (SN) and consequent motor dysfunctions. Additionally, studies found that PD was accompanied by extensive neuroinflammation of SN. However, it remains unclear as to whether microglia participate in the mαSyn pathology. This issue is addressed by using AAV-mα-Syn (A30P-A53T) to overexpress the human mαSyn in the SN in view of establishing the PD model. Subsequently, minocycline (Mino) was used to inhibit microglia activity, and an interleukin-1 receptor (IL-1R1) antagonist was used to hinder the IL-1R1 function. Finally, immunohistochemistry was used to analyze phosphorylated αSyn (Ser129) and TH-positive cells in the SN. Dopamine levels were analyzed by high performance liquid chromatography. mαSyn overexpression in the SN induced motor dysfunction, decreased striatal dopamine levels, and increased pathological αSyn 12 weeks after AAV injection. The data demonstrated that inhibiting microglial activation or hindering IL-1R1 reversed the persistent motor deficits, neurodegeneration of the nigrostriatal dopaminergic system, and development of Lewy body pathology caused by human mαSyn overexpression in the SN. Additionally, these findings indicate that neuroinflammation promotes the loss of neuronal cells.

    Topics: alpha-Synuclein; Animals; Behavior, Animal; Corpus Striatum; Dopamine; Dopaminergic Neurons; Humans; Lewy Bodies; Male; Mice; Mice, Inbred C57BL; Microglia; Minocycline; Motor Activity; Mutation; Nerve Degeneration; Parkinson Disease; Receptors, Interleukin-1 Type I; Substantia Nigra

2020
Tau knockout exacerbates degeneration of parvalbumin-positive neurons in substantia nigra pars reticulata in Parkinson's disease-related α-synuclein A53T mice.
    FASEB journal : official publication of the Federation of American Societies for Experimental Biology, 2020, Volume: 34, Issue:9

    Topics: alpha-Synuclein; Animals; Disks Large Homolog 4 Protein; Homeodomain Proteins; Mice; Mice, Inbred C57BL; Microtubule-Associated Proteins; Mutation; Nerve Degeneration; Parkinson Disease; Parvalbumins; Peptide Fragments; Protein Aggregates; Receptors, N-Methyl-D-Aspartate; Substantia Nigra; tau Proteins; Transcription Factors

2020
Neuronal activity modulates alpha-synuclein aggregation and spreading in organotypic brain slice cultures and in vivo.
    Acta neuropathologica, 2020, Volume: 140, Issue:6

    Topics: alpha-Synuclein; Animals; Axons; Brain; Dopamine; Dopaminergic Neurons; Humans; Lewy Body Disease; Mice; Nerve Degeneration; Neurons; Synucleinopathies

2020
Effects of Grape Polyphenols on the Life Span and Neuroinflammatory Alterations Related to Neurodegenerative Parkinson Disease-Like Disturbances in Mice.
    Molecules (Basel, Switzerland), 2020, Nov-16, Volume: 25, Issue:22

    Functional nutrition is a valuable supplementation to dietary therapy. Functional foods are enriched with biologically active substances. Plant polyphenols attract particular attention due to multiple beneficial properties attributed to their high antioxidant and other biological activities. We assessed the effect of grape polyphenols on the life span of C57BL/6 mice and on behavioral and neuroinflammatory alterations in a transgenic mouse model of Parkinson disease (PD) with overexpression of the A53T-mutant human α-synuclein. C57BL/6 mice were given a dietary supplement containing grape polyphenol concentrate (GPC-1.5 mL/kg/day) with drinking water from the age of 6-8 weeks for life. Transgenic PD mice received GPC beginning at the age of 10 weeks for four months. GPC significantly influenced the cumulative proportion of surviving and substantially augmented the average life span in mice. In the transgenic PD model, the grape polyphenol (GP) diet enhanced memory reconsolidation and diminished memory extinction in a passive avoidance test. Behavioral effects of GP treatment were accompanied by a decrease in α-synuclein accumulation in the frontal cortex and a reduction in the expression of neuroinflammatory markers (IBA1 and CD54) in the frontal cortex and hippocampus. Thus, a GP-rich diet is recommended as promising functional nutrition for aging people and patients with neurodegenerative disorders.

    Topics: alpha-Synuclein; Animals; Behavior, Animal; Brain; Dietary Supplements; Inflammation; Mice, Inbred C57BL; Mutant Proteins; Nerve Degeneration; Parkinson Disease; Polyphenols; Vitis; Weight Gain

2020
Tyrosol, a simple phenol from EVOO, targets multiple pathogenic mechanisms of neurodegeneration in a C. elegans model of Parkinson's disease.
    Neurobiology of aging, 2019, Volume: 82

    Parkinson's disease (PD) is a common neurodegenerative disorder involving α-synuclein (α-syn) aggregation, oxidative stress, dysregulation of redox metal homeostasis, and neurotoxicity. Different phenolic compounds with known antioxidant or antichelating properties have been shown to also interfere with aggregation of amyloid proteins and modulate intracellular signaling pathways. The present study aims to investigate for the first time the effect of tyrosol (TYR), a simple phenol present in extra-virgin olive oil, on α-syn aggregation in a Caenorhabditis elegans model of PD and evaluate its potential to prevent α-syn toxicity, neurodegeneration, and oxidative stress in this model organism. Our results show that TYR is effective in reducing α-syn inclusions, resulting in a lower toxicity and extended life span of treated nematodes. Moreover, TYR delayed α-syn-dependent degeneration of dopaminergic neurons in vivo. TYR treatment also reduced reactive oxygen species level and promoted the expression of specific chaperones and antioxidant enzymes. Overall, our study puts into perspective TYR potential to be considered as nutraceutical that targets pivotal causal factors in PD.

    Topics: alpha-Synuclein; Animals; Animals, Genetically Modified; Antioxidants; Caenorhabditis elegans; Dietary Supplements; Disease Models, Animal; Drug Delivery Systems; Nerve Degeneration; Olive Oil; Parkinsonian Disorders; Phenylethyl Alcohol

2019
Microglia affect α-synuclein cell-to-cell transfer in a mouse model of Parkinson's disease.
    Molecular neurodegeneration, 2019, 08-16, Volume: 14, Issue:1

    Cell-to-cell propagation of α-synuclein (α-syn) aggregates is thought to contribute to the pathogenesis of Parkinson's disease (PD) and underlie the spread of α-syn neuropathology. Increased pro-inflammatory cytokine levels and activated microglia are present in PD and activated microglia can promote α-syn aggregation. However, it is unclear how microglia influence α-syn cell-to-cell transfer.. We developed a clinically relevant mouse model to monitor α-syn prion-like propagation between cells; we transplanted wild-type mouse embryonic midbrain neurons into a mouse striatum overexpressing human α-syn (huα-syn) following adeno-associated viral injection into the substantia nigra. In this system, we depleted or activated microglial cells and determined the effects on the transfer of huα-syn from host nigrostriatal neurons into the implanted dopaminergic neurons, using the presence of huα-syn within the grafted cells as a readout.. First, we compared α-syn cell-to-cell transfer between host mice with a normal number of microglia to mice in which we had pharmacologically ablated 80% of the microglia from the grafted striatum. With fewer host microglia, we observed increased accumulation of huα-syn in grafted dopaminergic neurons. Second, we assessed the transfer of α-syn into grafted neurons in the context of microglia activated by one of two stimuli, lipopolysaccharide (LPS) or interleukin-4 (IL-4). LPS exposure led to a strong activation of microglial cells (as determined by microglia morphology, cytokine production and an upregulation in genes involved in the inflammatory response in the LPS-injected mice by RNA sequencing analysis). LPS-injected mice had significantly higher amounts of huα-syn in grafted neurons. In contrast, injection of IL-4 did not change the proportion of grafted dopamine neurons that contained huα-syn relative to controls. As expected, RNA sequencing analysis on striatal tissue revealed differential gene expression between LPS and IL-4-injected mice; with the genes upregulated in tissue from mice injected with LPS including several of those involved in an inflammatory response.. The absence or the hyperstimulation of microglia affected α-syn transfer in the brain. Our results suggest that under resting, non-inflammatory conditions, microglia modulate the transfer of α-syn. Pharmacological regulation of neuroinflammation could represent a future avenue for limiting the spread of PD neuropathology.

    Topics: alpha-Synuclein; Animals; Brain; Disease Models, Animal; Dopaminergic Neurons; Female; Lipopolysaccharides; Macrophages; Mice, Inbred C57BL; Microglia; Nerve Degeneration; Parkinson Disease

2019
Trichloroethylene and its metabolite TaClo lead to degeneration of substantia nigra dopaminergic neurones: Effects in wild type and human A30P mutant α-synuclein mice.
    Neuroscience letters, 2019, 10-15, Volume: 711

    Parkinson's disease (PD) is characterised pathologically by degeneration of the dopaminergic (DA) neurones of the substantia nigra pars compacta (SNpc) and the presence of α-synuclein containing Lewy body inclusions. Trichloroethylene (TCE) has been suggested as a potential environmental chemical that may contribute to the development of PD, via conversion to the neurotoxin, 1-Trichloromethyl-1,2,3,4-tetrahydro-β-carboline (TaClo). We investigated the effect of an 8 week exposure to TCE or TaClo on wild type and, as an experimental model of PD, A30P mutant α-synuclein overexpressing mice using a combination of behaviour and pathology. TCE or TaClo exposure caused significant DA neuronal loss within the SNpc in both wild type and transgenic mice. Cell numbers were lower in A30P animals than wild type, however, no additive effect of TCE or TaClo exposure and A30P overexpression was found. TCE or TaClo did not appear to lead to acceleration of motor or cognitive deficits in either wild type or A30P mutant mice, potentially because of the modest reductions of DA neuronal number in the SNpc. Our results do however suggest that TCE exposure could be a possible factor in development of PD like changes following exposure.

    Topics: alpha-Synuclein; Animals; Dopaminergic Neurons; Female; Humans; Male; Mice; Mice, Inbred C57BL; Mice, Transgenic; Mutation; Nerve Degeneration; Neurotoxins; Parkinsonian Disorders; Substantia Nigra; Trichloroethylene

2019
Blunting neuroinflammation with resolvin D1 prevents early pathology in a rat model of Parkinson's disease.
    Nature communications, 2019, 09-02, Volume: 10, Issue:1

    Neuroinflammation is one of the hallmarks of Parkinson's disease (PD) and may contribute to midbrain dopamine (DA) neuron degeneration. Recent studies link chronic inflammation with failure to resolve early inflammation, a process operated by specialized pro-resolving mediators, including resolvins. However, the effects of stimulating the resolution of inflammation in PD - to modulate disease progression - still remain unexplored. Here we show that rats overexpressing human α-synuclein (Syn) display altered DA neuron properties, reduced striatal DA outflow and motor deficits prior to nigral degeneration. These early alterations are coupled with microglia activation and perturbations of inflammatory and pro-resolving mediators, namely IFN-γ and resolvin D1 (RvD1). Chronic and early RvD1 administration in Syn rats prevents central and peripheral inflammation, as well as neuronal dysfunction and motor deficits. We also show that endogenous RvD1 is decreased in human patients with early-PD. Our results suggest there is an imbalance between neuroinflammatory and pro-resolving processes in PD.

    Topics: alpha-Synuclein; Animals; Disease Models, Animal; Docosahexaenoic Acids; Dopaminergic Neurons; Humans; Inflammation; Male; Microglia; Nerve Degeneration; Parkinson Disease; Rats; Rats, Sprague-Dawley; Rats, Transgenic; Substantia Nigra

2019
Parkinson's disease-associated
    Proceedings of the National Academy of Sciences of the United States of America, 2019, 10-08, Volume: 116, Issue:41

    Mutations in the

    Topics: alpha-Synuclein; Animals; Animals, Genetically Modified; Brain; Cell Membrane; Dopaminergic Neurons; Drosophila melanogaster; Drosophila Proteins; Endoplasmic Reticulum Stress; Female; Group VI Phospholipases A2; Group X Phospholipases A2; Humans; Male; Mitochondria; Mitochondrial Proteins; Nerve Degeneration; Parkinson Disease; Phospholipids; Synaptic Transmission

2019
Histamine-4 receptor antagonist JNJ7777120 inhibits pro-inflammatory microglia and prevents the progression of Parkinson-like pathology and behaviour in a rat model.
    Brain, behavior, and immunity, 2019, Volume: 76

    The activation of microglial cells is presumed to play a key role in the pathogenesis of Parkinson's disease (PD). The activity of microglia is regulated by the histamine-4 receptor (H

    Topics: alpha-Synuclein; Animals; Behavior, Animal; Brain; Corpus Striatum; Disease Models, Animal; Disease Progression; Dopaminergic Neurons; Histamine; Indoles; Inflammation; Male; Microglia; Nerve Degeneration; Parkinson Disease; Parkinsonian Disorders; Piperazines; Rats; Rats, Sprague-Dawley; Receptors, Histamine H4; Rotenone

2019
Anle138b modulates α-synuclein oligomerization and prevents motor decline and neurodegeneration in a mouse model of multiple system atrophy.
    Movement disorders : official journal of the Movement Disorder Society, 2019, Volume: 34, Issue:2

    MSA is a fatal neurodegenerative disease characterized by autonomic failure and severe motor impairment. Its main pathological hallmark is the accumulation of α-synuclein in oligodendrocytes, leading to glial and neuronal dysfunction and neurodegeneration. These features are recapitulated in the PLP-hαSyn mouse model expressing human α-synuclein in oligodendrocytes. At present, there is no effective disease-modifying therapy. Previous experiments have shown that the aggregation inhibitor, anle138b, reduces neurodegeneration and behavioral deficits in mouse models of other proteinopathies.. To test the therapeutic potential of anle138b in a mouse model of MSA.. Two-month-old PLP-hαSyn mice were fed over a period of 4 months with pellets containing anle138b at two different doses (0.6 and 2 g/kg) and compared to healthy controls and PLP-hαSyn mice fed with placebo pellets. At the end of the treatment, behavioral and histological analyses were performed.. We observed a reversal of motor function to healthy control levels when PLP-hαSyn mice were treated with both doses of anle138b. Histological and molecular analyses showed a significant reduction in α-synuclein oligomers and glial cytoplasmic inclusions in animals fed with anle138b compared to nontreated mice. These animals also present preservation of dopaminergic neurons and reduction in microglial activation in SN correlating with the α-synuclein reduction observed.. Anle138b reduces α-synuclein accumulation in PLP-hαSyn mice, leading to neuroprotection, reduction of microglial activation, and preservation of motor function supporting the use of anle138b in a future clinical trial for MSA. © 2018 The Authors. Movement Disorders published by Wiley Periodicals, Inc. on behalf of International Parkinson and Movement Disorder Society.

    Topics: alpha-Synuclein; Animals; Benzodioxoles; Disease Models, Animal; Mice, Transgenic; Movement Disorders; Multiple System Atrophy; Nerve Degeneration; Neuroglia; Neurons; Oligodendroglia; Pyrazoles

2019
Lipidomic Analysis of α-Synuclein Neurotoxicity Identifies Stearoyl CoA Desaturase as a Target for Parkinson Treatment.
    Molecular cell, 2019, 03-07, Volume: 73, Issue:5

    In Parkinson's disease (PD), α-synuclein (αS) pathologically impacts the brain, a highly lipid-rich organ. We investigated how alterations in αS or lipid/fatty acid homeostasis affect each other. Lipidomic profiling of human αS-expressing yeast revealed increases in oleic acid (OA, 18:1), diglycerides, and triglycerides. These findings were recapitulated in rodent and human neuronal models of αS dyshomeostasis (overexpression; patient-derived triplication or E46K mutation; E46K mice). Preventing lipid droplet formation or augmenting OA increased αS yeast toxicity; suppressing the OA-generating enzyme stearoyl-CoA-desaturase (SCD) was protective. Genetic or pharmacological SCD inhibition ameliorated toxicity in αS-overexpressing rat neurons. In a C. elegans model, SCD knockout prevented αS-induced dopaminergic degeneration. Conversely, we observed detrimental effects of OA on αS homeostasis: in human neural cells, excess OA caused αS inclusion formation, which was reversed by SCD inhibition. Thus, monounsaturated fatty acid metabolism is pivotal for αS-induced neurotoxicity, and inhibiting SCD represents a novel PD therapeutic approach.

    Topics: alpha-Synuclein; Animals; Antiparkinson Agents; Caenorhabditis elegans; Cell Line; Cerebral Cortex; Diglycerides; Disease Models, Animal; Dopaminergic Neurons; Drug Discovery; Enzyme Inhibitors; Humans; Induced Pluripotent Stem Cells; Lipid Droplets; Lipid Metabolism; Metabolomics; Mice, Inbred C57BL; Mice, Transgenic; Molecular Targeted Therapy; Nerve Degeneration; Neural Stem Cells; Neurons; Oleic Acid; Parkinson Disease; Rats, Sprague-Dawley; Saccharomyces cerevisiae; Stearoyl-CoA Desaturase; Triglycerides

2019
Anti-Parkinson activity of bioactive substances extracted from Holothuria leucospilota.
    Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie, 2019, Volume: 109

    Parkinson's disease (PD) is a well-known neurodegenerative disorder characterized by dopaminergic (DA) neuron loss and α-synuclein aggregation. Recent study revealed that the extracts from sea cucumber, Holothuroidea spp., exhibited neuroprotective and lifespan extension effects in Caenorhabditis elegans model. Interestingly, the black sea cucumber, Holothuria leucospilota, possesses body wall and a specialized organ called cuvierian tubules containing high amount of bioactive compounds. In this study, the neuroprotective effects of the body wall (BW) and cuvierian tubules (CT) from this sea cucumber against PD were evaluated using C. elegans as a model. H. leucospilota were extracted using ethanol (ET), ethyl acetate (EA), butanol (BU) and aqueous (AQ) fractions. Extracts from these fractions were used to treat the 6-OHDA-induced BZ555 and α-synuclein expressing NL5901 strains of C. elegans. Treatment with ET, EA, BU and AQ fractions of H. leucospilota extracts could significantly prevent degeneration of DA neurons in 6-OHDA-induced worms, improve food-sensing behavior mediated by DA neurons, and up-regulate cat-2 and sod-3 gene expressions. These results indicate the neuroprotective activity of the extracts which may be attributed to the anti-oxidant activity of the bioactive compounds. Moreover, α-synuclein aggregation was significantly reduced together with the recovery of lipid deposition upon the treatment with H. leucospilota extracts. In addition, treatment with H. leucospilota extracts was able to increase the lifespan of 6-OHDA-induced N2. NMR analysis revealed the major chemical components in the effective EA fractions were terpenoids, steroids, saponins, and glycosides. In summary, H. leucospilota extracts exhibited anti-Parkinson effect in both toxin-induced and transgenic C. elegans models of PD. Further study will be performed to elucidate the most effective anti-PD molecules which will lead to the development of anti-PD drug.

    Topics: alpha-Synuclein; Animals; Animals, Genetically Modified; Antiparkinson Agents; Disease Models, Animal; Dopamine; Dopaminergic Neurons; Holothuria; Nerve Degeneration; Neuroprotective Agents; Parkinson Disease

2019
Long-term RNAi knockdown of α-synuclein in the adult rat substantia nigra without neurodegeneration.
    Neurobiology of disease, 2019, Volume: 125

    α-Synuclein plays a central role in the pathogenesis of Parkinson's disease (PD); interventions that decrease its expression appear neuroprotective in PD models. Successful translation of these observations into effective therapies will be dependent on the safety of suppressing α-synuclein expression in the adult brain. We investigated long-term α-synuclein knockdown in the adult rat CNS. 8-month old animals received either AAV-sh[Snca] (an RNA interference vector targeting the Snca mRNA transcript) or AAV-sh[Ctrl] (a control vector) unilaterally into the substantia nigra. No signs of systemic toxicity or motor dysfunction were observed in either experimental group over 12 months. Viral transgene expression persisted to 12 months post-inoculation, at which point Snca mRNA expression in substantia nigra dopaminergic neurons of animals that received AAV-sh[Snca] was decreased by ≈90%, and α-synuclein immunoreactivity by >70% relative to the control side. Stereological quantification of Nissl-labeled neurons showed no evidence of neurodegeneration in the substantia nigra 12 months after inoculation with either vector, and we observed abundant dopaminergic neurons with minimal α-synuclein immunoreactivity that appeared otherwise unremarkable in the AAV-sh[Snca] group. Despite the absence of neurodegeneration, some loss of TH expression was evident in nigral neurons after transduction with either vector, presumably a non-specific consequence of vector delivery, cellular transduction, or expression of shRNA or GFP. We conclude that long-term α-synuclein knockdown in the substantia nigra does not cause significant functional deficits in the ascending dopaminergic projection, or neurodegeneration. These findings are encouraging that it may be feasible to target α-synuclein expression therapeutically in PD.

    Topics: alpha-Synuclein; Animals; Dependovirus; Gene Knockdown Techniques; Genetic Vectors; Male; Nerve Degeneration; Rats; Rats, Inbred Lew; RNA Interference; RNA, Small Interfering; RNAi Therapeutics; Substantia Nigra

2019
Spermine protects alpha-synuclein expressing dopaminergic neurons from manganese-induced degeneration.
    Cell biology and toxicology, 2019, Volume: 35, Issue:2

    Manganese exposure is among the many environmental risk factors linked to the progression of neurodegenerative diseases, such as manganese-induced parkinsonism. In animal models, chronic exposure to manganese causes loss of cell viability, neurodegeneration, and functional deficits. Polyamines, such as spermine, have been shown to rescue animals from age-induced neurodegeneration in an autophagy-dependent manner; nonetheless, it is not understood whether polyamines can prevent manganese-induced toxicity. In this study, we used two model systems, the Caenorhabditis elegans UA44 strain and SK-MEL-28 cells, both expressing the protein alpha-synuclein (α-syn) to determine whether spermine could ameliorate manganese-induced toxicity. Manganese caused a substantial reduction in the viability of SK-MEL-28 cells and hastened neurodegeneration in the UA44 strain. Spermine protected both the SK-MEL-28 cells and the UA44 strain from manganese-induced toxicity. Spermine also reduced the age-associated neurodegeneration observed in the UA44 strain compared with a control strain without α-syn expression and led to improved avoidance behavior in a functional assay. Treatment with berenil, an inhibitor of polyamine catabolism, which leads to increased intracellular polyamine levels, also showed similar cellular protection against manganese toxicity. While both translation blocker cycloheximide and autophagy blocker chloroquine caused a reduction in the cytoprotective effect of spermine, transcription blocker actinomycin D had no effect. This study provides new insights on the effect of spermine in preventing manganese-induced toxicity, which is most likely via translational regulation of several candidate genes, including those of autophagy. Thus, our results indicate that polyamines positively influence neuronal health, even when exposed to high levels of manganese and α-syn, and supplementing polyamines through diet might delay the onset of diseases involving degeneration of dopaminergic neurons.

    Topics: alpha-Synuclein; Animals; Caenorhabditis elegans; Cell Line; Cell Survival; Diminazene; Dopaminergic Neurons; Humans; Manganese; Nerve Degeneration; Neuroprotective Agents; Spermine

2019
l-3,4-dihydroxyphenylalanine (l-DOPA) modulates brain iron, dopaminergic neurodegeneration and motor dysfunction in iron overload and mutant alpha-synuclein mouse models of Parkinson's disease.
    Journal of neurochemistry, 2019, Volume: 150, Issue:1

    Treatment with the dopamine (DA) precursor l-3,4-dihydroxyphenylalanine (l-DOPA) provides symptomatic relief arising from DA denervation in Parkinson's disease. Mounting evidence that DA autooxidation to neurotoxic quinones is involved in Parkinson's disease pathogenesis has raised concern about potentiation of oxidative stress by l-DOPA. The rate of DA quinone formation increases in the presence of excess redox-active iron (Fe), which is a pathological hallmark of Parkinson's disease. Conversely, l-DOPA has pH-dependent Fe-chelating properties, and may act to 'redox silence' Fe and partially allay DA autoxidation. We examined the effects of l-DOPA in three murine models of parkinsonian neurodegeneration: early-life Fe overexposure in wild-type mice, transgenic human (h)A53T mutant α-synuclein (α-syn) over-expression, and a combined 'multi-hit' model of Fe-overload in hA53T mice. We found that l-DOPA was neuroprotective and prevented age-related Fe accumulation in the substantia nigra pars compacta (SNc), similar to the mild-affinity Fe chelator clioquinol. Chronic l-DOPA treatment showed no evidence of increased oxidative stress in wild-type midbrain and normalized motor performance, when excess Fe was present. Similarly, l-DOPA also did not exacerbate protein oxidation levels in hA53T mice, with or without excess nigral Fe, and showed evidence of neuroprotection. The effects of l-DOPA in Fe-fed hA53T mice were somewhat muted, suggesting that Fe-chelation alone is insufficient to attenuate neuron loss in an animal model also recapitulating altered DA metabolism. In summary, we found no evidence in any of our model systems that l-DOPA treatment accentuated neurodegeneration, suggesting DA replacement therapy does not contribute to oxidative stress in the Parkinson's disease brain.

    Topics: alpha-Synuclein; Animals; Antiparkinson Agents; Brain; Disease Models, Animal; Dopaminergic Neurons; Humans; Iron; Iron Overload; Levodopa; Mice; Mice, Transgenic; Nerve Degeneration; Oxidative Stress; Parkinson Disease

2019
Cardiolipin remodeling by ALCAT1 links mitochondrial dysfunction to Parkinson's diseases.
    Aging cell, 2019, Volume: 18, Issue:3

    Cardiolipin (CL) is a mitochondrial signature phospholipid that is required for membrane structure, respiration, dynamics, and mitophagy. Oxidative damage of CL by reactive oxygen species is implicated in the pathogenesis of Parkinson's disease (PD), but the underlying cause remains elusive. This work investigated the role of ALCAT1, an acyltransferase that catalyzes pathological remodeling of CL in various aging-related diseases, in a mouse model of PD induced by 1-methyl-4-phenyl-1,2,4,6-tetrahydropyridine (MPTP). We show that MPTP treatment caused oxidative stress, mtDNA mutations, and mitochondrial dysfunction in the midbrain. In contrast, ablation of the ALCAT1 gene or pharmacological inhibition of ALCAT1 prevented MPTP-induced neurotoxicity, apoptosis, and motor deficits. ALCAT1 deficiency also mitigated mitochondrial dysfunction by modulating DRP1 translocation to the mitochondria. Moreover, pharmacological inhibition of ALCAT1 significantly improved mitophagy by promoting the recruitment of Parkin to dysfunctional mitochondria. Finally, ALCAT1 expression was upregulated by MPTP and by α-synucleinopathy, a key hallmark of PD, whereas ALCAT1 deficiency prevented α-synuclein oligomerization and S-129 phosphorylation, implicating a key role of ALCAT1 in the etiology of mouse models of PD. Together, these findings identify ALCAT1 as a novel drug target for the treatment of PD.

    Topics: Acyltransferases; alpha-Synuclein; Animals; Apoptosis; Cardiolipins; Cell Line, Tumor; Disease Models, Animal; Humans; Locomotion; Mice; Mice, Transgenic; Mitochondria; Mitochondrial Dynamics; Mitophagy; MPTP Poisoning; Nerve Degeneration; Phosphorylation; Reactive Oxygen Species; Ubiquitin-Protein Ligases

2019
Polyphenolic flavonoid (Myricetin) upregulated proteasomal degradation mechanisms: Eliminates neurodegenerative proteins aggregation.
    Journal of cellular physiology, 2019, Volume: 234, Issue:11

    Major neurodegenerative disorders are characterized by the formation of misfolded proteins aggregates inside or outside the neuronal cells. Previous studies suggest that aberrant proteins aggregates play a critical role in protein homeostasis imbalance and failure of protein quality control (PQC) mechanism, leading to disease conditions. However, we still do not understand the precise mechanisms of PQC failure and cellular dysfunctions associated with neurodegenerative diseases caused by the accumulation of protein aggregates. Here, we show that Myricetin, a flavonoid, can eliminate various abnormal proteins from the cellular environment via modulating endogenous levels of Hsp70 chaperone and quality control (QC)-E3 ubiquitin ligase E6-AP. We have observed that Myricetin treatment suppresses the aggregation of different aberrant proteins. Myricetin also enhances the elimination of various toxic neurodegenerative diseases associated proteins from the cells, which could be reversed by the addition of putative proteasome inhibitor (MG132). Remarkably, Myricetin can also stabilize E6-AP and reduce the misfolded proteins inclusions, which further alleviates cytotoxicity. Taken together these findings suggested that new mechanistic and therapeutic insights based on small molecules mediated regulation of disturbed protein quality control mechanism, which may result in the maintenance of the state of proteostasis.

    Topics: A549 Cells; alpha-Synuclein; Enzyme Stability; Flavonoids; Humans; Inclusion Bodies; Luciferases; Models, Biological; Mutant Proteins; Nerve Degeneration; Peptides; Polyphenols; Proteasome Endopeptidase Complex; Protein Aggregates; Protein Folding; Proteolysis; Solubility; Stress, Physiological; Superoxide Dismutase; Ubiquitin-Protein Ligases; Up-Regulation

2019
Layer-specific axonal degeneration of serotonergic fibers in the prefrontal cortex of aged A53T α-synuclein-expressing mice.
    Neurobiology of aging, 2019, Volume: 80

    Axonal pathology precedes dopaminergic cell loss in Parkinson's disease (PD), indicating a dying back axonopathy of nigrostriatal projections. Although most attention focused on the dopaminergic system, increasing evidence implies a compromised serotonergic system in PD as well. By combining immunohistological and biochemical approaches, a profound layer-specific reduction of the serotonergic input to the prefrontal cortex (PFC) layers II and V/VI in aged mutant A53T α-synuclein-expressing mice (A53T mice) was detected. In addition, the altered fiber network was characterized by swollen axons and enlarged axonal varicosities within all PFC layers, but most pronounced in PFC layer I. Although prefrontal serotonin levels and synaptic protein expression were preserved, aged A53T mice showed increased levels of kinesin family member 1a and vesicular monoamine transporter 2. Together with increased tryptophan hydroxylase 2 mRNA levels in the raphe nuclei and an elevated serotonin receptor 1b expression in the PFC, these findings point to compensatory mechanisms within the serotonergic system to overcome the reduced neuritic input to the PFC in this transgenic animal model for PD.

    Topics: Aging; alpha-Synuclein; Animals; Axons; Disease Models, Animal; Mice; Nerve Degeneration; Parkinson Disease; Prefrontal Cortex; Serotonergic Neurons

2019
Lycopodium Attenuates Loss of Dopaminergic Neurons by Suppressing Oxidative Stress and Neuroinflammation in a Rat Model of Parkinson's Disease.
    Molecules (Basel, Switzerland), 2019, Jun-10, Volume: 24, Issue:11

    Parkinson's disease, a chronic, age related neurodegenerative disorder, is characterized by a progressive loss of nigrostriatal dopaminergic neurons. Several studies have proven that the activation of glial cells, presence of alpha-synuclein aggregates, and oxidative stress, fuels neurodegeneration, and currently there is no definitive treatment for PD. In this study, a rotenone-induced rat model of PD was used to understand the neuroprotective potential of Lycopodium (Lyc), a commonly-used potent herbal medicine. Immunohistochemcial data showed that rotenone injections significantly increased the loss of dopaminergic neurons in the substantia nigra, and decreased the striatal expression of tyrosine hydroxylase. Further, rotenone administration activated microglia and astroglia, which in turn upregulated the expression of α-synuclein, pro-inflammatory, and oxidative stress factors, resulting in PD pathology. However, rotenone-injected rats that were orally treated with lycopodium (50 mg/kg) were protected against dopaminergic neuronal loss by diminishing the expression of matrix metalloproteinase-3 (MMP-3) and MMP-9, as well as reduced activation of microglia and astrocytes. This neuroprotective mechanism not only involves reduction in pro-inflammatory response and α-synuclein expression, but also synergistically enhanced antioxidant defense system by virtue of the drug's multimodal action. These findings suggest that Lyc has the potential to be further developed as a therapeutic candidate for PD.

    Topics: alpha-Synuclein; Animals; Antioxidants; Brain; Catalase; Cyclooxygenase 2; Cytokines; Disease Models, Animal; Dopaminergic Neurons; Glutathione; Inflammation; Inflammation Mediators; Lipid Peroxidation; Lycopodium; Male; Malondialdehyde; Matrix Metalloproteinases; Microglia; Nerve Degeneration; Neuroprotection; Nitric Oxide; Nitric Oxide Synthase Type II; Nitrites; Oxidative Stress; Parkinson Disease; Plant Extracts; Rats, Wistar; Rotenone; Superoxide Dismutase

2019
Glial α-synuclein promotes neurodegeneration characterized by a distinct transcriptional program in vivo.
    Glia, 2019, Volume: 67, Issue:10

    α-Synucleinopathies are neurodegenerative diseases that are characterized pathologically by α-synuclein inclusions in neurons and glia. The pathologic contribution of glial α-synuclein in these diseases is not well understood. Glial α-synuclein may be of particular importance in multiple system atrophy (MSA), which is defined pathologically by glial cytoplasmic α-synuclein inclusions. We have previously described Drosophila models of neuronal α-synucleinopathy, which recapitulate key features of the human disorders. We have now expanded our model to express human α-synuclein in glia. We demonstrate that expression of α-synuclein in glia alone results in α-synuclein aggregation, death of dopaminergic neurons, impaired locomotor function, and autonomic dysfunction. Furthermore, co-expression of α-synuclein in both neurons and glia worsens these phenotypes as compared to expression of α-synuclein in neurons alone. We identify unique transcriptomic signatures induced by glial as opposed to neuronal α-synuclein. These results suggest that glial α-synuclein may contribute to the burden of pathology in the α-synucleinopathies through a cell type-specific transcriptional program. This new Drosophila model system enables further mechanistic studies dissecting the contribution of glial and neuronal α-synuclein in vivo, potentially shedding light on mechanisms of disease that are especially relevant in MSA but also the α-synucleinopathies more broadly.

    Topics: alpha-Synuclein; Animals; Animals, Genetically Modified; Cell Death; Constipation; Disease Models, Animal; Dopaminergic Neurons; Drosophila; Humans; Movement Disorders; Nerve Degeneration; Neurodegenerative Diseases; Neuroglia; Protein Aggregation, Pathological; Transcription, Genetic; Transcriptome

2019
Alteration of autophagy-related proteins in peripheral blood mononuclear cells of patients with Parkinson's disease.
    Neurobiology of aging, 2018, Volume: 63

    Previous postmortem studies demonstrated dysregulation of autophagy in patients with Parkinson's disease (PD). To clarify whether this alteration reflects a fundamental aspect of PD or represents the final stage of autophagy dysregulation resulting from a long neurodegenerative process, we focused on basal autophagy in peripheral blood mononuclear cells (PBMCs) of PD patients (n = 35) and controls (n = 23). The whole-transcriptome assay revealed downregulation of mRNAs for 6 core regulators of autophagy (UNC-51-like kinase [ULK] 3, autophagy-related [Atg] 2A, Atg4B, Atg5, Atg16L1, and histone deacetylase 6). Reverse transcription-polymerase chain reaction and Western blot analysis confirmed significantly increased protein levels of upstream autophagy (ULK1, Beclin1, and autophagy/beclin1 regulator 1) with negative feedback of mRNA expression for these proteins in PD. These protein levels were correlated with increased levels of α-synuclein in PBMCs. The expression level of the oligomeric form of α-synuclein in PBMCs paralleled the clinical severity of PD and the degeneration of cardiac sympathetic nerves. Basal activity of autophagy can be lower in patients with PD. Alteration of basal autophagy may be a fundamental aspect of PD.

    Topics: Aged; Aged, 80 and over; alpha-Synuclein; Autophagy; Autophagy-Related Proteins; Female; Humans; Leukocytes, Mononuclear; Male; Middle Aged; Nerve Degeneration; Parkinson Disease; RNA, Messenger; Severity of Illness Index

2018
Toxic properties of microsome-associated alpha-synuclein species in mouse primary neurons.
    Neurobiology of disease, 2018, Volume: 111

    α-synuclein (αS) is a small protein that self-aggregates into α-helical oligomer species and subsequently into larger insoluble amyloid fibrils that accumulate in intraneuronal inclusions during the development of Parkinson's disease. Toxicity of αS oligomers and fibrils has been long debated and more recent data are suggesting that both species can induce neurodegeneration. However while most of these data are based on differences in structure between oligomer and aggregates, often preassembled in vitro, the in vivo situation might be more complex and subcellular locations where αS species accumulate, rather than their conformation, might contribute to enhanced toxicity. In line with this observation, we have shown that αS oligomers and aggregates are associated with the endoplasmic reticulum/microsomes (ER/M) membrane in vivo and how accumulation of soluble αS oligomers at the ER/M level precedes neuronal degeneration in a mouse model of α-synucleinopathies. In this paper we took a further step, investigating the biochemical and functional features of αS species associated with the ER/M membrane. We found that by comparison with non-microsomal associated αS (P10), the ER/M-associated αS pool is a unique population of oligomers and aggregates with specific biochemical traits such as increased aggregation, N- and C-terminal truncations and phosphorylation at serine 129. Moreover, when administered to murine primary neurons, ER/M-associated αS species isolated from diseased A53T human αS transgenic mice induced neuronal changes in a time- and dose-dependent manner. In fact the addition of small amounts of ER/M-associated αS species from diseased mice to primary cultures induced the formation of beads-like structures or strings of fibrous αS aggregates along the neurites, occasionally covering the entire process or localizing at the soma level. By comparison treatment with P10 fractions from the same diseased mice resulted in the formation of scarce and small puncta only when administered at high amount. Moreover, increasing the amount of P100/M fractions obtained from diseased and, more surprisingly, from presymptomatic mice induced a significant level of neuronal death that was prevented when neurons were treated with ER/M fractions immunodepleted of αS high molecular weight (HMW) species. These data provide the first evidence of the existence of two different populations of αS HMW species in vivo, putting the spotlight on the association to ER/M membrane

    Topics: alpha-Synuclein; Animals; Apoptosis; Cell Line, Tumor; Cerebral Cortex; Disease Models, Animal; Endoplasmic Reticulum; Humans; Mice, Transgenic; Microsomes; Molecular Weight; Nerve Degeneration; Neurodegenerative Diseases; Neurons; Primary Cell Culture; Protein Aggregation, Pathological

2018
α-synuclein Induces Mitochondrial Dysfunction through Spectrin and the Actin Cytoskeleton.
    Neuron, 2018, 01-03, Volume: 97, Issue:1

    Genetics and neuropathology strongly link α-synuclein aggregation and neurotoxicity to the pathogenesis of Parkinson's disease and related α-synucleinopathies. Here we describe a new Drosophila model of α-synucleinopathy based on widespread expression of wild-type human α-synuclein, which shows robust neurodegeneration, early-onset locomotor deficits, and abundant α-synuclein aggregation. We use results of forward genetic screening and genetic analysis in our new model to demonstrate that α-synuclein expression promotes reorganization of the actin filament network and consequent mitochondrial dysfunction through altered Drp1 localization. Similar changes are present in a mouse α-synucleinopathy model and in postmortem brain tissue from patients with α-synucleinopathy. Importantly, we provide evidence that the interaction of α-synuclein with spectrin initiates pathological alteration of the actin cytoskeleton and downstream neurotoxicity. These findings suggest new therapeutic approaches for α-synuclein induced neurodegeneration.

    Topics: Actin Cytoskeleton; Aged; Aged, 80 and over; alpha-Synuclein; Animals; Cytoskeletal Proteins; Disease Models, Animal; Drosophila melanogaster; Female; GTP-Binding Proteins; Humans; Lewy Body Disease; Male; Middle Aged; Mitochondria; Nerve Degeneration; Spectrin

2018
Neuronal complex I deficiency occurs throughout the Parkinson's disease brain, but is not associated with neurodegeneration or mitochondrial DNA damage.
    Acta neuropathologica, 2018, Volume: 135, Issue:3

    Mitochondrial complex I deficiency occurs in the substantia nigra of individuals with Parkinson's disease. It is generally believed that this phenomenon is caused by accumulating mitochondrial DNA damage in neurons and that it contributes to the process of neurodegeneration. We hypothesized that if these theories are correct, complex I deficiency should extend beyond the substantia nigra to other affected brain regions in Parkinson's disease and correlate tightly with neuronal mitochondrial DNA damage. To test our hypothesis, we employed a combination of semiquantitative immunohistochemical analyses, Western blot and activity measurements, to assess complex I quantity and function in multiple brain regions from an extensively characterized population-based cohort of idiopathic Parkinson's disease (n = 18) and gender and age matched healthy controls (n = 11). Mitochondrial DNA was assessed in single neurons from the same areas by real-time PCR. Immunohistochemistry showed that neuronal complex I deficiency occurs throughout the Parkinson's disease brain, including areas spared by the neurodegenerative process such as the cerebellum. Activity measurements in brain homogenate confirmed a moderate decrease of complex I function, whereas Western blot was less sensitive, detecting only a mild reduction, which did not reach statistical significance at the group level. With the exception of the substantia nigra, neuronal complex I loss showed no correlation with the load of somatic mitochondrial DNA damage. Interestingly, α-synuclein aggregation was less common in complex I deficient neurons in the substantia nigra. We show that neuronal complex I deficiency is a widespread phenomenon in the Parkinson's disease brain which, contrary to mainstream theory, does not follow the anatomical distribution of neurodegeneration and is not associated with the neuronal load of mitochondrial DNA mutation. Our findings suggest that complex I deficiency in Parkinson's disease can occur independently of mitochondrial DNA damage and may not have a pathogenic role in the neurodegenerative process.

    Topics: Aged; Aged, 80 and over; alpha-Synuclein; Brain; DNA Damage; DNA, Mitochondrial; Electron Transport Complex I; Female; Humans; Male; Middle Aged; Mitochondria; Mitochondrial Diseases; Nerve Degeneration; Neurons; Parkinson Disease; Prospective Studies; Protein Aggregation, Pathological

2018
Trichloroethylene and Parkinson's Disease: Risk Assessment.
    Molecular neurobiology, 2018, Volume: 55, Issue:7

    This study was conducted to investigate the mechanism of action and extent of selective dopaminergic neurodegeneration caused by exposure to trichloroethylene (TCE) leading to the endogenous formation of the neurotoxin 1-trichloromethyl-1,2,3,4-tetrahydro-β-carboline (TaClo) in rodents. Beginning at 3 months of age, male C57BL/6 mice received oral TCE dissolved in vehicle for 8 months. Dopaminergic neuronal loss was assessed by nigral tyrosine hydroxylase (TH) immunoreactivity. Selective dopaminergic neurodegeneration was determined based on histological analysis of non-dopaminergic neurons in the brain. Behavioral assays were evaluated using open field activity and rotarod tests. Mitochondrial complex I activity, oxidative stress markers, and microglial activation were also examined in the substantia nigra. The level of TaClo was detected using HPLC-electrospray ionization tandem mass spectrometry. Dopaminergic neurotoxicity of TaClo was determined in midbrain organotypic cultures from rat pups. Following 8 months of TCE treatment, there was a progressive and selective loss of 50% of the dopaminergic neurons in mouse substantia nigra (SN) and about 50% loss of dopamine and 72% loss of 3,4-dihydroxyphenylacetic acid in the striatum, respectively. In addition, motor deficits, mitochondrial impairment, oxidative stress, and inflammation were measured. TaClo content was quantified in the brain after TCE treatment. In organotypic cultures, TaClo rather than TCE induced dopaminergic neuronal loss, similar to MPP

    Topics: Administration, Oral; alpha-Synuclein; Animals; Corpus Striatum; Dopamine; Inflammation; Male; Mice, Inbred C57BL; Mitochondria; Nerve Degeneration; Neurotoxins; Oxidative Stress; Parkinson Disease; Protein Folding; Risk Assessment; Substantia Nigra; Trichloroethylene

2018
α-Synuclein accumulation and GBA deficiency due to L444P GBA mutation contributes to MPTP-induced parkinsonism.
    Molecular neurodegeneration, 2018, 01-08, Volume: 13, Issue:1

    Mutations in glucocerebrosidase (GBA) cause Gaucher disease (GD) and increase the risk of developing Parkinson's disease (PD) and Dementia with Lewy Bodies (DLB). Since both genetic and environmental factors contribute to the pathogenesis of sporadic PD, we investigated the susceptibility of nigrostriatal dopamine (DA) neurons in L444P GBA heterozygous knock-in (GBA. We used GBA. L444P GBA heterozygous mutation reduced GBA protein levels, enzymatic activity and a concomitant accumulation of α-synuclein in the midbrain of GBA. Our results suggest that GBA deficiency due to L444P GBA heterozygous mutation and the accompanying accumulation of α-synuclein render DA neurons more susceptible to MPTP intoxication. Thus, GBA and α-synuclein play dual physiological roles in the survival of DA neurons in response to the mitochondrial dopaminergic neurotoxin, MPTP.

    Topics: alpha-Synuclein; Animals; Dopaminergic Neurons; Gene Knock-In Techniques; Glucosylceramidase; Humans; Mesencephalon; Mice; Mice, Knockout; Mutation; Nerve Degeneration; Parkinsonian Disorders

2018
Nigral depigmentation reflects monoamine exhaustion as initial step to Parkinson's disease.
    Medical hypotheses, 2018, Volume: 110

    This hypothesis discusses exposure and response to various stressors as cause for chronic neurodegeneration. Predisposing genetic and environmental factors in conjunction with exposure to exogenous and endogenous toxins cause stress, which consumes dopamine and related biogenic amines. To compensate monoamine exhaustion, conversion of endogenous levodopa to dopamine by tyrosine hydroxylase is up regulated. Concomitantly, tyrosine mediated levodopa degradation to dopaquinone is reduced. Dopaquinone is the essential precursor of neuromelanin. Its deficiency may cause irreversible nigral fading as initial feature of Parkinson's disease.

    Topics: alpha-Synuclein; Biogenic Monoamines; Humans; Models, Neurological; Monophenol Monooxygenase; Nerve Degeneration; Neurons; Parkinson Disease; Protein Folding; Substantia Nigra

2018
Lewy body-like alpha-synuclein inclusions trigger reactive microgliosis prior to nigral degeneration.
    Journal of neuroinflammation, 2018, May-01, Volume: 15, Issue:1

    Converging evidence suggests a role for microglia-mediated neuroinflammation in Parkinson's disease (PD). Animal models of PD can serve as a platform to investigate the role of neuroinflammation in degeneration in PD. However, due to features of the previously available PD models, interpretations of the role of neuroinflammation as a contributor to or a consequence of neurodegeneration have remained elusive. In the present study, we investigated the temporal relationship of neuroinflammation in a model of synucleinopathy following intrastriatal injection of pre-formed alpha-synuclein fibrils (α-syn PFFS).. Male Fischer 344 rats (N = 114) received unilateral intrastriatal injections of α-syn PFFs, PBS, or rat serum albumin with cohorts euthanized at monthly intervals up to 6 months. Quantification of dopamine neurons, total neurons, phosphorylated α-syn (pS129) aggregates, major histocompatibility complex-II (MHC-II) antigen-presenting microglia, and ionized calcium-binding adaptor molecule-1 (Iba-1) immunoreactive microglial soma size was performed in the substantia nigra. In addition, the cortex and striatum were also examined for the presence of pS129 aggregates and MHC-II antigen-presenting microglia to compare the temporal patterns of pSyn accumulation and reactive microgliosis.. Intrastriatal injection of α-syn PFFs to rats resulted in widespread accumulation of phosphorylated α-syn inclusions in several areas that innervate the striatum followed by significant loss (~ 35%) of substantia nigra pars compacta dopamine neurons within 5-6 months. The peak magnitudes of α-syn inclusion formation, MHC-II expression, and reactive microglial morphology were all observed in the SN 2 months following injection and 3 months prior to nigral dopamine neuron loss. Surprisingly, MHC-II immunoreactivity in α-syn PFF injected rats was relatively limited during the later interval of degeneration. Moreover, we observed a significant correlation between substantia nigra pSyn inclusion load and number of microglia expressing MHC-II. In addition, we observed a similar relationship between α-syn inclusion load and number of microglia expressing MHC-II in cortical regions, but not in the striatum.. Our results demonstrate that increases in microglia displaying a reactive morphology and MHC-II expression occur in the substantia nigra in close association with peak numbers of pSyn inclusions, months prior to nigral dopamine neuron degeneration, and suggest that reactive microglia may contribute to vulnerability of SNc neurons to degeneration. The rat α-syn PFF model provides an opportunity to examine the innate immune response to accumulation of pathological α-syn in the context of normal levels of endogenous α-syn and provides insight into the earliest neuroinflammatory events in PD.

    Topics: alpha-Synuclein; Animals; Injections, Intraventricular; Lewy Bodies; Male; Microglia; Nerve Degeneration; Rats; Rats, Inbred F344; Substantia Nigra

2018
Soluble epoxide hydrolase plays a key role in the pathogenesis of Parkinson's disease.
    Proceedings of the National Academy of Sciences of the United States of America, 2018, 06-19, Volume: 115, Issue:25

    Parkinson's disease (PD) is characterized as a chronic and progressive neurodegenerative disorder, and the deposition of specific protein aggregates of α-synuclein, termed Lewy bodies, is evident in multiple brain regions of PD patients. Although there are several available medications to treat PD symptoms, these medications do not prevent the progression of the disease. Soluble epoxide hydrolase (sEH) plays a key role in inflammation associated with the pathogenesis of PD. Here we found that MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine)-induced neurotoxicity in the mouse striatum was attenuated by subsequent repeated administration of TPPU, a potent sEH inhibitor. Furthermore, deletion of the sEH gene protected against MPTP-induced neurotoxicity, while overexpression of sEH in the striatum significantly enhanced MPTP-induced neurotoxicity. Moreover, the expression of the sEH protein in the striatum from MPTP-treated mice or postmortem brain samples from patients with dementia of Lewy bodies (DLB) was significantly higher compared with control groups. Interestingly, there was a positive correlation between sEH expression and phosphorylation of α-synuclein in the striatum. Oxylipin analysis showed decreased levels of 8,9-epoxy-5Z,11Z,14Z-eicosatrienoic acid in the striatum of MPTP-treated mice, suggesting increased activity of sEH in this region. Interestingly, the expression of sEH mRNA in human PARK2 iPSC-derived neurons was higher than that of healthy control. Treatment with TPPU protected against apoptosis in human PARK2 iPSC-derived dopaminergic neurons. These findings suggest that increased activity of sEH in the striatum plays a key role in the pathogenesis of neurodegenerative disorders such as PD and DLB. Therefore, sEH may represent a promising therapeutic target for α-synuclein-related neurodegenerative disorders.

    Topics: 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine; alpha-Synuclein; Animals; Cell Line; Corpus Striatum; Disease Models, Animal; Dopaminergic Neurons; Epoxide Hydrolases; HEK293 Cells; Humans; Lewy Bodies; Male; Mice; Mice, Inbred C57BL; MPTP Poisoning; Nerve Degeneration; Parkinson Disease; RNA, Messenger

2018
Limited effects of dysfunctional macroautophagy on the accumulation of extracellularly derived α-synuclein in oligodendroglia: implications for MSA pathogenesis.
    BMC neuroscience, 2018, 05-21, Volume: 19, Issue:1

    The progressive neurodegenerative disorder multiple system atrophy (MSA) is characterized by α-synuclein-positive (oligodendro-) glial cytoplasmic inclusions (GCIs). A connection between the abnormal accumulation of α-synuclein in GCIs and disease initiation and progression has been postulated. Mechanisms involved in the formation of GCIs are unclear. Abnormal uptake of α-synuclein from extracellular space, oligodendroglial overexpression of α-synuclein, and/or dysfunctional protein degradation including macroautophagy have all been discussed. In the current study, we investigated whether dysfunctional macroautophagy aggravates accumulation of extracellular α-synuclein in the oligodendroglia.. We show that oligodendroglia uptake monomeric and fibrillar extracellular α-synuclein. Blocking macroautophagy through bafilomycin A1 treatment or genetic knockdown of LC3B does not consistently change the level of incorporated α-synuclein in oligodendroglia exposed to extracellular soluble/monomeric or fibrillar α-synuclein, however leads to higher oxidative stress in combination with fibrillar α-synuclein treatment. Finally, we detected no evidence for GCI-like formation resulting from dysfunctional macroautophagy in oligodendroglia using confocal microscopy.. In summary, isolated dysfunctional macroautophagy is not sufficient to enhance abnormal accumulation of uptaken α-synuclein in vitro, but may lead to increased production of reactive oxygen species in the presence of fibrillar α-synuclein. Multiple complementary pathways are likely to contribute to GCI formation in MSA.

    Topics: alpha-Synuclein; Animals; Autophagy; Brain; Humans; Inclusion Bodies; Mice, Inbred C57BL; Multiple System Atrophy; Nerve Degeneration; Oligodendroglia

2018
The c-Abl inhibitor, Radotinib HCl, is neuroprotective in a preclinical Parkinson's disease mouse model.
    Human molecular genetics, 2018, 07-01, Volume: 27, Issue:13

    Accumulating evidence suggests that the non-receptor tyrosine kinase c-Abl plays an important role in the progression of Parkinson's disease (PD) and c-Abl inhibition could be neuroprotective in PD and related α-synucleinopathies. Nilotinib, a c-Abl inhibitor, has shown improved motor and cognitive symptoms in PD patients. However, issues concerning blood-brain barrier (BBB) penetration, lack of selectivity and safety still remain. Radotinib HCl is a selective Bcr-Abl kinase inhibitor that not only effectively access the brain, but also exhibits greater pharmacokinetic properties and safety profiles compared to Nilotinib and other c-Abl inhibitors. Here, we show the neuroprotective efficacy of Radotinib HCl, a brain penetrant c-Abl inhibitor, in a pre-clinical model of PD. Importantly, in vitro studies demonstrate that the treatment of Radotinib HCl protects the α-synuclein preformed fibrils (PFF)-induced neuronal toxicity, reduces the α-synuclein PFF-induced Lewy bodies (LB)/Lewy neurites (LN)-like pathology and inhibits the α-synuclein PFF-induced c-Abl activation in primary cortical neurons. Furthermore, administration of Radotinib HCl inhibits c-Abl activation and prevents dopaminergic neuron loss, neuroinflammation and behavioral deficits following α-synuclein PFF-induced toxicity in vivo. Taken together, our findings indicate that Radotinib HCl has beneficial neuroprotective effects in PD and provides an evidence that selective and brain permeable c-Abl inhibitors can be potential therapeutic agents for the treatment of PD and related α-synucleinopathies.

    Topics: alpha-Synuclein; Animals; Blood-Brain Barrier; Brain; Disease Models, Animal; Dopamine; Dopaminergic Neurons; Humans; Lewy Bodies; Mice; Nerve Degeneration; Neuroprotective Agents; Parkinson Disease; Proto-Oncogene Proteins c-abl; Pyrimidines; Sesquiterpenes

2018
Alpha-synuclein delays mitophagy and targeting Miro rescues neuron loss in Parkinson's models.
    Acta neuropathologica, 2018, Volume: 136, Issue:4

    Alpha-synuclein is a component of Lewy bodies, the pathological hallmark of Parkinson's disease (PD), and is also mutated in familial PD. Here, by extensively analyzing PD patient brains and neurons, and fly models, we show that alpha-synuclein accumulation results in upregulation of Miro protein levels. Miro is a motor/adaptor on the outer mitochondrial membrane that mediates mitochondrial motility, and is removed from damaged mitochondria to facilitate mitochondrial clearance via mitophagy. PD patient neurons abnormally accumulate Miro on the mitochondrial surface leading to delayed mitophagy. Partial reduction of Miro rescues mitophagy phenotypes and neurodegeneration in human neurons and flies. Upregulation of Miro by alpha-synuclein requires an interaction via the N-terminus of alpha-synuclein. Our results highlight the importance of mitochondria-associated alpha-synuclein in human disease, and present Miro as a novel therapeutic target.

    Topics: alpha-Synuclein; Animals; Behavior, Animal; Brain; Cell Differentiation; Drosophila melanogaster; Drosophila Proteins; Humans; Induced Pluripotent Stem Cells; Mitochondria; Mitophagy; Movement Disorders; Mutation; Nerve Degeneration; Neurons; Parkinson Disease; rho GTP-Binding Proteins; Signal Transduction; Up-Regulation

2018
MANF protects dopamine neurons and locomotion defects from a human α-synuclein induced Parkinson's disease model in C. elegans by regulating ER stress and autophagy pathways.
    Experimental neurology, 2018, Volume: 308

    Many studies have demonstrated that mesencephalic astrocyte-derived neurotrophic factor (MANF) has been shown protective effects on neurotoxin based models of Parkinson's disease (PD). It still remains unclear whether MANF can rescue dopaminergic (DA) neurons in an α-synuclein model. Glial cell line-derived neurotrophic factor (GDNF) and its related neurturin (NRTN) can protect DA neurons in the neurotoxin but not α-synuclein animal models of PD, it failed in the clinical trials. Since α-synuclein model can better mimic the progression of human PD, in our study we overexpressed MANF specifically in DA neurons by using an α-synuclein Caenorhabditis elegans (C. elegans) model. Our results showed MANF alleviated progressive neuronal degeneration and prevented locomotion defects. Indeed, MANF can protect cilia of DA neurons at an early stage, suggested that MANF participated in the whole process of neuronal degeneration. Furthermore, we found MANF facilitated the removal of misfolded α-synuclein proteins and rescued the function of damaged DA neurons. By using RNAi approach, we inhibited ER stress and autophagy related genes and effects of MANF were decreased, which demonstrated ER stress and autophagy pathways were involved in the MANF-mediated neuroprotection. Our study suggests MANF exhibits potential as a neuroprotective agent for PD therapy.

    Topics: alpha-Synuclein; Animals; Animals, Genetically Modified; Autophagy; Caenorhabditis elegans; Disease Models, Animal; Dopaminergic Neurons; Endoplasmic Reticulum Stress; Humans; Locomotion; Nerve Degeneration; Nerve Growth Factors; Neuroprotection; Parkinsonian Disorders

2018
Synapsin III deficiency hampers α-synuclein aggregation, striatal synaptic damage and nigral cell loss in an AAV-based mouse model of Parkinson's disease.
    Acta neuropathologica, 2018, Volume: 136, Issue:4

    Parkinson's disease (PD), the most common neurodegenerative movement disorder, is characterized by the progressive loss of nigral dopamine neurons. The deposition of fibrillary aggregated α-synuclein in Lewy bodies (LB), that is considered to play a causative role in the disease, constitutes another key neuropathological hallmark of PD. We have recently described that synapsin III (Syn III), a synaptic phosphoprotein that regulates dopamine release in cooperation with α-synuclein, is present in the α-synuclein insoluble fibrils composing the LB of patients affected by PD. Moreover, we observed that silencing of Syn III gene could prevent α-synuclein fibrillary aggregation in vitro. This evidence suggests that Syn III might be crucially involved in α-synuclein pathological deposition. To test this hypothesis, we studied whether mice knock-out (ko) for Syn III might be protected from α-synuclein aggregation and nigrostriatal neuron degeneration resulting from the unilateral injection of adeno-associated viral vectors (AAV)-mediating human wild-type (wt) α-synuclein overexpression (AAV-hαsyn). We found that Syn III ko mice injected with AAV-hαsyn did not develop fibrillary insoluble α-synuclein aggregates, showed reduced amount of α-synuclein oligomers detected by in situ proximity ligation assay (PLA) and lower levels of Ser129-phosphorylated α-synuclein. Moreover, the nigrostriatal neurons of Syn III ko mice were protected from both synaptic damage and degeneration triggered by the AAV-hαsyn injection. Our observations indicate that Syn III constitutes a crucial mediator of α-synuclein aggregation and toxicity and identify Syn III as a novel therapeutic target for PD.

    Topics: alpha-Synuclein; Amphetamine; Animals; Central Nervous System Stimulants; Dependovirus; Mice; Mice, Inbred C57BL; Mice, Knockout; Neostriatum; Nerve Degeneration; Neurons; Parkinson Disease; Stereotyped Behavior; Substantia Nigra; Synapses; Synapsins; Vesicle-Associated Membrane Protein 2

2018
Immunotherapy targeting toll-like receptor 2 alleviates neurodegeneration in models of synucleinopathy by modulating α-synuclein transmission and neuroinflammation.
    Molecular neurodegeneration, 2018, 08-09, Volume: 13, Issue:1

    Synucleinopathies of the aging population are an heterogeneous group of neurological disorders that includes Parkinson's disease (PD) and dementia with Lewy bodies (DLB) and are characterized by the progressive accumulation of α-synuclein in neuronal and glial cells. Toll-like receptor 2 (TLR2), a pattern recognition immune receptor, has been implicated in the pathogenesis of synucleinopathies because TLR2 is elevated in the brains of patients with PD and TLR2 is a mediator of the neurotoxic and pro-inflammatory effects of extracellular α-synuclein aggregates. Therefore, blocking TLR2 might alleviate α-synuclein pathological and functional effects. For this purpose, herein, we targeted TLR2 using a functional inhibitory antibody (anti-TLR2).. Two different human α-synuclein overexpressing transgenic mice were used in this study. α-synuclein low expresser mouse (α-syn-tg, under the PDGFβ promoter, D line) was stereotaxically injected with TLR2 overexpressing lentivirus to demonstrate that increment of TLR2 expression triggers neurotoxicity and neuroinflammation. α-synuclein high expresser mouse (α-Syn-tg; under mThy1 promoter, Line 61) was administrated with anti-TLR2 to examine that functional inhibition of TLR2 ameliorates neuropathology and behavioral defect in the synucleinopathy animal model. In vitro α-synuclein transmission live cell monitoring system was used to evaluate the role of TLR2 in α-synuclein cell-to-cell transmission.. We demonstrated that administration of anti-TLR2 alleviated α-synuclein accumulation in neuronal and astroglial cells, neuroinflammation, neurodegeneration, and behavioral deficits in an α-synuclein tg mouse model of PD/DLB. Moreover, in vitro studies with neuronal and astroglial cells showed that the neuroprotective effects of anti-TLR2 antibody were mediated by blocking the neuron-to-neuron and neuron-to-astrocyte α-synuclein transmission which otherwise promotes NFκB dependent pro-inflammatory responses.. This study proposes TLR2 immunotherapy as a novel therapeutic strategy for synucleinopathies of the aging population.

    Topics: alpha-Synuclein; Animals; Brain; Disease Models, Animal; Humans; Immunotherapy; Lewy Body Disease; Mice; Mice, Transgenic; Nerve Degeneration; Parkinsonian Disorders; Toll-Like Receptor 2

2018
Amyloid oligomerization of the Parkinson's disease related protein α-synuclein impacts on its curvature-membrane sensitivity.
    Journal of neurochemistry, 2018, Volume: 147, Issue:4

    The amyloid aggregation of the presynaptic protein α-synuclein (AS) is pathognomonic of Parkinson's disease and other neurodegenerative disorders. Physiologically, AS contributes to synaptic homeostasis by participating in vesicle maintenance, trafficking, and release. Its avidity for highly curved acidic membranes has been related to the distinct chemistry of the N-terminal amphipathic helix adopted upon binding to appropriated lipid interfaces. Pathologically, AS populate a myriad of toxic aggregates ranging from soluble oligomers to insoluble amyloid fibrils. Different gain-of-toxic function mechanisms are linked to prefibrillar oligomers which are considered as the most neurotoxic species. Here, we investigated if amyloid oligomerization could hamper AS function as a membrane curvature sensor. We used fluorescence correlation spectroscopy to quantitatively evaluate the interaction of oligomeric species, produced using a popular method based on lyophilization and rehydration, to lipid vesicles of different curvatures and compositions. We found that AS oligomerization has a profound impact on protein-lipid interaction, altering binding affinity and/or curvature sensitivity depending on membrane composition. Our work provides novel insights into how the formation of prefibrillar intermediate species could contribute to neurodegeneration due to a loss-of-function mechanism. OPEN PRACTICES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. The complete Open Science Disclosure form for this article can be found at the end of the article. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/.

    Topics: alpha-Synuclein; Amyloid beta-Peptides; Cell Membrane; Humans; Lipid Bilayers; Nerve Degeneration; Parkinson Disease; Protein Binding; Protein Conformation; Spectrometry, Fluorescence; Synaptic Vesicles

2018
Motor neuron loss and neuroinflammation in a model of α-synuclein-induced neurodegeneration.
    Neurobiology of disease, 2018, Volume: 120

    Mechanisms underlying α-synuclein (αSyn) mediated neurodegeneration are poorly understood. Intramuscular (IM) injection of αSyn fibrils in human A53T transgenic M83

    Topics: alpha-Synuclein; Animals; Brain; Female; Humans; Inflammation; Male; Mice; Mice, Transgenic; Motor Neurons; Nerve Degeneration; Spinal Cord

2018
Inflammasome inhibition prevents α-synuclein pathology and dopaminergic neurodegeneration in mice.
    Science translational medicine, 2018, 10-31, Volume: 10, Issue:465

    Parkinson's disease (PD) is characterized by a profound loss of dopaminergic neurons in the substantia nigra, accompanied by chronic neuroinflammation, mitochondrial dysfunction, and widespread accumulation of α-synuclein-rich protein aggregates in the form of Lewy bodies. However, the mechanisms linking α-synuclein pathology and dopaminergic neuronal death to chronic microglial neuroinflammation have not been completely elucidated. We show that activation of the microglial NLR family pyrin domain containing 3 (NLRP3) inflammasome is a common pathway triggered by both fibrillar α-synuclein and dopaminergic degeneration in the absence of α-synuclein aggregates. Cleaved caspase-1 and the inflammasome adaptor protein apoptosis-associated speck-like protein containing a C-terminal caspase recruitment domain (ASC) were elevated in the substantia nigra of the brains of patients with PD and in multiple preclinical PD models. NLRP3 activation by fibrillar α-synuclein in mouse microglia resulted in a delayed but robust activation of the NLRP3 inflammasome leading to extracellular interleukin-1β and ASC release in the absence of pyroptosis. Nanomolar doses of a small-molecule NLRP3 inhibitor, MCC950, abolished fibrillar α-synuclein-mediated inflammasome activation in mouse microglial cells and extracellular ASC release. Furthermore, oral administration of MCC950 in multiple rodent PD models inhibited inflammasome activation and effectively mitigated motor deficits, nigrostriatal dopaminergic degeneration, and accumulation of α-synuclein aggregates. These findings suggest that microglial NLRP3 may be a sustained source of neuroinflammation that could drive progressive dopaminergic neuropathology and highlight NLRP3 as a potential target for disease-modifying treatments for PD.

    Topics: Administration, Oral; alpha-Synuclein; Animals; CARD Signaling Adaptor Proteins; Disease Models, Animal; Dopaminergic Neurons; Extracellular Space; Furans; Heterocyclic Compounds, 4 or More Rings; Humans; Indenes; Inflammasomes; Mice; Microglia; Nerve Degeneration; NLR Family, Pyrin Domain-Containing 3 Protein; Parkinson Disease; Protein Aggregates; Pyroptosis; Substantia Nigra; Sulfonamides; Sulfones

2018
Distinct functional roles of Vps41-mediated neuroprotection in Alzheimer's and Parkinson's disease models of neurodegeneration.
    Human molecular genetics, 2018, 12-15, Volume: 27, Issue:24

    Commonalities and, in some cases, pathological overlap between neurodegenerative diseases have led to speculation that targeting of underlying mechanisms might be of potentially shared therapeutic benefit. Alzheimer's disease is characterized by the formation of plaques, composed primarily of the amyloid-β 1-42 (Aβ) peptide in the brain, resulting in neurodegeneration. Previously, we have shown that overexpression of the lysosomal-trafficking protein, human Vps41 (hVps41), is neuroprotective in a transgenic worm model of Parkinson's disease, wherein progressive dopaminergic neurodegeneration is induced by α-synuclein overexpression. Here, we report the results of a systematic comparison of hVps41-mediated neuroprotection between α-synuclein and Aβ in transgenic nematode models of Caenorhabditis elegans. Our results indicate that an ARF-like GTPase gene product, ARL-8, mitigates endocytic Aβ neurodegeneration in a VPS-41-dependent manner, rather than through RAB-7 and AP3 as with α-synuclein. Furthermore, the neuroprotective effect of ARL-8 or hVps41 appears to be dependent on their colocalization and the activity of ARL-8. Additionally, we demonstrate that the LC3 orthologue, LGG-2, plays a critical role in Aβ toxicity with ARL-8. Further analysis of functional effectors of Aβ protein processing via the lysosomal pathway will assist in the elucidation of the underlying mechanism involving VPS-41-mediated neuroprotection. These results reveal functional distinctions in the intracellular management of neurotoxic proteins that serve to better inform the path for development of therapeutic interventions to halt neurodegeneration.

    Topics: ADP-Ribosylation Factors; alpha-Synuclein; Alzheimer Disease; Amyloid beta-Peptides; Animals; Animals, Genetically Modified; Caenorhabditis elegans; Caenorhabditis elegans Proteins; Disease Models, Animal; Dopamine; Gene Expression Regulation; Humans; Membrane Proteins; Microtubule-Associated Proteins; Nerve Degeneration; Neuroprotection; Parkinson Disease; Peptide Fragments; Vesicular Transport Proteins

2018
Holothuria scabra extracts exhibit anti-Parkinson potential in C. elegans: A model for anti-Parkinson testing.
    Nutritional neuroscience, 2018, Volume: 21, Issue:6

    Parkinson's disease (PD) is associated with aggregation of α-synuclein and selective death of dopaminergic (DA) neurons in the substantia nigra, thereby leading to cognitive and motor impairments. Nowadays, the drugs commonly used for PD treatment, such as levodopa, provide only symptomatic relief. Therefore, seeking new drugs against PD, especially from plants and marine organisms, is one of the major research areas to be explored. This study aimed to investigate the anti-Parkinson activity of the extracts from the sea cucumber, Holothuria scabra, by using Caenorhabditis elegans as a model.. H. scabra was solvent-extracted and subdivided into six fractions including whole body-ethyl acetate (WBEA), body wall-ethyl acetate (BWEA), viscera-ethyl acetate (VIEA), whole body-butanol (WBBU), body wall-butanol (BWBU), and viscera-butanol (VIBU). The extracts were tested in C. elegans BZ555 strain expressing the green fluorescent protein (GFP) specifically in the DA neurons and NL5901 strain expressing human α-synuclein in the muscle cells.. WBEA, BWEA, and WBBU fractions of H. scabra extracts at 500 µg/ml significantly attenuated DA neuron-degeneration induced by selective cathecholamine neurotoxin 6-hydroxydopamine (6-OHDA) in the BZ555 strain. Moreover, the extracts also reduced α-synuclein aggregation and restored lipid content in NL5901, as well as improved food-sensing behavior and prolonged lifespan in the 6-OHDA-treated wild-type strain.. The study indicated that the H. scabra extracts have anti-Parkinson potential in the C. elegans model. These findings encourage further investigations on using the H. scabra extract, as well as its active constituent compounds, as a possible preventive and/or therapeutic intervention against PD.

    Topics: alpha-Synuclein; Animals; Antiparkinson Agents; Biological Products; Caenorhabditis elegans; Disease Models, Animal; Dopaminergic Neurons; Holothuria; Nerve Degeneration; Neuroprotective Agents; Oxidopamine; Parkinson Disease

2018
DT-diaphorase Protects Against Autophagy Induced by Aminochrome-Dependent Alpha-Synuclein Oligomers.
    Neurotoxicity research, 2017, Volume: 32, Issue:3

    Alpha-synuclein (SNCA) oligomers have been reported to inhibit autophagy. Aminochrome-induced SNCA oligomers are neurotoxic, but the flavoenzyme DT-diaphorase prevents both their formation and their neurotoxicity. However, the possible protective role of DT-diaphorase against autophagy impairment by aminochrome-induced SNCA oligomers remains unclear. To test this idea, we used the cell line RCSN-3NQ7SNCA, with constitutive expression of a siRNA against DT-diaphorase and overexpression SNCA, and RCSN-3 as control cells. A significant increase in LC3-II expression was observed in RCSN-3 cells treated with 20 μM aminochrome and 10 μM rapamycin followed by a decrease in cell death compared to RCSN-3 cells incubated with 20 μM aminochrome alone. The incubation of RCSN-3NQ7SNCA cells with 20 μM aminochrome and 10 μM rapamycin does not change the expression of LC3-II in comparison with RCSN-3NQ7SNCA cells incubated with 20 μM aminochrome alone. The incubation of both cell lines preincubated with 100 nM bafilomycin and 20 μM aminochrome increases the level of LC3-II. Under the same conditions, cell death increases in both cell lines in comparison with cells incubated with 20 μM aminochrome. These results support the protective role of DT-diaphorase against SNCA oligomers-induced autophagy inhibition.

    Topics: alpha-Synuclein; Animals; Autophagy; Cell Line; Cell Survival; Gene Expression; HEK293 Cells; Humans; Indolequinones; Macrolides; Microtubule-Associated Proteins; NAD(P)H Dehydrogenase (Quinone); Nerve Degeneration; Neuroprotection; Rats; RNA, Small Interfering; Sirolimus

2017
Allelic difference in Mhc2ta confers altered microglial activation and susceptibility to α-synuclein-induced dopaminergic neurodegeneration.
    Neurobiology of disease, 2017, Volume: 106

    Parkinson's Disease (PD) is a complex and heterogeneous neurodegenerative disease characterized by a progressive loss of dopaminergic neurons in the substantia nigra pars compacta and pathological intracellular accumulation of alpha-synuclein (α-syn). In the vast majority of PD patients, the disease has a complex etiology, defined by multiple genetic and environmental risk factors. Common genetic variants in the human leukocyte-antigen (HLA) region have been associated to PD risk and the carriage of these can double the risk to develop PD. Among these common genetic variants are the ones that modulate the expression of MHCII genes. MHCII molecules encoded in the HLA-region are responsible for antigen presentation to the adaptive immune system and have a key role in inflammatory processes. In addition to cis‑variants affecting MHCII expression, a transactivator encoded by the Mhc2ta gene is the major regulator of MHCII expression. We have previously identified variations in the promoter region of Mhc2ta, encoded in the VRA4 region, to regulate MHCII expression in rats. The expression of MHCII is known to be required in the response to α-syn. However, how the expression of MHCII affects the activation of microglial or the impact of physiological, differential Mhc2ta expression on degeneration of dopaminergic neurons has not previously been addressed. Here we addressed the implications of common genetic allelic variants of the major regulator of MHCII expression on α-syn-induced microglia activation and the severity of the dopaminergic neurodegeneration. We used a viral vector technology to overexpress α-syn in two rat strains; Dark agouti (DA) wild type and DA.VRA4-congenic rats. The congenic strain carries PVG alleles in the VRA4 locus and therefore displays lower Mhc2ta expression levels compared to DA rats. We analyzed the impact of this physiological differential Mhc2ta expression on gliosis, inflammation, degeneration of the nigro-striatal dopamine system and behavioral deficits after α-syn overexpression. We report that allelic variants of Mhc2ta differently modified the microglial activation in response to overexpression of human α-syn in rats. Overexpression of α-syn led to a larger denervation of the nigro-striatal system and significant behavioral deficits in DA.VRA4 congenic rats with lower Mhc2ta expression compared to DA rats. These results indicate that Mhc2ta is a key upstream regulator of the inflammatory response in PD pathology.

    Topics: Alleles; alpha-Synuclein; Animals; Corpus Striatum; Dependovirus; Dopaminergic Neurons; Genes, MHC Class II; Genetic Predisposition to Disease; Genetic Variation; Genetic Vectors; Humans; Male; Microglia; Motor Activity; Nerve Degeneration; Nuclear Proteins; Parkinsonian Disorders; Rats, Transgenic; Trans-Activators

2017
Decelerated neurodegeneration after intravitreal injection of α-synuclein antibodies in a glaucoma animal model.
    Scientific reports, 2017, 07-24, Volume: 7, Issue:1

    Although elevated intraocular pressure (IOP) remains the major risk factor in glaucoma, neurodegenerative processes continue despite effective IOP lowering. Altered α-synuclein antibody (Abs) levels have been reported to play a crucial role. This study aimed at identifying whether α-synuclein Abs are capable to decelerate neuronal decay while providing insights into proteomic changes. Four groups of Sprague Dawley rats received episcleral vein occlusion: (1) CTRL, no intravitreal injection, n = 6, (2) CTRL IgG, intravitreal injection of unspecific IgG, n = 5, (3) Buffer, intravitreal injection of buffer, n = 6, (4), α-synuclein Ab, intravitreal injection of α-synuclein Ab, n = 5. IOP and retinal nerve fiber layer thickness (RNFLT) were monitored and immunohistochemistry, microarray and proteomic analysis were performed. RNFLT was reduced in CTRL, CTRL IgG and Buffer group (all p < 0.01) and α-synuclein Ab group (p = 0.17). Axon and RGC density showed an increased neurodegeneration in CTRL, CTRL IgG and Buffer group (all p < 0.01) and increased neuronal survival in α-synuclein Ab group (p = 0.38 and 0.06, respectively) compared with fellow eyes. Proteomic analysis revealed alterations of cofilin 1 and superoxide dismutase 1 expression. This data indicate that α-synuclein Ab might indirectly modulate the actin cytoskeleton organization and negatively regulate apoptotic processes via cofilin 1 and superoxide dismutase 1.

    Topics: alpha-Synuclein; Animals; Antibodies, Monoclonal; Apoptosis; Deceleration; Disease Models, Animal; Female; Glaucoma; Intraocular Pressure; Intravitreal Injections; Nerve Degeneration; Neuroprotective Agents; Proteomics; Rats; Rats, Sprague-Dawley; Retina; Retinal Ganglion Cells

2017
Brain monoamine oxidase B and A in human parkinsonian dopamine deficiency disorders.
    Brain : a journal of neurology, 2017, Sep-01, Volume: 140, Issue:9

    See Jellinger (doi:10.1093/awx190) for a scientific commentary on this article. The enzyme monoamine oxidases (B and A subtypes, encoded by MAOB and MAOA, respectively) are drug targets in the treatment of Parkinson's disease. Inhibitors of MAOB are used clinically in Parkinson's disease for symptomatic purposes whereas the potential disease-modifying effect of monoamine oxidase inhibitors is debated. As astroglial cells express high levels of MAOB, the enzyme has been proposed as a brain imaging marker of astrogliosis, a cellular process possibly involved in Parkinson's disease pathogenesis as elevation of MAOB in astrocytes might be harmful. Since brain monoamine oxidase status in Parkinson's disease is uncertain, our objective was to measure, by quantitative immunoblotting in autopsied brain homogenates, protein levels of both monoamine oxidases in three different degenerative parkinsonian disorders: Parkinson's disease (n = 11), multiple system atrophy (n = 11), and progressive supranuclear palsy (n = 16) and in matched controls (n = 16). We hypothesized that if MAOB is 'substantially' localized to astroglial cells, MAOB levels should be generally associated with standard astroglial protein measures (e.g. glial fibrillary acidic protein). MAOB levels were increased in degenerating putamen (+83%) and substantia nigra (+10%, non-significant) in multiple system atrophy; in caudate (+26%), putamen (+27%), frontal cortex (+31%) and substantia nigra (+23%) of progressive supranuclear palsy; and in frontal cortex (+33%), but not in substantia nigra of Parkinson's disease, a region we previously reported no increase in astrocyte protein markers. Although the magnitude of MAOB increase was less than those of standard astrocytic markers, significant positive correlations were observed amongst the astrocyte proteins and MAOB. Despite suggestions that MAOA (versus MAOB) is primarily responsible for metabolism of dopamine in dopamine neurons, there was no loss of the enzyme in the parkinsonian substantia nigra; instead, increased nigral levels of a MAOA fragment and 'turnover' of the enzyme were observed in the conditions. Our findings provide support that MAOB might serve as a biochemical imaging marker, albeit not entirely specific, for astrocyte activation in human brain. The observation that MAOB protein concentration is generally increased in degenerating brain areas in multiple system atrophy (especially putamen) and in progressive supranuclear palsy, but no

    Topics: Adolescent; Adult; alpha-Synuclein; Brain; Case-Control Studies; Caudate Nucleus; Dopamine; Female; Frontal Lobe; Glial Fibrillary Acidic Protein; Humans; Isoenzymes; Male; Middle Aged; Monoamine Oxidase; Multiple System Atrophy; Nerve Degeneration; Parkinson Disease; Peptide Fragments; Phosphopyruvate Hydratase; Putamen; Substantia Nigra; Supranuclear Palsy, Progressive; Tubulin; Young Adult

2017
Age-Dependent Dopaminergic Neurodegeneration and Impairment of the Autophagy-Lysosomal Pathway in LRRK-Deficient Mice.
    Neuron, 2017, Nov-15, Volume: 96, Issue:4

    LRRK2 mutations are the most common genetic cause of Parkinson's disease, but LRRK2's normal physiological role in the brain is unclear. Here, we show that inactivation of LRRK2 and its functional homolog LRRK1 results in earlier mortality and age-dependent, selective neurodegeneration. Loss of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNpc) and of noradrenergic neurons in the locus coeruleus is accompanied with increases in apoptosis, whereas the cerebral cortex and cerebellum are unaffected. Furthermore, selective age-dependent neurodegeneration is only present in LRRK

    Topics: Adrenergic Neurons; Aging; alpha-Synuclein; Animals; Autophagy; Cerebellum; Cerebral Cortex; Dopaminergic Neurons; Female; Leucine-Rich Repeat Serine-Threonine Protein Kinase-2; Locus Coeruleus; Male; Mice; Mice, Knockout; Mutation; Nerve Degeneration; Protein Serine-Threonine Kinases; Signal Transduction; Substantia Nigra; Vacuoles

2017
Lewy Bodies and the Mechanisms of Neuronal Cell Death in Parkinson's Disease and Dementia with Lewy Bodies.
    Brain pathology (Zurich, Switzerland), 2017, Volume: 27, Issue:1

    Neuronal loss in specific brain regions and neurons with intracellular inclusions termed Lewy bodies are the pathologic hallmark in both Parkinson's disease (PD) and dementia with Lewy bodies (DLB). Lewy bodies comprise of aggregated intracellular vesicles and proteins and α-synuclein is reported to be a major protein component. Using human brain tissue from control, PD and DLB and light and confocal immunohistochemistry with antibodies to superoxide dismutase 2 as a marker for mitochondria, α-synuclein for Lewy bodies and βIII Tubulin for microtubules we have examined the relationship between Lewy bodies and mitochondrial loss. We have shown microtubule regression and mitochondrial and nuclear degradation in neurons with developing Lewy bodies. In PD, multiple Lewy bodies were often observed with α-synuclein interacting with DNA to cause marked nuclear degradation. In DLB, the mitochondria are drawn into the Lewy body and the mitochondrial integrity is lost. This work suggests that Lewy bodies are cytotoxic. In DLB, we suggest that microtubule regression and mitochondrial loss results in decreased cellular energy and axonal transport that leads to cell death. In PD, α-synuclein aggregations are associated with intact mitochondria but interacts with and causes nuclear degradation which may be the major cause of cell death.

    Topics: Aged; Aged, 80 and over; alpha-Synuclein; Biomarkers; Cell Death; Cell Nucleus; Electron Transport Complex IV; Female; Humans; Lewy Bodies; Lewy Body Disease; Male; Microtubules; Middle Aged; Mitochondria; Nerve Degeneration; Nerve Tissue Proteins; Neurons; Parkinson Disease; Superoxide Dismutase

2017
Progression of alpha-synuclein pathology in multiple system atrophy of the cerebellar type.
    Neuropathology and applied neurobiology, 2017, Volume: 43, Issue:4

    The aim of this study was to identify early foci of α-synuclein (α-syn pathology) accumulation, subsequent progression and neurodegeneration in multiple system atrophy of the cerebellar type (MSA-C).. We analysed 70-μm-thick sections of 10 cases with MSA-C and 24 normal controls.. MSA-C cases with the lowest burden of pathology showed α-syn glial cytoplasmic inclusions (GCIs) in the cerebellum as well as in medullary and pontine cerebellar projections. Cerebellar pathology was highly selective and severely involved subcortical white matter, whereas deep white matter and granular layer were only mildly affected and the molecular layer was spared. Loss of Purkinje cells increased with disease duration and was associated with neuronal and axonal abnormalities. Neocortex, basal ganglia and spinal cord became consecutively involved with the increasing burden of α-syn pathology, followed by hippocampus, amygdala, and, finally, the visual cortex. GCIs were associated with myelinated axons, and the severity of GCIs correlated with demyelination.. Our findings indicate that cerebellar subcortical white matter and cerebellar brainstem projections are likely the earliest foci of α-syn pathology in MSA-C, followed by involvement of more widespread regions of the central nervous system and neurodegeneration with disease progression.

    Topics: Aged; alpha-Synuclein; Central Nervous System; Cerebellum; Disease Progression; Female; Humans; Male; Middle Aged; Multiple System Atrophy; Nerve Degeneration

2017
Sorbus alnifolia protects dopaminergic neurodegeneration in Caenorhabditis elegans.
    Pharmaceutical biology, 2017, Volume: 55, Issue:1

    The twigs of Sorbus alnifolia (Sieb. et Zucc.) K. Koch (Rosaceae) have been used to treat neurological disorders as a traditional medicine in Korea. However, there are limited data describing the efficacy of S. alnifolia in Parkinson's disease (PD).. This study was conducted to identify the protective effects of the methanol extracts of S. alnifolia (MESA) on the dopaminergic (DA) neurodegeneration in Caenorhabditis elegans.. To test the neuroprotective action of MESA, viability assay was performed after 48 h exposure to 1-methyl-4-phenylpyridine (MMP. MESA restored MPP. These results revealed that MESA protects DA neurodegeneration and recovers diminished DA neuronal functions, thereby can be a valuable candidate for the treatment of PD.

    Topics: 1-Methyl-4-phenylpyridinium; alpha-Synuclein; Animals; Animals, Genetically Modified; Behavior, Animal; Caenorhabditis elegans; Caenorhabditis elegans Proteins; Cell Survival; Cytoprotection; Dopaminergic Neurons; Dose-Response Relationship, Drug; Feeding Behavior; Longevity; Methanol; Motor Activity; Nerve Degeneration; Neuroprotective Agents; PC12 Cells; Phytotherapy; Plant Components, Aerial; Plant Extracts; Plants, Medicinal; Protein Aggregates; Rats; Solvents; Sorbus; Time Factors

2017
AAV1/2-induced overexpression of A53T-α-synuclein in the substantia nigra results in degeneration of the nigrostriatal system with Lewy-like pathology and motor impairment: a new mouse model for Parkinson's disease.
    Acta neuropathologica communications, 2017, Feb-01, Volume: 5, Issue:1

    Topics: Aged; Aged, 80 and over; alpha-Synuclein; Animals; Corpus Striatum; Dependovirus; Dopamine; Dopamine Plasma Membrane Transport Proteins; Genetic Vectors; Homovanillic Acid; Humans; Male; Mice, Inbred C57BL; Motor Activity; Mutation; Nerve Degeneration; Neurons; Parkinsonian Disorders; Substantia Nigra; Tyrosine 3-Monooxygenase

2017
Propagation of pathological α-synuclein in marmoset brain.
    Acta neuropathologica communications, 2017, Feb-02, Volume: 5, Issue:1

    α-Synuclein is a defining, key component of Lewy bodies and Lewy neurites in Parkinson's disease (PD) and dementia with Lewy bodies (DLB), as well as glial cytoplasmic inclusions in multiple system atrophy (MSA). The distribution and spreading of these pathologies are closely correlated with disease progression. Recent studies have revealed that intracerebral injection of synthetic α-synuclein fibrils or pathological α-synuclein prepared from DLB or MSA brains into wild-type or transgenic animal brains induced prion-like propagation of phosphorylated α-synuclein pathology. The common marmoset is a very small primate that is expected to be a useful model of human diseases. Here, we show that intracerebral injection of synthetic α-synuclein fibrils into adult wild-type marmoset brains (caudate nucleus and/or putamen) resulted in spreading of abundant α-synuclein pathologies, which were positive for various antibodies to α-synuclein, including phospho Ser129-specific antibody, anti-ubiquitin and anti-p62 antibodies, at three months after injection. Remarkably, robust Lewy body-like inclusions were formed in tyrosine hydroxylase (TH)-positive neurons in these marmosets, strongly suggesting the retrograde spreading of abnormal α-synuclein from striatum to substantia nigra. Moreover, a significant decrease in the numbers of TH-positive neurons was observed in the injection-side of the brain, where α-synuclein inclusions were deposited. Furthermore, most of the α-synuclein inclusions were positive for 1-fluoro-2,5-bis (3-carboxy-4-hydroxystyryl) benzene (FSB) and thioflavin-S, which are dyes widely used to visualize the presence of amyloid. Thus, injection of synthetic α-synuclein fibrils into brains of non-transgenic primates induced PD-like α-synuclein pathologies within only 3 months after injection. Finally, we provide evidence indicating that neurons with abnormal α-synuclein inclusions may be cleared by microglial cells. This is the first marmoset model for α-synuclein propagation. It should be helpful in studies to elucidate mechanisms of disease progression and in development and evaluation of disease-modifying drugs for α-synucleinopathies.

    Topics: alpha-Synuclein; Animals; Benzothiazoles; Brain; Callithrix; Female; Immunohistochemistry; Lewy Bodies; Microglia; Nerve Degeneration; Neurons; Parkinsonian Disorders; Protein Aggregation, Pathological; Recombinant Proteins; Sequence Homology, Amino Acid; Thiazoles; Tyrosine 3-Monooxygenase

2017
Neuronal Loss and Α-Synuclein Pathology in the Superior Colliculus and Its Relationship to Visual Hallucinations in Dementia with Lewy Bodies.
    The American journal of geriatric psychiatry : official journal of the American Association for Geriatric Psychiatry, 2017, Volume: 25, Issue:6

    Patients with dementia with Lewy bodies (DLB) often experience visual hallucinations, which are related to decreased quality of life for patients and increased caregiver distress. The pathologic changes that contribute to visual hallucinations are not known, but several hypotheses implicate deficient attentional processing. The superior colliculus has a role in visual attention and planning eye movements and has been directly implicated in several models of visual hallucinations. Therefore, the present study sought to identify neurodegenerative changes that may contribute to hallucinations in DLB.. Postmortem superior colliculus tissue from 13 comparison, 10 DLB, and 10 Alzheimer disease (AD) cases was evaluated using quantitative neuropathologic methods.. α-Synuclein and tau deposition were more severe in deeper layers of the superior colliculus. DLB cases had neuronal density reductions in the stratum griseum intermedium, an important structure in directing attention toward visual targets. In contrast, neuronal density was reduced in all laminae of the superior colliculus in AD.. These findings suggest that regions involved in directing attention toward visual targets are subject to neurodegenerative changes in DLB. Considering several hypotheses of visual hallucinations implicating dysfunctional attention toward external stimuli, these findings may provide evidence of pathologic changes that contribute to the manifestation of visual hallucinations in DLB.

    Topics: Aged; Aged, 80 and over; alpha-Synuclein; Alzheimer Disease; Case-Control Studies; Cell Count; Female; Hallucinations; Humans; Lewy Body Disease; Male; Middle Aged; Nerve Degeneration; Superior Colliculi; Tauopathies

2017
Fenpropathrin, a Widely Used Pesticide, Causes Dopaminergic Degeneration.
    Molecular neurobiology, 2016, Volume: 53, Issue:2

    Fenpropathrin is one of the widely used pyrethroids in agriculture and household and also reported to have neurotoxic effects in rodent models. In our Parkinson's disease (PD) clinic, there was a unique patient with a history of daily exposure to fenpropathrin for 6 months prior to developing Parkinsonian symptoms progressively. Since whether fenpropathrin is related to any dopaminergic degeneration was unknown, we aimed in this study to evaluate the neurotoxic effects of fenpropathrin on the dopaminergic system and associated mechanisms in vitro and in vivo. In cultured SH-SY5Y cells, fenpropathrin caused cell death, reactive oxygen species generation, Lewy body-associated proteins aggregation, and Lewy body-like intracytoplasmic inclusions formation. In rodent animals, two different injections of fenpropathrin were used for administrations, intraperitoneal (i.p), or stereotaxical (ST). The rats exhibited lower number of pokes 60 days after first i.p injection, while the rats in ST group showed a significant upregulation of apomorphine-evoked rotations 60 days after first injection. Decreased tyrosine hydroxylase (TH) and vesicular monoamine transporter 2 (VMAT2) immunoreactivity, while increased dopamine transporter (DAT) immunoreactivity were observed in rats of either i.p or ST group 60 days after the last exposure to fenpropathrin. However, the number of TH-positive cells in the substantia nigra was more reduced 120 days after the first i.p injection than those of 60 days. Our data demonstrated that exposure to fenpropathrin could mimic the pathologic and pathogenetic features of PD especially in late onset cases. These results imply fenpropathrin as a DA neurotoxin and a possible environmental risk factor for PD.

    Topics: alpha-Synuclein; Animals; Apoptosis; Behavior, Animal; Body Weight; Cell Line, Tumor; Cell Proliferation; Cell Shape; Cell Survival; Dopamine; Dopamine Plasma Membrane Transport Proteins; Down-Regulation; Humans; Lewy Bodies; Matrix Metalloproteinases; Motor Activity; Nerve Degeneration; Neurons; Pesticides; Protein Aggregates; Pyrethrins; Rats, Sprague-Dawley; Reactive Oxygen Species; RNA-Binding Proteins; Time Factors; Ubiquitin; Up-Regulation; Vesicular Monoamine Transport Proteins

2016
α-Synuclein interferes with the ESCRT-III complex contributing to the pathogenesis of Lewy body disease.
    Human molecular genetics, 2016, Mar-15, Volume: 25, Issue:6

    α-Synuclein (α-syn) has been implicated in neurological disorders with parkinsonism, including Parkinson's disease and Dementia with Lewy body. Recent studies have shown α-syn oligomers released from neurons can propagate from cell-to-cell in a prion-like fashion exacerbating neurodegeneration. In this study, we examined the role of the endosomal sorting complex required for transport (ESCRT) pathway on the propagation of α-syn. α-syn, which is transported via the ESCRT pathway through multivesicular bodies for degradation, can also target the degradation of the ESCRT protein-charged multivesicular body protein (CHMP2B), thus generating a roadblock of endocytosed α-syn. Disruption of the ESCRT transport system also resulted in increased exocytosis of α-syn thus potentially increasing cell-to-cell propagation of synuclein. Conversely, delivery of a lentiviral vector overexpressing CHMP2B rescued the neurodegeneration in α-syn transgenic mice. Better understanding of the mechanisms of intracellular trafficking of α-syn might be important for understanding the pathogenesis and developing new treatments for synucleinopathies.

    Topics: alpha-Synuclein; Animals; Brain; Case-Control Studies; Cell Line; Disease Models, Animal; Endosomal Sorting Complexes Required for Transport; Humans; Lewy Bodies; Lewy Body Disease; Male; Mice; Mice, Inbred C57BL; Nerve Degeneration; Neurons; Parkinson Disease; Parkinsonian Disorders

2016
Synphilin-1 attenuates mutant LRRK2-induced neurodegeneration in Parkinson's disease models.
    Human molecular genetics, 2016, Feb-15, Volume: 25, Issue:4

    Mutations in leucine-rich repeat kinase 2 (LRRK2) cause autosomal-dominant Parkinsonism with pleomorphic pathology including deposits of aggregated protein and neuronal degeneration. The pathogenesis of LRRK2-linked Parkinson's disease (PD) is not fully understood. Here, using co-immunoprecipitation, we found that LRRK2 interacted with synphilin-1 (SP1), a cytoplasmic protein that interacts with α-synuclein and has implications in PD pathogenesis. LRRK2 interacted with the N-terminus of SP1 whereas SP1 predominantly interacted with the C-terminus of LRRK2, including kinase domain. Co-expression of SP1 with LRRK2 increased LRRK2-induced cytoplasmic aggregation in cultured cells. Moreover, SP1 also attenuated mutant LRRK2-induced toxicity and reduced LRRK2 kinase activity in cultured cells. Knockdown of SP1 by siRNA enhanced LRRK2 neuronal toxicity. In vivo Drosophila studies, co-expression of SP1 and mutant G2019S-LRRK2 in double transgenic Drosophila increased survival and improved locomotor activity. Expression of SP1 protects against G2019S-LRRK2-induced dopamine neuron loss and reduced LRRK2 phosphorylation in double transgenic fly brains. Our findings demonstrate that SP1 attenuates mutant LRRK2-induced PD-like phenotypes and plays a neural protective role.

    Topics: alpha-Synuclein; Animals; Animals, Genetically Modified; Carrier Proteins; Cell Line, Tumor; Disease Models, Animal; Dopamine; Drosophila; Gene Knockdown Techniques; HEK293 Cells; Humans; Leucine-Rich Repeat Serine-Threonine Protein Kinase-2; Mice; Mutation; Nerve Degeneration; Nerve Tissue Proteins; Neurons; Parkinson Disease; Phosphorylation; Protein Interaction Domains and Motifs

2016
α-Mangostin Inhibits α-Synuclein-Induced Microglial Neuroinflammation and Neurotoxicity.
    Cellular and molecular neurobiology, 2016, Volume: 36, Issue:5

    Microglia-mediated neuroinflammation induced by α-synuclein in the substantianigra likely either initiates or aggravates nigral neuro degeneration in Parkinson's disease (PD). We aimed to explore the effects of α-mangostin (α-M), a polyphenolicxanthone derivative from mangosteen on α-synuclein-stimulated DA neurodegeneration. Primary microglia, mesencephalic neuron, mesencephalic neuron-glianeuronal cultures, and transwell co-cultures were prepared separately. Liquid scintillation counting was used to determine the radioactivity in DA uptake. Enzyme-linked immunosorbent assay (ELISA) was performed in the IL-1β, IL-6, and TNF-α assay. The expression of proteins was analyzed by Western blot. α-M inhibited the increased levels of pro-inflammatory cytokines, NO, and ROS in α-synuclein-stimulated primary microglia. Mechanistic study revealed that α-M functioned by inhibition of nuclear factor kappa B (NF-κB) and NADPH oxidase. Further, α-M protected α-synuclein-induced microglial and direct neurotoxicity. Although detailed mechanisms remain to be defined, our observations suggest a potential compound, which inhibits microglial activation induced by α-synuclein by targeting NADPH oxidase, might be a therapeutic possibility in preventing PD progression.

    Topics: alpha-Synuclein; Animals; Cells, Cultured; Dopamine; Humans; Mesencephalon; Microglia; NADPH Oxidases; Nerve Degeneration; Neurons; Parkinson Disease; Rats, Sprague-Dawley; Xanthones

2016
Glucocerebrosidase Deficiency in Drosophila Results in α-Synuclein-Independent Protein Aggregation and Neurodegeneration.
    PLoS genetics, 2016, Volume: 12, Issue:3

    Mutations in the glucosidase, beta, acid (GBA1) gene cause Gaucher's disease, and are the most common genetic risk factor for Parkinson's disease (PD) and dementia with Lewy bodies (DLB) excluding variants of low penetrance. Because α-synuclein-containing neuronal aggregates are a defining feature of PD and DLB, it is widely believed that mutations in GBA1 act by enhancing α-synuclein toxicity. To explore this hypothesis, we deleted the Drosophila GBA1 homolog, dGBA1b, and compared the phenotypes of dGBA1b mutants in the presence and absence of α-synuclein expression. Homozygous dGBA1b mutants exhibit shortened lifespan, locomotor and memory deficits, neurodegeneration, and dramatically increased accumulation of ubiquitinated protein aggregates that are normally degraded through an autophagic mechanism. Ectopic expression of human α-synuclein in dGBA1b mutants resulted in a mild enhancement of dopaminergic neuron loss and increased α-synuclein aggregation relative to controls. However, α-synuclein expression did not substantially enhance other dGBA1b mutant phenotypes. Our findings indicate that dGBA1b plays an important role in the metabolism of protein aggregates, but that the deleterious consequences of mutations in dGBA1b are largely independent of α-synuclein. Future work with dGBA1b mutants should reveal the mechanism by which mutations in dGBA1b lead to accumulation of protein aggregates, and the potential influence of this protein aggregation on neuronal integrity.

    Topics: alpha-Synuclein; Animals; Dopaminergic Neurons; Drosophila melanogaster; Gaucher Disease; Glucosylceramidase; Humans; Lysosomes; Nerve Degeneration; Parkinson Disease; Phenotype; Protein Aggregation, Pathological

2016
Post-translational modifications of α-synuclein contribute to neurodegeneration in the colon of elderly individuals.
    Molecular medicine reports, 2016, Volume: 13, Issue:6

    Synucleinopathies and abnormalities in the nerves of the enteric nervous system are hypothesized to be involved in age-associated motility disorders. The aim of the present study was to investigate the expression of various antigens, including α‑synuclein (Syn) and its post‑translational modified forms, in the human colon at various ages. In addition, the study aimed to correlate the expression of Syn with neurodegeneration. Immunohistochemistry was used to detect the expression of neurofilament (NF), Syn, as well as its nitrated (N) form in the healthy colonic tissue of 12 young (34.08±5.12 years), 10 middle‑aged (51.80±3.52 years), and 11 elderly (75.82±7.70 years) individuals. To the best of our knowledge, the current study is the first to demonstrate the presence of N‑Syn in the colonic tissue. N‑Syn was identified in the upper layer of the mucosa and submucosa layer. Furthermore, Syn (wild‑type) was present in the mucosa and submucosa. The number of NF‑positive neurons in the submucosal layer declined significantly with age (P<0.01). In addition, Syn and N‑Syn significantly increased during aging (P<0.01). Furthermore, a negative correlation was identified between neuron number and synucleinopathies, indicating the abnormal accumulation of both wild-type Syn and N‑Syn in the mucosa, submucosa, muscle layer and myenteric plexus. The present study demonstrates that the Syn pathology may be linked to colic neuronal degeneration during normal aging, and this link may cause functional deficits.

    Topics: Adult; Aged; Aged, 80 and over; alpha-Synuclein; Colon; Female; Humans; Immunohistochemistry; Male; Middle Aged; Myenteric Plexus; Nerve Degeneration; Neurons; Protein Processing, Post-Translational; Submucous Plexus

2016
Interactions of GFAP with ceftriaxone and phenytoin: SRCD and molecular docking and dynamic simulation.
    Biochimica et biophysica acta, 2016, Volume: 1860, Issue:10

    GFAP is the major intermediate filament protein in mature astrocytes. Its increased expression and aggregation was firstly associated to Alexander's disease, and successively in different neurological diseases including scrapie, Alzheimer's and Creutzfeld-Jacob diseases. Recently, ceftriaxone a multi-potent β-lactam antibiotic able to overcome the blood-brain barrier, successfully eliminated the cellular toxic effects of misfolded mutated GFAP, similarly to phenytoin sodium, in a cellular model of Alexander's disease and inhibited α-synuclein aggregation protecting PC12 cells from the exposure to 6-hydroxydopamine.. In this study, synchrotron radiation circular dichroism spectroscopy has been used to obtain structural information about the GFAP-ceftriaxone (phenytoin) interactions, while computational methods allowed the identification of the relevant putative binding site of either ceftriaxone or phenytoin on the dimer structure of GFAP, permitting to rationalize the spectroscopic experimental results.. We found that GFAP exhibited enhanced stability upon the addition of two equivalents of each ligands with ceftriaxone imparting a more spontaneous interactions and a more ordered complex system than phenytoin.. SRCD data and MD models indicate a stronger protective effect of ceftriaxone in neurological disorders characterized by an increased production and polymerization of GFAP.. This result, in addition to our previous works in which we documented that ceftriaxone interacts with α-synuclein inhibiting its pathological aggregation and that a cyclical treatment with this molecule in a patient with adult-onset Alexander's disease halted, and partly reversed, the progression of neurodegeneration, suggests the possibility of a chaperone-like effect of ceftriaxone on protein involved in specific neurodegenerative diseases.

    Topics: Alexander Disease; alpha-Synuclein; Animals; Astrocytes; Binding Sites; Ceftriaxone; Glial Fibrillary Acidic Protein; Humans; Intermediate Filament Proteins; Molecular Docking Simulation; Molecular Dynamics Simulation; Nerve Degeneration; PC12 Cells; Phenytoin; Protein Aggregation, Pathological; Rats

2016
α-Synuclein induced toxicity in brain stem serotonin neurons mediated by an AAV vector driven by the tryptophan hydroxylase promoter.
    Scientific reports, 2016, 05-23, Volume: 6

    We studied the impact of α-synuclein overexpression in brainstem serotonin neurons using a novel vector construct where the expression of human wildtype α-synuclein is driven by the tryptophan hydroxylase promoter, allowing expression of α-synuclein at elevated levels, and with high selectivity, in serotonergic neurons. α-Synuclein induced degenerative changes in axons and dendrites, displaying a distorted appearance, suggesting accumulation and aggregation of α-synuclein as a result of impaired axonal transport, accompanied by a 40% loss of terminals, as assessed in the hippocampus. Tissue levels of serotonin and its major metabolite 5-HIAA remained largely unaltered, and the performance of the α-synuclein overexpressing rats in tests of spatial learning (water maze), anxiety related behavior (elevated plus maze) and depressive-like behavior (forced swim test) was not different from control, suggesting that the impact of the developing axonal pathology on serotonin neurotransmission was relatively mild. Overexpression of α-synuclein in the raphe nuclei, combined with overexpression in basal forebrain cholinergic neurons, resulted in more pronounced axonal pathology and significant impairment in the elevated plus maze. We conclude that α-synuclein pathology in serotonergic or cholinergic neurons alone is not sufficient to impair non-motor behaviors, but that it is their simultaneous involvement that determines severity of such symptoms.

    Topics: alpha-Synuclein; Animals; Behavior, Animal; Brain Stem; Cholinergic Neurons; Dependovirus; Female; Genetic Vectors; Humans; Maze Learning; Nerve Degeneration; Promoter Regions, Genetic; Raphe Nuclei; Rats; Rats, Sprague-Dawley; Recombinant Proteins; Serotonergic Neurons; Tryptophan Hydroxylase; Up-Regulation

2016
Molecular Features Underlying Neurodegeneration Identified through In Vitro Modeling of Genetically Diverse Parkinson's Disease Patients.
    Cell reports, 2016, 06-14, Volume: 15, Issue:11

    The fact that Parkinson's disease (PD) can arise from numerous genetic mutations suggests a unifying molecular pathology underlying the various genetic backgrounds. To address this hypothesis, we took an integrated approach utilizing in vitro disease modeling and comprehensive transcriptome profiling to advance our understanding of PD progression and the concordant downstream signaling pathways across divergent genetic predispositions. To model PD in vitro, we generated neurons harboring disease-causing mutations from patient-specific, induced pluripotent stem cells (iPSCs). We observed signs of degeneration in midbrain dopaminergic neurons, reflecting the cardinal feature of PD. Gene expression signatures of PD neurons provided molecular insights into disease phenotypes observed in vitro, including oxidative stress vulnerability and altered neuronal activity. Notably, PD neurons show that elevated RBFOX1, a gene previously linked to neurodevelopmental diseases, underlies a pattern of alternative RNA-processing associated with PD-specific phenotypes.

    Topics: alpha-Synuclein; Alternative Splicing; Cell Differentiation; Cell Line; Dopaminergic Neurons; Genes, Mitochondrial; Genetic Heterogeneity; Humans; Induced Pluripotent Stem Cells; Mesencephalon; Models, Biological; Nerve Degeneration; Neural Stem Cells; Neurites; Neurotoxins; Oxidative Stress; Parkinson Disease; Phenotype; RNA Splicing Factors; Sequence Analysis, RNA; Transcriptome

2016
Aberrant adenosine A2A receptor signaling contributes to neurodegeneration and cognitive impairments in a mouse model of synucleinopathy.
    Experimental neurology, 2016, Volume: 283, Issue:Pt A

    Synucleinopathy is characterized by abnormal accumulation of misfolded α-synuclein (α-Syn)-positive cytoplasmic inclusions and by neurodegeneration and cognitive impairments, but the pathogenesis mechanism of synucleinopathy remains to be defined. Using a transmission model of synucleinopathy by intracerebral injection of preformed A53T α-Syn fibrils, we investigated whether aberrant adenosine A2A receptor (A2AR) signaling contributed to pathogenesis of synucleinopathy. We demonstrated that intra-hippocampal injection of preformed mutant α-Syn fibrils triggered a striking and selective induction of A2AR expression which was closely co-localized with pSer129 α-Syn-rich inclusions in neurons and glial cells of hippocampus. Importantly, by abolishing aberrant A2AR signaling triggered by mutant α-Syn, genetic deletion of A2ARs blunted a cascade of pathological events leading to synucleinopathy, including pSer129 α-Syn-rich and p62-positive aggregates, NF-κB activation and astrogliosis, apoptotic neuronal cell death and working memory deficits without affecting motor activity. These findings define α-Syn-triggered aberrant A2AR signaling as a critical pathogenesis mechanism of synucleinopathy with dual controls of cognition and neurodegeneration by modulating α-Syn aggregates. Thus, aberrant A2AR signaling represents a useful biomarker as well as a therapeutic target of synucleinopathy.

    Topics: alpha-Synuclein; Analysis of Variance; Animals; Cognition Disorders; Disease Models, Animal; Exploratory Behavior; Hippocampus; In Situ Nick-End Labeling; Mice; Mice, Inbred C57BL; Mice, Knockout; Mutation; Nerve Degeneration; Nerve Tissue Proteins; Neuroglia; Receptor, Adenosine A2A; RNA, Messenger; Signal Transduction

2016
Neurodegeneration in tauopathies and synucleinopathies.
    Revue neurologique, 2016, Volume: 172, Issue:11

    While increasing life expectancy is a major achievement, the global aging of societies raises a number of medical issues, such as the development of age-related disorders, including neurodegenerative diseases. The three main disease groups constituting the majority of neurodegenerative diseases are tauopathies, alpha-synucleinopathies and diseases due to repetitions of glutamine (including Huntington's disease). In each neurodegenerative disease, the accumulation of one or more aggregated proteins has been identified as the molecular signature of the disease (as seen, for example, in Alzheimer's disease, Parkinson's disease, dementia with Lewy bodies, amyotrophic lateral sclerosis and frontotemporal dementia). The etiology of neurodegenerative diseases is often multifactorial, and the known risk factors include, in addition to genetic polymorphisms and age, some other possible causes, such as certain immune and metabolic conditions, endocrine pathologies, gender, socioeconomic or professional status, oxidative stress or inflammation, vitamin deficiencies and environmental factors (chemical exposure, metals). However, innovative strategies to elaborate suitable diagnostic and therapeutic approaches (aiming to at least delay or possibly even reverse disease progression) require further knowledge of the genetic and adaptive immunological characteristics of neurodegenerative diseases.

    Topics: alpha-Synuclein; Brain; Cytoskeleton; Humans; Inclusion Bodies; Lewy Body Disease; Nerve Degeneration; Neurodegenerative Diseases; Parkinsonian Disorders; Tauopathies

2016
Impairment of chaperone-mediated autophagy induces dopaminergic neurodegeneration in rats.
    Autophagy, 2016, Volume: 12, Issue:11

    Chaperone-mediated autophagy (CMA) involves the selective lysosomal degradation of cytosolic proteins such as SNCA (synuclein α), a protein strongly implicated in Parkinson disease (PD) pathogenesis. However, the physiological role of CMA and the consequences of CMA failure in the living brain remain elusive. Here we show that CMA inhibition in the adult rat substantia nigra via adeno-associated virus-mediated delivery of short hairpin RNAs targeting the LAMP2A receptor, involved in CMA's rate limiting step, was accompanied by intracellular accumulation of SNCA-positive puncta, which were also positive for UBIQUITIN, and in accumulation of autophagic vacuoles within LAMP2A-deficient nigral neurons. Strikingly, LAMP2A downregulation resulted in progressive loss of nigral dopaminergic neurons, severe reduction in striatal dopamine levels/terminals, increased astro- and microgliosis and relevant motor deficits. Thus, this study highlights for the first time the importance of the CMA pathway in the dopaminergic system and suggests that CMA impairment may underlie PD pathogenesis.

    Topics: alpha-Synuclein; Animals; Autophagy; Behavior, Animal; Dependovirus; Dopaminergic Neurons; Down-Regulation; Female; Gene Silencing; Genetic Vectors; Lysosomal-Associated Membrane Protein 2; Molecular Chaperones; Neostriatum; Nerve Degeneration; Phenotype; Rats, Wistar; RNA, Small Interfering; Sequestosome-1 Protein; Substantia Nigra; Transduction, Genetic; Ubiquitin; Vacuoles

2016
Caspase-1 clipping causes complications for α-synuclein.
    Proceedings of the National Academy of Sciences of the United States of America, 2016, Sep-06, Volume: 113, Issue:36

    Topics: alpha-Synuclein; Caspase 1; Humans; Nerve Degeneration; Neurons; Parkinson Disease; Protein Aggregation, Pathological; Proteolysis

2016
Munc18-1 is a molecular chaperone for α-synuclein, controlling its self-replicating aggregation.
    The Journal of cell biology, 2016, 09-12, Volume: 214, Issue:6

    Munc18-1 is a key component of the exocytic machinery that controls neurotransmitter release. Munc18-1 heterozygous mutations cause developmental defects and epileptic phenotypes, including infantile epileptic encephalopathy (EIEE), suggestive of a gain of pathological function. Here, we used single-molecule analysis, gene-edited cells, and neurons to demonstrate that Munc18-1 EIEE-causing mutants form large polymers that coaggregate wild-type Munc18-1 in vitro and in cells. Surprisingly, Munc18-1 EIEE mutants also form Lewy body-like structures that contain α-synuclein (α-Syn). We reveal that Munc18-1 binds α-Syn, and its EIEE mutants coaggregate α-Syn. Likewise, removal of endogenous Munc18-1 increases the aggregative propensity of α-Syn(WT) and that of the Parkinson's disease-causing α-Syn(A30P) mutant, an effect rescued by Munc18-1(WT) expression, indicative of chaperone activity. Coexpression of the α-Syn(A30P) mutant with Munc18-1 reduced the number of α-Syn(A30P) aggregates. Munc18-1 mutations and haploinsufficiency may therefore trigger a pathogenic gain of function through both the corruption of native Munc18-1 and a perturbed chaperone activity for α-Syn leading to aggregation-induced neurodegeneration.

    Topics: alpha-Synuclein; Animals; Animals, Newborn; Genotype; Haploinsufficiency; Lewy Bodies; Microscopy, Fluorescence; Models, Molecular; Molecular Chaperones; Munc18 Proteins; Mutation; Nerve Degeneration; Neurons; Parkinson Disease; PC12 Cells; Protein Aggregates; Protein Binding; Protein Conformation; Rats; Rats, Sprague-Dawley; Recombinant Fusion Proteins; RNA Interference; Time Factors; Transfection

2016
DNA damage preceding dopamine neuron degeneration in A53T human α-synuclein transgenic mice.
    Biochemical and biophysical research communications, 2016, Dec-02, Volume: 481, Issue:1-2

    Defective DNA repair has been linked with age-associated neurodegenerative disorders. Parkinson's disease (PD) is a progressive neurodegenerative disorder caused by genetic and environmental factors. Whether damages to nuclear DNA contribute to neurodegeneration of PD still remain obscure. in this study we aim to explore whether nuclear DNA damage induce dopamine neuron degeneration in A53T human α-Synuclein over expressed mouse model. We investigated the effects of X-ray irradiation on A53T-α-Syn MEFs and A53T-α-Syn transgene mice. Our results indicate that A53T-α-Syn MEFs show a prolonged DNA damage repair process and senescense phenotype. DNA damage preceded onset of motor phenotype in A53T-α-Syn transgenic mice and decrease the number of nigrostriatal dopaminergic neurons. Neurons of A53T-α-Syn transgenic mice are more fragile to DNA damages.

    Topics: alpha-Synuclein; Animals; Cell Line; DNA Damage; Dopaminergic Neurons; Humans; Mice; Mice, Transgenic; Nerve Degeneration; Parkinson Disease

2016
Lipidomic and proteomic characterization of platelet extracellular vesicle subfractions from senescent platelets.
    Transfusion, 2015, Volume: 55, Issue:3

    Platelets (PLTs) in stored PLT concentrates (PLCs) release PLT extracellular vesicles (PL-EVs) induced by senescence and activation, resembling the PLT storage lesion. No comprehensive classification or molecular characterization of senescence-induced PL-EVs exists to understand PL-EV heterogeneity.. PL-EVs from 5-day-stored PLCs from healthy individuals were isolated and subfractionated by differential centrifugation, filtration, and density gradient ultracentrifugation into five PLT microvesicle (PL-MV) subfractions (Fraction [F]1-F5) and PLT exosomes (PL-EXs). PL-EV size, concentration, and composition were analyzed by nanoparticle tracking analysis, flow cytometry, and lipid and protein mass spectrometry. Protein data were verified by Western blot.. PL-EVs showed overlapping mean particle sizes of 180 to 260 nm, but differed significantly in composition. Less dense, intermediate, and dense PL-MVs enriched specific lipidomic and proteomic markers related to the plasma membrane, intracellular membranes, PLT granules, mitochondria, and PLT activation. α-Synuclein (81% of total) accumulated in F1 and F2, amyloid-β (Aβ) precursor protein in F3 and F4 (84%), and apolipoprotein (Apo)E (88%) and ApoJ (92%) in F3 to F5. PL-EXs enriched lipid species and proteins, with high abundance of lipid raft, PLT adhesion, and immune response-related markers.. Differential lipid and protein compositions of PL-EVs suggest their unique cellular origins and functions, partly overlapping with PLT granule secretion. Dense PL-MVs might represent autophagic vesicles released during PLT activation and apoptosis and PL-EXs resemble lipid rafts, with a potential role in PLT aggregation and immunity. Segregation of α-synuclein and Aβ precursor protein, ApoE, and ApoJ into less dense and dense PL-MVs, respectively, show their differential carrier role of neurologic disease-related cargo.

    Topics: Adult; alpha-Synuclein; Blood Platelets; Blood Preservation; Blood Proteins; Blotting, Western; Cell-Derived Microparticles; Cellular Senescence; Centrifugation; Erythrocytes; Exosomes; Filtration; Flow Cytometry; Humans; Immunomagnetic Separation; Lipids; Mass Spectrometry; Membrane Lipids; Membrane Proteins; Nanoparticles; Nerve Degeneration; Platelet Activation; Plateletpheresis

2015
The Parkinson's disease-related protein DJ-1 protects dopaminergic neurons in vivo and cultured cells from alpha-synuclein and 6-hydroxydopamine toxicity.
    Neuro-degenerative diseases, 2015, Volume: 15, Issue:1

    Dopaminergic degeneration is a major finding in brains of patients with Parkinson's disease (PD), together with Lewy bodies, intraneuronal inclusions mainly composed of the fibrillogenic protein α-synuclein (α-syn). The familial-PD-related protein DJ-1 was reported to reduce dopaminergic degeneration triggered by α-syn or by the dopaminergic-selective neurotoxin 6-hydroxydopamine (6-OHDA).. The aim was to further investigate the role of DJ-1 in dopaminergic degeneration and to see whether a cell-permeable recombinant form of DJ-1 (TAT-DJ-1) could restore dopamine depletion in vivo, thus representing an innovative therapeutic approach.. We developed in vitro (PC12/TetOn cells and mouse primary mesencephalic neurons) and in vivo models [including DJ-1 knockout (-/-) mice] to investigate DJ-1 in dopaminergic degeneration.. We found that in PC12/TetOn cells overexpressing α-syn with the familial-PD linked mutation A30P, DJ-1 silencing increased α-syn (A30P) toxicity. Primary mesencephalic neurons from DJ-1 (-/-) mice were more vulnerable to a cell-permeable form of α-syn (TAT-α-syn) and to 6-OHDA. Intrastriatally administered TAT-DJ-1 reduced 6-OHDA toxicity in vivo in C57BL/6 mice. Finally, when we injected TAT-α-syn (A30P) in the striatum of DJ-1 (-/-) animals, dopamine was depleted more than in the control strain.. DJ-1 appears to have a protective role against dopaminergic degeneration triggered by α-syn or 6-OHDA, reinforcing the possible therapeutic importance of this protein in PD.

    Topics: alpha-Synuclein; Animals; Cells, Cultured; Disease Models, Animal; Dopaminergic Neurons; Mesencephalon; Mice; Mice, Inbred C57BL; Mice, Knockout; Mutation; Nerve Degeneration; Oncogene Proteins; Oxidopamine; Parkinson Disease; Peroxiredoxins; Protein Deglycase DJ-1; Up-Regulation

2015
Aneuploidy in Lewy body diseases.
    Neurobiology of aging, 2015, Volume: 36, Issue:3

    An increase in DNA content is associated with neuronal degeneration in Alzheimer's disease but has not been evaluated in Lewy body diseases. Using stereological principles, flow cytometry, and standard histopathologic methods, we evaluated the number and DNA content of neurons and all cells and the severity of Lewy and Alzheimer pathologies, in brain regions affected at different stages in Lewy body diseases compared with controls. An increase in neuronal DNA content was observed in all the affected brain regions examined, although this change was related to different pathologies. In the substantia nigra, increased neuronal DNA content related to neuronal loss, whereas in the cortex and hippocampus, increased neuronal DNA content related to Alzheimer pathologies. Of note, increased neuronal DNA content did not relate to the deposition of Lewy bodies in any region examined. These data support the concept that increased DNA content increases neuronal susceptibility to degeneration and Alzheimer pathologies.

    Topics: Aged; Aged, 80 and over; alpha-Synuclein; Aneuploidy; Cerebral Cortex; DNA; Female; Hippocampus; Humans; Lewy Bodies; Lewy Body Disease; Male; Nerve Degeneration; Neurofibrillary Tangles

2015
A53T human α-synuclein overexpression in transgenic mice induces pervasive mitochondria macroautophagy defects preceding dopamine neuron degeneration.
    The Journal of neuroscience : the official journal of the Society for Neuroscience, 2015, Jan-21, Volume: 35, Issue:3

    In vitro evidence suggests that the inefficient removal of damaged mitochondria by macroautophagy contributes to Parkinson's disease (PD). Using a tissue-specific gene amplification strategy, we generated a transgenic mouse line with human α-synuclein A53T overexpression specifically in dopamine (DA) neurons. Transgenic mice showed profound early-onset mitochondria abnormalities, characterized by macroautophagy marker-positive cytoplasmic inclusions containing mainly mitochondrial remnants, which preceded the degeneration of DA neurons. Genetic deletion of either parkin or PINK1 in these transgenic mice significantly worsened mitochondrial pathologies, including drastically enlarged inclusions and loss of total mitochondria contents. These data suggest that mitochondria are the main targets of α-synuclein and their defective autophagic clearance plays a significant role during pathogenesis. Moreover, endogenous PINK1 or parkin is indispensable for the proper autophagic removal of damaged mitochondria. Our data for the first time establish an essential link between mitochondria macroautophagy impairments and DA neuron degeneration in an in vivo model based on known PD genetics. The model, its well-defined pathologies, and the demonstration of a main pathogenesis pathway in the present study have set the stage and direction of emphasis for future studies.

    Topics: alpha-Synuclein; Animals; Autophagy; Cell Count; Corpus Striatum; Disease Models, Animal; Dopaminergic Neurons; Humans; Mice; Mice, Transgenic; Mitochondria; Nerve Degeneration; Parkinson Disease; Protein Kinases; Ubiquitin-Protein Ligases

2015
X-linked Parkinson's disease.
    Movement disorders : official journal of the Movement Disorder Society, 2015, Volume: 30, Issue:2

    Topics: alpha-Synuclein; Female; Genes, X-Linked; Humans; Intellectual Disability; Male; Nerve Degeneration; Parkinson Disease; rab GTP-Binding Proteins

2015
PGC-1α activity in nigral dopamine neurons determines vulnerability to α-synuclein.
    Acta neuropathologica communications, 2015, Apr-01, Volume: 3

    Mitochondrial dysfunction and oxidative stress are critical factors in the pathogenesis of age-dependent neurodegenerative diseases. PGC-1α, a master regulator of mitochondrial biogenesis and cellular antioxidant defense, has emerged as a possible therapeutic target for Parkinson's disease, with important roles in the function and survival of dopaminergic neurons in the substantia nigra. The objective of this study is to determine if the loss of PGC-1α activity contributes to α-synuclein-induced degeneration.. We explore the vulnerability of PGC-1α null mice to the accumulation of human α-synuclein in nigral neurons, and assess the neuroprotective effect of AAV-mediated PGC-1α expression in this experimental model. Using neuronal cultures derived from these mice, mitochondrial respiration and production of reactive oxygen species are assessed in conditions of human α-synuclein overexpression. We find ultrastructural evidence for abnormal mitochondria and fragmented endoplasmic reticulum in the nigral dopaminergic neurons of PGC-1α null mice. Furthermore, PGC-1α null nigral neurons are more prone to degenerate following overexpression of human α-synuclein, an effect more apparent in male mice. PGC-1α overexpression restores mitochondrial morphology, oxidative stress detoxification and basal respiration, which is consistent with the observed neuroprotection against α-synuclein toxicity in male PGC-1α null mice.. Altogether, our results highlight an important role for PGC-1α in controlling the mitochondrial function of nigral neurons accumulating α-synuclein, which may be critical for gender-dependent vulnerability to Parkinson's disease.

    Topics: alpha-Synuclein; Animals; Cell Culture Techniques; Dopaminergic Neurons; Female; Humans; Male; Mice; Mice, Inbred C57BL; Mice, Knockout; Mitochondria; Nerve Degeneration; Oxidative Stress; Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha; Sex Factors; Substantia Nigra; Transcription Factors

2015
Neuron-to-neuron α-synuclein propagation in vivo is independent of neuronal injury.
    Acta neuropathologica communications, 2015, Mar-24, Volume: 3

    Interneuronal propagation of α-synuclein has been demonstrated in a variety of experimental models and may be involved in disease progression during the course of human synucleinopathies. The aim of this study was to assess the role that neuronal injury or, vice versa, cell integrity could have in facilitating interneuronal α-synuclein transfer and consequent protein spreading in an in vivo animal model.. Viral vectors carrying the DNA for human α-synuclein were injected into the rat vagus nerve to trigger protein overexpression in the medulla oblongata and consequent spreading of human α-synuclein toward pons, midbrain and forebrain. Two vector preparations sharing the same viral construct were manufactured using identical procedures with the exception of methods for their purification. They were also injected at concentrations that induced comparable levels of α-synuclein transduction/overexpression in the medulla oblongata. α-Synuclein load was associated with damage (at 6 weeks post injection) and death (at 12 weeks) of medullary neurons after treatment with only one of the two vector preparations. Of note, neuronal injury and degeneration was accompanied by a substantial reduction of caudo-rostral propagation of human α-synuclein.. Interneuronal α-synuclein transfer, which underlies protein spreading from the medulla oblongata to more rostral brain regions in this rat model, is not a mere consequence of passive release from damaged or dead neurons. Neuronal injury and degeneration did not exacerbate α-synuclein propagation. In fact, data suggest that cell-to-cell passage of α-synuclein may be particularly efficient between intact, relatively healthy neurons.

    Topics: alpha-Synuclein; Animals; Brain; Disease Models, Animal; Female; Gene Transfer Techniques; Humans; Medulla Oblongata; Nerve Degeneration; Neural Pathways; Neurons; Parkinson Disease; Rats; Rats, Sprague-Dawley

2015
Phosphorylated α-synuclein in skin nerve fibres differentiates Parkinson's disease from multiple system atrophy.
    Brain : a journal of neurology, 2015, Volume: 138, Issue:Pt 8

    Deposition of phosphorylated SNCA (also known as α-synuclein) in cutaneous nerve fibres has been shown pre- and post-mortem in Parkinson's disease. Thus far, no pre-mortem studies investigating the presence of phosphorylated SNCA in skin sympathetic nerve fibres of multiple system atrophy, another synucleinopathy, have been conducted. In this in vivo study, skin from the ventral forearm of 10 patients with multiple system atrophy and 10 with Parkinson's disease, together with six control subjects with essential tremor, were examined by immunohistochemistry. Phosphorylated SNCA deposits in skin sympathetic nerve fibres and dermal nerve fibre density were assessed. All patients with Parkinson's disease expressed phosphorylated SNCA in sympathetic skin nerve fibres, correlating with an age-independent denervation of autonomic skin elements. In contrast, no phosphorylated SNCA was found in autonomic skin nerve fibres of patients with multiple system atrophy and essential tremor control subjects. These findings support that phosphorylated SNCA deposition is causative for nerve fibre degeneration in Parkinson's disease. Moreover, pre-mortem investigation of phosphorylated SNCA in cutaneous nerve fibres may prove a relevant and easily conductible diagnostic procedure to differentiate Parkinson's disease from multiple system atrophy.

    Topics: Aged; Aged, 80 and over; alpha-Synuclein; Brain; Female; Humans; Male; Middle Aged; Multiple System Atrophy; Nerve Degeneration; Nerve Fibers; Parkinson Disease; Phosphorylation; Skin

2015
Mutant alpha-synuclein causes age-dependent neuropathology in monkey brain.
    The Journal of neuroscience : the official journal of the Society for Neuroscience, 2015, May-27, Volume: 35, Issue:21

    Parkinson's disease (PD) is an age-dependent neurodegenerative disease that often occurs in those over age 60. Although rodents and small animals have been used widely to model PD and investigate its pathology, their short life span makes it difficult to assess the aging-related pathology that is likely to occur in PD patient brains. Here, we used brain tissues from rhesus monkeys at 2-3, 7-8, and >15 years of age to examine the expression of Parkin, PINK1, and α-synuclein, which are known to cause PD via loss- or gain-of-function mechanisms. We found that α-synuclein is increased in the older monkey brains, whereas Parkin and PINK1 are decreased or remain unchanged. Because of the gain of toxicity of α-synuclein, we performed stereotaxic injection of lentiviral vectors expressing mutant α-synuclein (A53T) into the substantia nigra of monkeys and found that aging also increases the accumulation of A53T in neurites and its associated neuropathology. A53T also causes more extensive reactive astrocytes and axonal degeneration in monkey brain than in mouse brain. Using monkey brain tissues, we found that A53T interacts with neurofascin, an adhesion molecule involved in axon subcellular targeting and neurite outgrowth. Aged monkey brain tissues show an increased interaction of neurofascin with A53T. Overexpression of A53T causes neuritic toxicity in cultured neuronal cells, which can be attenuated by transfected neurofascin. These findings from nonhuman primate brains reveal age-dependent pathological and molecular changes that could contribute to the age-dependent neuropathology in PD.

    Topics: Aging; alpha-Synuclein; Animals; Brain; Haplorhini; Macaca mulatta; Male; Mice; Mice, Inbred C57BL; Mutation; Nerve Degeneration

2015
α-Synuclein Shows High Affinity Interaction with Voltage-dependent Anion Channel, Suggesting Mechanisms of Mitochondrial Regulation and Toxicity in Parkinson Disease.
    The Journal of biological chemistry, 2015, Jul-24, Volume: 290, Issue:30

    Participation of the small, intrinsically disordered protein α-synuclein (α-syn) in Parkinson disease (PD) pathogenesis has been well documented. Although recent research demonstrates the involvement of α-syn in mitochondrial dysfunction in neurodegeneration and suggests direct interaction of α-syn with mitochondria, the molecular mechanism(s) of α-syn toxicity and its effect on neuronal mitochondria remain vague. Here we report that at nanomolar concentrations, α-syn reversibly blocks the voltage-dependent anion channel (VDAC), the major channel of the mitochondrial outer membrane that controls most of the metabolite fluxes in and out of the mitochondria. Detailed analysis of the blockage kinetics of VDAC reconstituted into planar lipid membranes suggests that α-syn is able to translocate through the channel and thus target complexes of the mitochondrial respiratory chain in the inner mitochondrial membrane. Supporting our in vitro experiments, a yeast model of PD shows that α-syn toxicity in yeast depends on VDAC. The functional interactions between VDAC and α-syn, revealed by the present study, point toward the long sought after physiological and pathophysiological roles for monomeric α-syn in PD and in other α-synucleinopathies.

    Topics: alpha-Synuclein; Animals; Gene Expression Regulation; Humans; Lipid Bilayers; Mitochondria; Nerve Degeneration; Parkinson Disease; Protein Binding; Protein Interaction Maps; Rats; Saccharomyces cerevisiae; Voltage-Dependent Anion Channel 1

2015
shRNA targeting α-synuclein prevents neurodegeneration in a Parkinson's disease model.
    The Journal of clinical investigation, 2015, Jul-01, Volume: 125, Issue:7

    Multiple convergent lines of evidence implicate both α-synuclein (encoded by SCNA) and mitochondrial dysfunction in the pathogenesis of sporadic Parkinson's disease (PD). Occupational exposure to the mitochondrial complex I inhibitor rotenone increases PD risk; rotenone-exposed rats show systemic mitochondrial defects but develop specific neuropathology, including α-synuclein aggregation and degeneration of substantia nigra dopaminergic neurons. Here, we inhibited expression of endogenous α-synuclein in the adult rat substantia nigra by adeno-associated virus-mediated delivery of a short hairpin RNA (shRNA) targeting the endogenous rat Snca transcript. Knockdown of α-synuclein by ~35% did not affect motor function or cause degeneration of nigral dopaminergic neurons in control rats. However, in rotenone-exposed rats, progressive motor deficits were substantially attenuated contralateral to α-synuclein knockdown. Correspondingly, rotenone-induced degeneration of nigral dopaminergic neurons, their dendrites, and their striatal terminals was decreased ipsilateral to α-synuclein knockdown. These data show that α-synuclein knockdown is neuroprotective in the rotenone model of PD and indicate that endogenous α-synuclein contributes to the specific vulnerability of dopaminergic neurons to systemic mitochondrial inhibition. Our findings are consistent with a model in which genetic variants influencing α-synuclein expression modulate cellular susceptibility to environmental exposures in PD patients. shRNA targeting the SNCA transcript should be further evaluated as a possible neuroprotective therapy in PD.

    Topics: alpha-Synuclein; Animals; Disease Models, Animal; Dopaminergic Neurons; Gene Knockdown Techniques; Male; Nerve Degeneration; Parkinsonian Disorders; Rats; Rats, Inbred Lew; Rats, Transgenic; RNA, Messenger; RNA, Small Interfering; Rotenone; Substantia Nigra

2015
Intrastriatal injection of pre-formed mouse α-synuclein fibrils into rats triggers α-synuclein pathology and bilateral nigrostriatal degeneration.
    Neurobiology of disease, 2015, Volume: 82

    Previous studies demonstrate that intrastriatal injections of fibrillar alpha-synuclein (α-syn) into mice induce Parkinson's disease (PD)-like Lewy body (LB) pathology formed by aggregated α-syn in anatomically interconnected regions and significant nigrostriatal degeneration. The aim of the current study was to evaluate whether exogenous mouse α-syn pre-formed fibrils (PFF) injected into the striatum of rats would result in accumulation of LB-like intracellular inclusions and nigrostriatal degeneration. Sprague-Dawley rats received unilateral intrastriatal injections of either non-fibrillized recombinant α-syn or PFF mouse α-syn in 1- or 2- sites and were euthanized at 30, 60 or 180 days post-injection (pi). Both non-fibrillized recombinant α-syn and PFF α-syn injections resulted in phosphorylated α-syn intraneuronal accumulations (i.e., diffuse Lewy neurite (LN)- and LB-like inclusions) with significantly greater accumulations following PFF injection. LB-like inclusions were observed in several areas that innervate the striatum, most prominently the frontal and insular cortices, the amygdala, and the substantia nigra pars compacta (SNpc). α-Syn accumulations co-localized with ubiquitin, p62, and were thioflavin-S-positive and proteinase-k resistant, suggesting that PFF-induced pathology exhibits properties similar to human LBs. Although α-syn inclusions within the SNpc remained ipsilateral to striatal injection, we observed bilateral reductions in nigral dopamine neurons at the 180-day time-point in both the 1- and 2-site PFF injection paradigms. PFF injected rats exhibited bilateral reductions in striatal dopaminergic innervation at 60 and 180 days and bilateral decreases in homovanillic acid; however, dopamine reduction was observed only in the striatum ipsilateral to PFF injection. Although the level of dopamine asymmetry in PFF injected rats at 180 days was insufficient to elicit motor deficits in amphetamine-induced rotations or forelimb use in the cylinder task, significant disruption of ultrasonic vocalizations was observed. Taken together, our findings demonstrate that α-syn PFF are sufficient to seed the pathological conversion and propagation of endogenous α-syn to induce a progressive, neurodegenerative model of α-synucleinopathy in rats.

    Topics: alpha-Synuclein; Animals; Corpus Striatum; Disease Models, Animal; Dopaminergic Neurons; Male; Mice; Motor Activity; Nerve Degeneration; Rats; Rats, Sprague-Dawley; Substantia Nigra; Vocalization, Animal

2015
Nigral overexpression of alpha-synuclein in the absence of parkin enhances alpha-synuclein phosphorylation but does not modulate dopaminergic neurodegeneration.
    Molecular neurodegeneration, 2015, 06-23, Volume: 10

    Alpha-synuclein is a key protein in the pathogenesis of Parkinson's disease. Mutations in the parkin gene are the most common cause of early-onset autosomal recessive Parkinson's disease, probably through a loss-of-function mechanism. However, the molecular mechanism by which loss of parkin function leads to the development of the disease and the role of alpha-synuclein in parkin-associated Parkinson's disease is still not elucidated. Conflicting results were reported about the effect of the absence of parkin on alpha-synuclein-mediated neurotoxicity using a transgenic approach. In this study, we investigated the effect of loss of parkin on alpha-synuclein neuropathology and toxicity in adult rodent brain using viral vectors. Therefore, we overexpressed human wild type alpha-synuclein in the substantia nigra of parkin knockout and wild type mice using two different doses of recombinant adeno-associated viral vectors.. No difference was observed in nigral dopaminergic cell loss between the parkin knockout mice and wild type mice up to 16 weeks after viral vector injection. However, the level of alpha-synuclein phosphorylated at serine residue 129 in the substantia nigra was significantly increased in the parkin knockout mice compared to the wild type mice while the total expression level of alpha-synuclein was similar in both groups. The increased alpha-synuclein phosphorylation was confirmed in a parkin knockdown cell line.. These findings support a functional relationship between parkin and alpha-synuclein phosphorylation in rodent brain.

    Topics: alpha-Synuclein; Animals; Cell Count; Cell Line, Tumor; Dependovirus; Dopaminergic Neurons; Female; Genes, Reporter; Genetic Vectors; Humans; Lentivirus; Male; Mice; Mice, Inbred C57BL; Mice, Knockout; Mice, Transgenic; MicroRNAs; Nerve Degeneration; Neuroblastoma; Phosphorylation; Phosphoserine; Protein Processing, Post-Translational; Substantia Nigra; Ubiquitin-Protein Ligases

2015
Alpha-Synuclein affects neurite morphology, autophagy, vesicle transport and axonal degeneration in CNS neurons.
    Cell death & disease, 2015, Jul-09, Volume: 6

    Many neuropathological and experimental studies suggest that the degeneration of dopaminergic terminals and axons precedes the demise of dopaminergic neurons in the substantia nigra, which finally results in the clinical symptoms of Parkinson disease (PD). The mechanisms underlying this early axonal degeneration are, however, still poorly understood. Here, we examined the effects of overexpression of human wildtype alpha-synuclein (αSyn-WT), a protein associated with PD, and its mutant variants αSyn-A30P and -A53T on neurite morphology and functional parameters in rat primary midbrain neurons (PMN). Moreover, axonal degeneration after overexpression of αSyn-WT and -A30P was analyzed by live imaging in the rat optic nerve in vivo. We found that overexpression of αSyn-WT and of its mutants A30P and A53T impaired neurite outgrowth of PMN and affected neurite branching assessed by Sholl analysis in a variant-dependent manner. Surprisingly, the number of primary neurites per neuron was increased in neurons transfected with αSyn. Axonal vesicle transport was examined by live imaging of PMN co-transfected with EGFP-labeled synaptophysin. Overexpression of all αSyn variants significantly decreased the number of motile vesicles and decelerated vesicle transport compared with control. Macroautophagic flux in PMN was enhanced by αSyn-WT and -A53T but not by αSyn-A30P. Correspondingly, colocalization of αSyn and the autophagy marker LC3 was reduced for αSyn-A30P compared with the other αSyn variants. The number of mitochondria colocalizing with LC3 as a marker for mitophagy did not differ among the groups. In the rat optic nerve, both αSyn-WT and -A30P accelerated kinetics of acute axonal degeneration following crush lesion as analyzed by in vivo live imaging. We conclude that αSyn overexpression impairs neurite outgrowth and augments axonal degeneration, whereas axonal vesicle transport and autophagy are severely altered.

    Topics: alpha-Synuclein; Amino Acid Substitution; Animals; Autophagy; Axons; Dopamine; Dopaminergic Neurons; Gene Expression Regulation; Humans; Nerve Degeneration; Neurites; Parkinson Disease; Rats; Substantia Nigra

2015
LK6/Mnk2a is a new kinase of alpha synuclein phosphorylation mediating neurodegeneration.
    Scientific reports, 2015, Jul-29, Volume: 5

    Parkinson's disease (PD) is a movement disorder due to the loss of dopaminergic (DA) neurons in the substantia nigra. Alpha-synuclein phosphorylation and α-synuclein inclusion (Lewy body) become a main contributor, but little is known about their formation mechanism. Here we used protein expression profiling of PD to construct a model of their signalling network from drsophila to human and nominate major nodes that regulate PD development. We found in this network that LK6, a serine/threonine protein kinase, plays a key role in promoting α-synuclein Ser129 phosphorylation by identification of LK6 knockout and overexpression. In vivo test was further confirmed that LK6 indeed enhances α-synuclein phosphorylation, accelerates the death of dopaminergic neurons, reduces the climbing ability and shortens the the life span of drosophila. Further, MAP kinase-interacting kinase 2a (Mnk2a), a human homolog of LK6, also been shown to make α-synuclein phosphorylation and leads to α-synuclein inclusion formation. On the mechanism, the phosphorylation mediated by LK6 and Mnk2a is controlled through ERK signal pathway by phorbolmyristate acetate (PMA) avtivation and PD98059 inhibition. Our findings establish pivotal role of Lk6 and Mnk2a in unprecedented signalling networks, may lead to new therapies preventing α-synuclein inclusion formation and neurodegeneration.

    Topics: alpha-Synuclein; Animals; Disease Models, Animal; Dopaminergic Neurons; Drosophila; Drosophila Proteins; Female; Male; MAP Kinase Signaling System; Mitogen-Activated Protein Kinase Kinases; Nerve Degeneration; Parkinson Disease; Phosphorylation; Phosphotransferases; Signal Transduction; Substantia Nigra

2015
Hot, Hotter, and Hottest Trends in α-Synuclein Research.
    Current protein & peptide science, 2015, Volume: 16, Issue:8

    Topics: alpha-Synuclein; Biomedical Research; Humans; Nerve Degeneration; Prions; Protein Aggregation, Pathological

2015
Acetylcorynoline attenuates dopaminergic neuron degeneration and α-synuclein aggregation in animal models of Parkinson's disease.
    Neuropharmacology, 2014, Volume: 82

    Parkinson's disease (PD), the second most common neurodegenerative disease, impairs motor skills and cognitive function. To date, the drugs used for PD treatment provide only symptomatic relief. The identification of new drugs that show benefit in slowing the decline seen in PD patients is the focus of much current research. Acetylcorynoline is the major alkaloid component derived from Corydalis bungeana, a traditional Chinese medical herb. It has been shown to have anti-inflammatory properties, but no studies have yet described the effects of acetylcorynoline on PD. The aim of this study was to evaluate the potential for acetylcorynoline to improve PD in Caenorhabditis elegans models. In the present study, we used a pharmacological strain (BZ555) that expresses green fluorescent protein specifically in dopaminergic neurons, and a transgenic strain (OW13) that expresses human α-synuclein in muscle cells to study the antiparkinsonian effects of acetylcorynoline. Our experimental data showed that treatment with up to 10 mM acetylcorynoline does not cause toxicity in animals. Acetylcorynoline significantly decreases dopaminergic neuron degeneration induced by 6-hydroxydopamine in BZ555 strain; prevents α-synuclein aggregation; recovers lipid content in OW13 strain; restores food-sensing behavior, and dopamine levels; and prolongs life-span in 6-hydroxydopamine-treated N2 strain, thus showing its potential as a possible antiparkinsonian drug. Acetylcorynoline may exert its effects by decreasing egl-1 expression to suppress apoptosis pathways and by increasing rpn5 expression to enhance the activity of proteasomes.

    Topics: alpha-Synuclein; Animals; Animals, Genetically Modified; Antiparkinson Agents; Appetitive Behavior; Berberine Alkaloids; Caenorhabditis elegans; Caenorhabditis elegans Proteins; Dopamine; Dopaminergic Neurons; Green Fluorescent Proteins; Humans; Molecular Structure; Muscle Cells; Nerve Degeneration; Oxidopamine; Parkinsonian Disorders; Repressor Proteins

2014
α-Synuclein A30P decreases neurodegeneration and increases synaptic vesicle release probability in CSPα-null mice.
    Neuropharmacology, 2014, Volume: 76 Pt A

    α-Synuclein and Cysteine-string protein-α (CSPα) are presynaptic proteins that participate in the maintenance of synaptic function. Mutations or overexpression of the wild type form of α-synuclein have been related to Parkinson's disease, and CSPα mutations cause one type of neuronal ceroid lipofuscinosis. Both are adult-onset neurodegenerative diseases characterized by neuronal protein aggregations. Strikingly, while in mouse the lack of CSPα produces defective neurotransmission and neurodegeneration of motor terminals, blindness and early lethality, the moderate overexpression of wild-type α-synuclein fully rescues the CSPα-null phenotype. Contrarily, the overexpression of the mutated human α-synuclein A30P (α-synuclein(hA30P)) has much less effect in CSPα KO mice. To explore how the A30P mutation affects the neuroprotective function of α-synuclein we investigated synaptic structure and neurotransmission in motor nerve terminals of wild-type and CSPα-null mice transgenic for α-synuclein(hA30P). We found that although α-synuclein(hA30P) did not fully prevent neurodegeneration, it significantly improved synaptic organization and function in CSPα-null mice by enhancing quantal content, release probability, synaptic vesicle content, active zone number, postsynaptic area, and microtubule appearance. These results demonstrate that α-synuclein(hA30P) is able to ameliorate synapse degeneration, despite its apparent lack of functionality and its long-term pathogenic effects in neurons. These findings may help to understand better the dual function of α-synuclein regarding neurodegeneration. This article is part of the Special Issue entitled 'The Synaptic Basis of Neurodegenerative Disorders'.

    Topics: alpha-Synuclein; Animals; Excitatory Postsynaptic Potentials; Gene Deletion; Gene Expression; HSP40 Heat-Shock Proteins; Humans; In Vitro Techniques; Membrane Proteins; Mice; Mice, Knockout; Mice, Transgenic; Motor Neurons; Nerve Degeneration; Neuromuscular Junction; Neuroprotective Agents; Synaptic Transmission; Synaptic Vesicles

2014
Lewy body extracts from Parkinson disease brains trigger α-synuclein pathology and neurodegeneration in mice and monkeys.
    Annals of neurology, 2014, Volume: 75, Issue:3

    Mounting evidence suggests that α-synuclein, a major protein component of Lewy bodies (LB), may be responsible for initiating and spreading the pathological process in Parkinson disease (PD). Supporting this concept, intracerebral inoculation of synthetic recombinant α-synuclein fibrils can trigger α-synuclein pathology in mice. However, it remains uncertain whether the pathogenic effects of recombinant synthetic α-synuclein may apply to PD-linked pathological α-synuclein and occur in species closer to humans.. Nigral LB-enriched fractions containing pathological α-synuclein were purified from postmortem PD brains by sucrose gradient fractionation and subsequently inoculated into the substantia nigra or striatum of wild-type mice and macaque monkeys. Control animals received non-LB fractions containing soluble α-synuclein derived from the same nigral PD tissue.. In both mice and monkeys, intranigral or intrastriatal inoculations of PD-derived LB extracts resulted in progressive nigrostriatal neurodegeneration starting at striatal dopaminergic terminals. No neurodegeneration was observed in animals receiving non-LB fractions from the same patients. In LB-injected animals, exogenous human α-synuclein was quickly internalized within host neurons and triggered the pathological conversion of endogenous α-synuclein. At the onset of LB-induced degeneration, host pathological α-synuclein diffusely accumulated within nigral neurons and anatomically interconnected regions, both anterogradely and retrogradely. LB-induced pathogenic effects required both human α-synuclein present in LB extracts and host expression of α-synuclein.. α-Synuclein species contained in PD-derived LB are pathogenic and have the capacity to initiate a PD-like pathological process, including intracellular and presynaptic accumulations of pathological α-synuclein in different brain areas and slowly progressive axon-initiated dopaminergic nigrostriatal neurodegeneration.

    Topics: alpha-Synuclein; Animals; Corpus Striatum; Dopaminergic Neurons; Female; Humans; Lewy Bodies; Macaca mulatta; Mice; Mice, Knockout; Microinjections; Nerve Degeneration; Parkinson Disease; Substantia Nigra; Tissue Extracts

2014
Enhanced ubiquitin-dependent degradation by Nedd4 protects against α-synuclein accumulation and toxicity in animal models of Parkinson's disease.
    Neurobiology of disease, 2014, Volume: 64

    Parkinson's disease is a neurodegenerative disorder, characterized by accumulation and misfolding of α-synuclein. Although the level of α-synuclein in neurons is fundamentally linked to the onset of neurodegeneration, multiple pathways have been implicated in its degradation, and it remains unclear which are the critical ubiquitination enzymes that protect against α-synuclein accumulation in vivo. The ubiquitin ligase Nedd4 targets α-synuclein to the endosomal-lysosomal pathway in cultured cells. Here we asked whether Nedd4-mediated degradation protects against α-synuclein-induced toxicity in the Drosophila and rodent models of Parkinson's disease. We show that overexpression of Nedd4 can rescue the degenerative phenotype from ectopic expression of α-synuclein in the Drosophila eye. Overexpressed Nedd4 in the Drosophila brain prevented the α-synuclein-induced locomotor defect whereas reduction in endogenous Nedd4 by RNAi led to worsening motor function and increased loss of dopaminergic neurons. Accordingly, AAV-mediated expression of wild-type but not the catalytically inactive Nedd4 decreased the α-synuclein-induced dopaminergic cell loss in the rat substantia nigra and reduced α-synuclein accumulation. Collectively, our data in two evolutionarily distant model organisms strongly suggest that Nedd4 is a modifier of α-synuclein pathobiology and thus a potential target for neuroprotective therapies.

    Topics: alpha-Synuclein; Animals; Animals, Genetically Modified; Brain; Dopaminergic Neurons; Drosophila; Endosomal Sorting Complexes Required for Transport; Eye; Female; Humans; Locomotion; Male; Mutation; Nedd4 Ubiquitin Protein Ligases; Nerve Degeneration; Parkinsonian Disorders; Rats; Rats, Sprague-Dawley; RNA, Messenger; Substantia Nigra; Ubiquitin-Protein Ligases

2014
Next-generation active immunization approach for synucleinopathies: implications for Parkinson's disease clinical trials.
    Acta neuropathologica, 2014, Volume: 127, Issue:6

    Immunotherapeutic approaches are currently in the spotlight for their potential as disease-modifying treatments for neurodegenerative disorders. The discovery that α-synuclein (α-syn) can transmit from cell to cell in a prion-like fashion suggests that immunization might be a viable option for the treatment of synucleinopathies. This possibility has been bolstered by the development of next-generation active vaccination technology with short peptides-AFFITOPEs(®) (AFF)- that do not elicit an α-syn-specific T cell response. This approach allows for the production of long term, sustained, more specific, non-cross reacting antibodies suitable for the treatment of synucleinopathies, such as Parkinson's disease (PD). In this context, we screened a large library of peptides that mimic the C-terminus region of α-syn and discovered a novel set of AFF that identified α-syn oligomers. Next, the peptide that elicited the most specific response against α-syn (AFF 1) was selected for immunizing two different transgenic (tg) mouse models of PD and Dementia with Lewy bodies, the PDGF- and the mThy1-α-syn tg mice. Vaccination with AFF 1 resulted in high antibody titers in CSF and plasma, which crossed into the CNS and recognized α-syn aggregates. Active vaccination with AFF 1 resulted in decreased accumulation of α-syn oligomers in axons and synapses, accompanied by reduced degeneration of TH fibers in the caudo-putamen nucleus and by improvements in motor and memory deficits in both in vivo models. Clearance of α-syn involved activation of microglia and increased anti-inflammatory cytokine expression, further supporting the efficacy of this novel active vaccination approach for synucleinopathies.

    Topics: alpha-Synuclein; Animals; Antibodies; Axons; Caudate Nucleus; Clinical Trials as Topic; Disease Models, Animal; Humans; Lewy Body Disease; Memory Disorders; Mice, Inbred C57BL; Mice, Transgenic; Microglia; Motor Activity; Nerve Degeneration; Parkinson Disease; Putamen; Synapses; T-Lymphocytes; Vaccination

2014
Autophagy-lysosome pathway associated neuropathology and axonal degeneration in the brains of alpha-galactosidase A-deficient mice.
    Acta neuropathologica communications, 2014, Feb-14, Volume: 2

    Mutations in the gene for alpha-galactosidase A result in Fabry disease, a rare, X-linked lysosomal storage disorder characterized by a loss of alpha-galactosidase A enzymatic activity. The resultant accumulation of glycosphingolipids throughout the body leads to widespread vasculopathy with particular detriment to the kidneys, heart and nervous system. Disruption in the autophagy-lysosome pathway has been documented previously in Fabry disease but its relative contribution to nervous system pathology in Fabry disease is unknown. Using an experimental mouse model of Fabry disease, alpha-galactosidase A deficiency, we examined brain pathology in 20-24 month old mice with particular emphasis on the autophagy-lysosome pathway.. Alpha-galactosidase A-deficient mouse brains exhibited enhanced punctate perinuclear immunoreactivity for the autophagy marker microtubule-associated protein light-chain 3 (LC3) in the parenchyma of several brain regions, as well as enhanced parenchymal and vascular immunoreactivity for lysosome-associated membrane protein-1 (LAMP-1). Ultrastructural analysis revealed endothelial cell inclusions with electron densities and a pronounced accumulation of electron-dense lipopigment. The pons of alpha-galactosidase A-deficient mice in particular exhibited a striking neuropathological phenotype, including the presence of large, swollen axonal spheroids indicating axonal degeneration, in addition to large interstitial aggregates positive for phosphorylated alpha-synuclein that co-localized with the axonal spheroids. Double-label immunofluorescence revealed co-localization of phosphorylated alpha-synuclein aggregates with ubiquitin and LC3.. Together these findings indicate widespread neuropathology and focused axonal neurodegeneration in alpha-galactosidase A-deficient mouse brain in association with disruption of the autophagy-lysosome pathway, and provide the basis for future mechanistic assessment of the contribution of the autophagy-lysosome pathway to this histologic phenotype.

    Topics: alpha-Galactosidase; alpha-Synuclein; Animals; Autophagy; Brain; Disease Models, Animal; Fabry Disease; Gene Expression Regulation; Inclusion Bodies; Lysosomal Membrane Proteins; Lysosomes; Mice; Mice, Knockout; Microscopy, Electron; Microtubule-Associated Proteins; Nerve Degeneration; Optical Imaging; Signal Transduction

2014
Targeting the intrinsically disordered structural ensemble of α-synuclein by small molecules as a potential therapeutic strategy for Parkinson's disease.
    PloS one, 2014, Volume: 9, Issue:2

    The misfolding of intrinsically disordered proteins such as α-synuclein, tau and the Aβ peptide has been associated with many highly debilitating neurodegenerative syndromes including Parkinson's and Alzheimer's diseases. Therapeutic targeting of the monomeric state of such intrinsically disordered proteins by small molecules has, however, been a major challenge because of their heterogeneous conformational properties. We show here that a combination of computational and experimental techniques has led to the identification of a drug-like phenyl-sulfonamide compound (ELN484228), that targets α-synuclein, a key protein in Parkinson's disease. We found that this compound has substantial biological activity in cellular models of α-synuclein-mediated dysfunction, including rescue of α-synuclein-induced disruption of vesicle trafficking and dopaminergic neuronal loss and neurite retraction most likely by reducing the amount of α-synuclein targeted to sites of vesicle mobilization such as the synapse in neurons or the site of bead engulfment in microglial cells. These results indicate that targeting α-synuclein by small molecules represents a promising approach to the development of therapeutic treatments of Parkinson's disease and related conditions.

    Topics: alpha-Synuclein; Animals; Binding Sites; Dopaminergic Neurons; Humans; Intrinsically Disordered Proteins; Mice; Models, Biological; Models, Molecular; Molecular Targeted Therapy; Nerve Degeneration; Parkinson Disease; Phagocytes; Small Molecule Libraries; Synapses

2014
Axon degeneration and PGC-1α-mediated protection in a zebrafish model of α-synuclein toxicity.
    Disease models & mechanisms, 2014, Volume: 7, Issue:5

    α-synuclein (aSyn) expression is implicated in neurodegenerative processes, including Parkinson's disease (PD) and dementia with Lewy bodies (DLB). In animal models of these diseases, axon pathology often precedes cell death, raising the question of whether aSyn has compartment-specific toxic effects that could require early and/or independent therapeutic intervention. The relevance of axonal pathology to degeneration can only be addressed through longitudinal, in vivo monitoring of different neuronal compartments. With current imaging methods, dopaminergic neurons do not readily lend themselves to such a task in any vertebrate system. We therefore expressed human wild-type aSyn in zebrafish peripheral sensory neurons, which project elaborate superficial axons that can be continuously imaged in vivo. Axonal outgrowth was normal in these neurons but, by 2 days post-fertilization (dpf), many aSyn-expressing axons became dystrophic, with focal varicosities or diffuse beading. Approximately 20% of aSyn-expressing cells died by 3 dpf. Time-lapse imaging revealed that focal axonal swelling, but not overt fragmentation, usually preceded cell death. Co-expressing aSyn with a mitochondrial reporter revealed deficits in mitochondrial transport and morphology even when axons appeared overtly normal. The axon-protective protein Wallerian degeneration slow (WldS) delayed axon degeneration but not cell death caused by aSyn. By contrast, the transcriptional coactivator PGC-1α, which has roles in the regulation of mitochondrial biogenesis and reactive-oxygen-species detoxification, abrogated aSyn toxicity in both the axon and the cell body. The rapid onset of axonal pathology in this system, and the relatively moderate degree of cell death, provide a new model for the study of aSyn toxicity and protection. Moreover, the accessibility of peripheral sensory axons will allow effects of aSyn to be studied in different neuronal compartments and might have utility in screening for novel disease-modifying compounds.

    Topics: alpha-Synuclein; Animals; Axons; Cell Death; Disease Models, Animal; Fertilization; Humans; Larva; Mitochondria; Nerve Degeneration; Protein Transport; Sensory Receptor Cells; Transcription Factors; Wallerian Degeneration; Zebrafish; Zebrafish Proteins

2014
Accumulation of oligomer-prone α-synuclein exacerbates synaptic and neuronal degeneration in vivo.
    Brain : a journal of neurology, 2014, Volume: 137, Issue:Pt 5

    In Parkinson's disease and dementia with Lewy bodies, α-synuclein aggregates to form oligomers and fibrils; however, the precise nature of the toxic α-synuclein species remains unclear. A number of synthetic α-synuclein mutations were recently created (E57K and E35K) that produce species of α-synuclein that preferentially form oligomers and increase α-synuclein-mediated toxicity. We have shown that acute lentiviral expression of α-synuclein E57K leads to the degeneration of dopaminergic neurons; however, the effects of chronic expression of oligomer-prone α-synuclein in synapses throughout the brain have not been investigated. Such a study could provide insight into the possible mechanism(s) through which accumulation of α-synuclein oligomers in the synapse leads to neurodegeneration. For this purpose, we compared the patterns of neurodegeneration and synaptic damage between a newly generated mThy-1 α-synuclein E57K transgenic mouse model that is prone to forming oligomers and the mThy-1 α-synuclein wild-type mouse model (Line 61), which accumulates various forms of α-synuclein. Three lines of α-synuclein E57K (Lines 9, 16 and 54) were generated and compared with the wild-type. The α-synuclein E57K Lines 9 and 16 were higher expressings of α-synuclein, similar to α-synuclein wild-type Line 61, and Line 54 was a low expressing of α-synuclein compared to Line 61. By immunoblot analysis, the higher-expressing α-synuclein E57K transgenic mice showed abundant oligomeric, but not fibrillar, α-synuclein whereas lower-expressing mice accumulated monomeric α-synuclein. Monomers, oligomers, and fibrils were present in α-synuclein wild-type Line 61. Immunohistochemical and ultrastructural analyses demonstrated that α-synuclein accumulated in the synapses but not in the neuronal cells bodies, which was different from the α-synuclein wild-type Line 61, which accumulates α-synuclein in the soma. Compared to non-transgenic and lower-expressing mice, the higher-expressing α-synuclein E57K mice displayed synaptic and dendritic loss, reduced levels of synapsin 1 and synaptic vesicles, and behavioural deficits. Similar alterations, but to a lesser extent, were seen in the α-synuclein wild-type mice. Moreover, although the oligomer-prone α-synuclein mice displayed neurodegeneration in the frontal cortex and hippocampus, the α-synuclein wild-type only displayed neuronal loss in the hippocampus. These results support the hypothesis that accumulating oligomeric α-synuclein may

    Topics: alpha-Synuclein; Alzheimer Disease; Animals; Brain; Disease Models, Animal; Gene Expression Regulation; Glutamic Acid; Humans; Lewy Body Disease; Lysine; Memory Disorders; Mice; Mice, Inbred C57BL; Mice, Transgenic; Mutation; Nerve Degeneration; Nerve Tissue Proteins; Neurons; Synapses; Thy-1 Antigens

2014
Cutaneous neuropathy in Parkinson's disease: a window into brain pathology.
    Acta neuropathologica, 2014, Volume: 128, Issue:1

    The deposition of alpha-synuclein in the brain, the neuropathological hallmark of Parkinson's disease (PD), follows a distinct anatomical and temporal sequence. This study aimed to characterize alpha-synuclein deposition in cutaneous nerves from patients with PD. We further strived to explore whether peripheral nerve involvement is intrinsic to PD and reflective of known features of brain pathology, which could render it a useful tool for pathogenetic studies and pre-mortem histological diagnosis of PD. We obtained skin biopsies from the distal and proximal leg, back and finger of 31 PD patients and 35 controls and quantified the colocalization of phosphorylated alpha-synuclein in somatosensory and autonomic nerve fibers and the pattern of loss of different subtypes of dermal fibers. Deposits of phosphorylated alpha-synuclein were identified in 16/31 PD patients but in 0/35 controls (p < 0.0001). Quantification of nerve fibers revealed two types of peripheral neurodegeneration in PD: (1) a length-dependent reduction of intraepidermal small nerve fibers (p < 0.05) and (2) a severe non-length-dependent reduction of substance P-immunoreactive intraepidermal nerve fibers (p < 0.0001). The latter coincided with a more pronounced proximal manifestation of alpha-synuclein pathology in the skin. The histological changes did not correlate with markers of levodopa toxicity such as vitamin B12 deficiency. Our findings suggest that loss of peripheral nerve fibers is an intrinsic feature of PD and that peripheral nerve changes may reflect the two types of central alpha-synuclein-related PD pathology, namely neuronal death and axonal degeneration. Detection of phosphorylated alpha-synuclein in dermal nerve fibers might be a useful diagnostic test for PD with high specificity but low sensitivity.

    Topics: Adult; Aged; Aged, 80 and over; alpha-Synuclein; Autonomic Pathways; Back; Brain; Female; Fingers; Humans; Leg; Male; Middle Aged; Nerve Degeneration; Neural Conduction; Parkinson Disease; Peripheral Nervous System Diseases; Phosphorylation; Skin; Substance P; Vitamins

2014
Aldehyde dehydrogenase 1 defines and protects a nigrostriatal dopaminergic neuron subpopulation.
    The Journal of clinical investigation, 2014, Volume: 124, Issue:7

    Subpopulations of dopaminergic (DA) neurons within the substantia nigra pars compacta (SNpc) display a differential vulnerability to loss in Parkinson's disease (PD); however, it is not clear why these subsets are preferentially selected in PD-associated neurodegeneration. In rodent SNpc, DA neurons can be divided into two subpopulations based on the expression of aldehyde dehydrogenase 1 (ALDH1A1). Here, we have shown that, in α-synuclein transgenic mice, a murine model of PD-related disease, DA neurodegeneration occurs mainly in a dorsomedial ALDH1A1-negative subpopulation that is also prone to cytotoxic aggregation of α-synuclein. Notably, the topographic ALDH1A1 pattern observed in α-synuclein transgenic mice was conserved in human SNpc. Postmortem evaluation of brains of patients with PD revealed a severe reduction of ALDH1A1 expression and neurodegeneration in the ventral ALDH1A1-positive DA subpopulations. ALDH1A1 expression was also suppressed in α-synuclein transgenic mice. Deletion of Aldh1a1 exacerbated α-synuclein-mediated DA neurodegeneration and α-synuclein aggregation, whereas Aldh1a1-null and control DA neurons were comparably susceptible to 1-methyl-4-phenylpyridinium-, glutamate-, or camptothecin-induced cell death. ALDH1A1 overexpression appeared to preferentially protect against α-synuclein-mediated DA neurodegeneration but did not rescue α-synuclein-induced loss of cortical neurons. Together, our findings suggest that ALDH1A1 protects subpopulations of SNpc DA neurons by preventing the accumulation of dopamine aldehyde intermediates and formation of cytotoxic α-synuclein oligomers.

    Topics: Aldehyde Dehydrogenase; Aldehyde Dehydrogenase 1 Family; alpha-Synuclein; Animals; Disease Models, Animal; Dopaminergic Neurons; Female; Humans; Male; Mice; Mice, Knockout; Mice, Mutant Strains; Mice, Transgenic; Mutant Proteins; Mutation, Missense; Nerve Degeneration; Parkinson Disease; Protein Multimerization; Recombinant Proteins; Retinal Dehydrogenase; Substantia Nigra

2014
The prion hypothesis of Parkinson's disease: this hot topic just got hotter.
    Movement disorders : official journal of the Movement Disorder Society, 2014, Volume: 29, Issue:8

    Topics: alpha-Synuclein; Animals; Dopaminergic Neurons; Female; Humans; Lewy Bodies; Nerve Degeneration; Parkinson Disease; Tissue Extracts

2014
ESCRT-mediated uptake and degradation of brain-targeted α-synuclein single chain antibody attenuates neuronal degeneration in vivo.
    Molecular therapy : the journal of the American Society of Gene Therapy, 2014, Volume: 22, Issue:10

    Parkinson's disease and dementia with Lewy bodies are neurodegenerative disorders characterized by accumulation of α-synuclein (α-syn). Recently, single-chain fragment variables (scFVs) have been developed against individual conformational species of α-syn. Unlike more traditional monoclonal antibodies, these scFVs will not activate or be endocytosed by Fc receptors. For this study, we investigated an scFV directed against oligomeric α-syn fused to the LDL receptor-binding domain from apolipoprotein B (apoB). The modified scFV showed enhanced brain penetration and was imported into neuronal cells through the endosomal sorting complex required for transport (ESCRT) pathway, leading to lysosomal degradation of α-syn aggregates. Further analysis showed that the scFV was effective at ameliorating neurodegenerative pathology and behavioral deficits observed in the mouse model of dementia with Lewy bodies/Parkinson's disease. Thus, the apoB modification had the effect of both increasing accumulation of the scFV in the brain and directing scFV/α-syn complexes for degradation through the ESCRT pathway, leading to improved therapeutic potential of immunotherapy.

    Topics: alpha-Synuclein; Amino Acid Motifs; Animals; Apolipoproteins B; Autophagy; Behavior, Animal; Brain; Cell Line; Endosomal Sorting Complexes Required for Transport; Gene Order; Genetic Vectors; Lentivirus; Mice; Mice, Transgenic; Nerve Degeneration; Nerve Tissue Proteins; Neuroglia; Neurons; Protein Binding; Protein Transport; Proteolysis; Rats; Single-Chain Antibodies; Transduction, Genetic

2014
S-Nitrosylating protein disulphide isomerase mediates α-synuclein aggregation caused by methamphetamine exposure in PC12 cells.
    Toxicology letters, 2014, Oct-01, Volume: 230, Issue:1

    Methamphetamine (METH) belongs to Amphetamine-type stimulants, METH abusers are at high risk of neurodegenerative disorders, including Parkinson's disease (PD). However, there are still no effective treatments to METH-induced neurodegeneration because its mechanism remains unknown. In order to investigate METH's neurotoxic mechanism, we established an in vitro PD pathology model by exposing PC12 cells to METH. We found the expression of nitric oxide synthase (NOS), nitric oxide (NO) and α-synuclein (α-syn) was significantly increased after METH treatment for 24h, in addition, the aggregattion of α-syn and the S-nitrosylation of protein disulphideisomerase(PDI) were also obviously enhanced. When we exposed PC12 cells to the NOS inhibitor N-nitro-L-arginine(L-NNA) with METH together, the L-NNA obviously inhibited these changes induced by METH. While when we exposed PC12 cells to the precursor of NO L-Arginine together with METH, the L-Arginine resulted in the opposite effect compared to L-NNA. And when we knocked down the PDI gene, the L-NNA did not have this effect. Therefore, PDI plays a significant role in neurological disorders related to α-syn aggregation, and it suggests that PDI could be as a potential target to prevent METH-induced neurodegeneration.

    Topics: alpha-Synuclein; Animals; Apoptosis; Cell Survival; Central Nervous System Stimulants; Enzyme Inhibitors; Gene Knockdown Techniques; Methamphetamine; Nerve Degeneration; Neurons; Neurotoxicity Syndromes; Nitric Oxide; Nitric Oxide Synthase; Nitroarginine; PC12 Cells; Protein Disulfide-Isomerases; Protein Processing, Post-Translational; Rats; RNA Interference; Time Factors; Transfection

2014
Cardiac sympathetic denervation in 6-OHDA-treated nonhuman primates.
    PloS one, 2014, Volume: 9, Issue:8

    Cardiac sympathetic neurodegeneration and dysautonomia affect patients with sporadic and familial Parkinson's disease (PD) and are currently proposed as prodromal signs of PD. We have recently developed a nonhuman primate model of cardiac dysautonomia by iv 6-hydroxydopamine (6-OHDA). Our in vivo findings included decreased cardiac uptake of a sympathetic radioligand and circulating catecholamines; here we report the postmortem characterization of the model. Ten adult rhesus monkeys (5-17 yrs old) were used in this study. Five animals received 6-OHDA (50 mg/kg i.v.) and five were age-matched controls. Three months post-neurotoxin the animals were euthanized; hearts and adrenal glands were processed for immunohistochemistry. Quantification of immunoreactivity (ir) of stainings was performed by an investigator blind to the treatment group using NIH ImageJ software (for cardiac bundles and adrenals, area above threshold and optical density) and MBF StereoInvestigator (for cardiac fibers, area fraction fractionator probe). Sympathetic cardiac nerve bundle analysis and fiber area density showed a significant reduction in global cardiac tyrosine hydroxylase-ir (TH; catecholaminergic marker) in 6-OHDA animals compared to controls. Quantification of protein gene protein 9.5 (pan-neuronal marker) positive cardiac fibers showed a significant deficit in 6-OHDA monkeys compared to controls and correlated with TH-ir fiber area. Semi-quantitative evaluation of human leukocyte antigen-ir (inflammatory marker) and nitrotyrosine-ir (oxidative stress marker) did not show significant changes 3 months post-neurotoxin. Cardiac nerve bundle α-synuclein-ir (presynaptic protein) was reduced (trend) in 6-OHDA treated monkeys; insoluble proteinase-K resistant α-synuclein (typical of PD pathology) was not observed. In the adrenal medulla, 6-OHDA monkeys had significantly reduced TH-ir and aminoacid decarboxylase-ir. Our results confirm that systemic 6-OHDA dosing to nonhuman primates induces cardiac sympathetic neurodegeneration and loss of catecholaminergic enzymes in the adrenal medulla, and suggests that this model can be used as a platform to evaluate disease-modifying strategies aiming to induce peripheral neuroprotection.

    Topics: Adrenal Medulla; alpha-Synuclein; Animals; Aromatic-L-Amino-Acid Decarboxylases; Autonomic Fibers, Postganglionic; Chromaffin Cells; Disease Models, Animal; Female; Heart; Macaca mulatta; Male; Myocardium; Nerve Degeneration; Oxidopamine; Parkinson Disease, Secondary; Sympathectomy; Tyrosine 3-Monooxygenase

2014
Engineered antibody therapies coming of age for aging brains.
    Molecular therapy : the journal of the American Society of Gene Therapy, 2014, Volume: 22, Issue:10

    Topics: alpha-Synuclein; Animals; Brain; Endosomal Sorting Complexes Required for Transport; Nerve Degeneration; Single-Chain Antibodies

2014
Mutations in RAB39B cause X-linked intellectual disability and early-onset Parkinson disease with α-synuclein pathology.
    American journal of human genetics, 2014, Dec-04, Volume: 95, Issue:6

    Advances in understanding the etiology of Parkinson disease have been driven by the identification of causative mutations in families. Genetic analysis of an Australian family with three males displaying clinical features of early-onset parkinsonism and intellectual disability identified a ∼45 kb deletion resulting in the complete loss of RAB39B. We subsequently identified a missense mutation (c.503C>A [p.Thr168Lys]) in RAB39B in an unrelated Wisconsin kindred affected by a similar clinical phenotype. In silico and in vitro studies demonstrated that the mutation destabilized the protein, consistent with loss of function. In vitro small-hairpin-RNA-mediated knockdown of Rab39b resulted in a reduction in the density of α-synuclein immunoreactive puncta in dendritic processes of cultured neurons. In addition, in multiple cell models, we demonstrated that knockdown of Rab39b was associated with reduced steady-state levels of α-synuclein. Post mortem studies demonstrated that loss of RAB39B resulted in pathologically confirmed Parkinson disease. There was extensive dopaminergic neuron loss in the substantia nigra and widespread classic Lewy body pathology. Additional pathological features included cortical Lewy bodies, brain iron accumulation, tau immunoreactivity, and axonal spheroids. Overall, we have shown that loss-of-function mutations in RAB39B cause intellectual disability and pathologically confirmed early-onset Parkinson disease. The loss of RAB39B results in dysregulation of α-synuclein homeostasis and a spectrum of neuropathological features that implicate RAB39B in the pathogenesis of Parkinson disease and potentially other neurodegenerative disorders.

    Topics: alpha-Synuclein; Amino Acid Substitution; Australia; Base Sequence; Dopamine; Female; Gene Expression Regulation; Genes, X-Linked; Humans; Intellectual Disability; Lewy Bodies; Male; Middle Aged; Models, Molecular; Molecular Sequence Data; Mutation, Missense; Nerve Degeneration; Parkinson Disease; Pedigree; rab GTP-Binding Proteins; Sequence Analysis, DNA; Sequence Deletion; Substantia Nigra

2014
Valproic acid ameliorates C. elegans dopaminergic neurodegeneration with implications for ERK-MAPK signaling.
    Neuroscience letters, 2013, Apr-29, Volume: 541

    Parkinson's disease (PD) is a currently incurable neurodegenerative disorder that affects the aging population. The loss of dopaminergic neurons in the substantia nigra is one of the pathological features of PD. The precise causes of PD remain unresolved but evidence supports both environmental and genetic contributions. Current efforts for the treatment of PD are directed toward the discovery of compounds that show promise in impeding age-dependent neurodegeneration in PD patients. Alpha-synuclein (α-Syn) is a human protein that is mutated in specific populations of patients with familial PD. Overexpression of α-Syn in animal models of PD replicates key symptoms of PD, including neurodegeneration. Here, we use the nematode Caenorhabditis elegans as a model system, whereby α-Syn toxicity causes dopaminergic neurodegeneration, to test the capacity of valproic acid (VA) to protect neurons. The results of our study showed that treatment of nematodes with moderate concentrations of VA significantly protects dopaminergic neurons against α-Syn toxicity. Consistent with previously established knowledge related to the mechanistic action of VA in the cell, we showed through genetic analysis that the neuroprotection conferred by VA is inhibited by cell-specific depletion of the C. elegans ortholog of the MAP extracellular signal-regulated kinase (ERK), MPK-1, in the dopaminergic neurons. These findings suggest that VA may exert its neuroprotective effect via ERK-MAPK, or alternately could act with MAPK signaling to additively provide dopaminergic neuroprotection.

    Topics: alpha-Synuclein; Animals; Anticonvulsants; Caenorhabditis elegans; Caenorhabditis elegans Proteins; Dopaminergic Neurons; Humans; MAP Kinase Kinase 2; MAP Kinase Signaling System; Mitogen-Activated Protein Kinase 1; Nerve Degeneration; Neuroprotective Agents; Parkinson Disease; RNA Interference; Valproic Acid

2013
β-synuclein aggregates and induces neurodegeneration in dopaminergic neurons.
    Annals of neurology, 2013, Volume: 74, Issue:1

    Whereas the contribution of α-synuclein to neurodegeneration in Parkinson disease is well accepted, the putative impact of its close homologue, β-synuclein, is enigmatic. β-Synuclein is widely expressed throughout the central nervous system, as is α-synuclein, but the physiological functions of both proteins remain unknown. Recent findings have supported the view that β-synuclein can act as an ameliorating regulator of α-synuclein-induced neurotoxicity, having neuroprotective rather than neurodegenerative capabilities, and being nonaggregating due to the absence of most of the aggregation-promoting NAC domain. However, a mutation of β-synuclein linked to dementia with Lewy bodies rendered the protein neurotoxic in transgenic mice, and fibrillation of β-synuclein has been demonstrated in vitro.. Neurotoxicity and aggregation properties of α-, β-, and γ-synuclein were comparatively elucidated in the rat nigro-striatal projection and in cultured neurons.. Supporting the hypothesis that β-synuclein can act as a neurodegeneration-inducing factor, we demonstrated that wild-type β-synuclein is neurotoxic for cultured primary neurons. Furthermore, β-synuclein formed proteinase K-resistant aggregates in dopaminergic neurons in vivo, leading to pronounced and progressive neurodegeneration in rats. Expression of β-synuclein caused mitochondrial fragmentation, but this fragmentation did not render mitochondria nonfunctional in terms of ion handling and respiration even at late stages of neurodegeneration. A comparison of the neurodegenerative effects induced by α-, β-, and γ-synuclein revealed that β-synuclein was eventually as neurotoxic as α-synuclein for nigral dopaminergic neurons, whereas γ-synuclein proved to be nontoxic and had very low aggregation propensity.. Our results suggest that the role of β-synuclein as a putative modulator of neuropathology in aggregopathies like Parkinson disease and dementia with Lewy bodies needs to be revisited.

    Topics: alpha-Synuclein; Animals; beta-Synuclein; Biophysical Phenomena; Calcium; Cells, Cultured; Dependovirus; Dopaminergic Neurons; Embryo, Mammalian; Female; gamma-Synuclein; Green Fluorescent Proteins; Membrane Potentials; Microscopy, Electron, Transmission; Mitochondria; Mutation; Nerve Degeneration; Rats; Rats, Wistar; Respiration; Substantia Nigra; Transfection; Vesicular Monoamine Transport Proteins

2013
α-Synuclein in CSF of patients with severe traumatic brain injury.
    Neurology, 2013, Apr-30, Volume: 80, Issue:18

    The study aims to examine α-synuclein in the CSF of patients with severe traumatic brain injury (TBI) and its relationship with clinical characteristics and long-term outcomes.. This prospective case-control study enrolled patients with severe TBI (Glasgow Coma Score ≤ 8) who underwent ventriculostomy. CSF samples were taken from each TBI patient at admission and daily for up to 8 days after injury and successively assessed by ELISA. Control CSF was collected for analysis from subjects receiving lumbar puncture for other medical reasons. We used trajectory analysis to identify distinct temporal profiles of CSF α-synuclein that were compared with clinical outcomes.. CSF α-synuclein was elevated in TBI patients after injury as compared to controls (p = 0.0008). Overall, patients who died had higher concentrations (area under the curve) over 8 days of observation compared to those who survived at 6 months postinjury (p = 0.002). Two distinct temporal α-synuclein profiles were recognized over time. Subjects who died had consistently elevated α-synuclein levels compared to those who survived with α-synuclein levels near controls. High-risk trajectory was a strong and accurate predictor of death with 100% specificity and a very high sensitivity (83%).. Taken together, these data support the hypothesis that in severe TBI patients, substantial increase of CSF α-synuclein may indicate widespread neurodegeneration and reflect secondary neuropathologic events occurring after injury. The determination of CSF α-synuclein may be a valuable prognostic marker, adding to the clinical assessment and creating opportunities for medical intervention.

    Topics: Adult; Aged; alpha-Synuclein; Biomarkers; Brain Injuries; Case-Control Studies; Female; Glasgow Coma Scale; Humans; Male; Middle Aged; Nerve Degeneration; Pilot Projects; Prognosis; Prospective Studies; Risk Factors; Survival Analysis; Young Adult

2013
Neurodegenerative disease status and post-mortem pathology in idiopathic rapid-eye-movement sleep behaviour disorder: an observational cohort study.
    The Lancet. Neurology, 2013, Volume: 12, Issue:5

    We postulated that idiopathic rapid-eye-movement (REM) sleep behaviour disorder (IRBD) represents the prodromal phase of a Lewy body disorder and that, with sufficient follow-up, most cases would eventually be diagnosed with a clinical defined Lewy body disorder, such as Parkinson's disease (PD) or dementia with Lewy bodies (DLB).. Patients from an IRBD cohort recruited between 1991 and 2003, and previously assessed in 2005, were followed up during an additional period of 7 years. In this original cohort, we sought to identify the nature and frequency of emerging defined neurodegenerative syndromes diagnosed by standard clinical criteria. We estimated rates of survival free from defined neurodegenerative disease by means of the Kaplan-Meier method. We further characterised individuals who remained diagnosed as having only IRBD, through dopamine transporter (DAT) imaging, transcranial sonography (TCS), and olfactory testing. We did a neuropathological assessment in three patients who died during follow-up and who had the antemortem diagnosis of PD or DLB.. Of the 44 participants from the original cohort, 36 (82%) had developed a defined neurodegenerative syndrome by the 2012 assessment (16 patients were diagnosed with PD, 14 with DLB, one with multiple system atrophy, and five with mild cognitive impairment). The rates of neurological-disease-free survival from time of IRBD diagnosis were 65·2% (95% CI 50·9 to 79·5) at 5 years, 26·6% (12·7 to 40·5) at 10 years, and 7·5% (-1·9 to 16·9) at 14 years. Of the four remaining neurological-disease-free individuals who underwent neuroimaging and olfactory tests, all four had decreased striatal DAT uptake, one had substantia nigra hyperechogenicity on TCS, and two had impaired olfaction. In three patients, the antemortem diagnoses of PD and DLB were confirmed by neuropathological examination showing widespread Lewy bodies in the brain, and α-synuclein aggregates in the peripheral autonomic nervous system in one case. In these three patients, neuronal loss and Lewy pathology (α-synuclein-containing Lewy bodies and Lewy neurites) were found in the brainstem nuclei that regulate REM sleep atonia.. Most IRBD individuals from our cohort developed a Lewy body disorder with time. Patients who remained disease-free at follow-up showed markers of increased short-term risk for developing PD and DLB in IRBD, such as decreased striatal DAT binding. Our findings indicate that in most patients diagnosed with IRBD this parasomnia represents the prodromal phase of a Lewy body disorder. IRBD is a candidate for the study of early events and progression of this prodromal phase, and to test disease-modifying strategies to slow or stop the neurodegenerative process.. None.

    Topics: Aged; Aged, 80 and over; alpha-Synuclein; Brain; Cohort Studies; Disease Progression; Disease-Free Survival; Female; Humans; Lewy Body Disease; Male; Nerve Degeneration; Prodromal Symptoms; REM Sleep Behavior Disorder

2013
MHCII is required for α-synuclein-induced activation of microglia, CD4 T cell proliferation, and dopaminergic neurodegeneration.
    The Journal of neuroscience : the official journal of the Society for Neuroscience, 2013, Jun-05, Volume: 33, Issue:23

    Accumulation of α-synuclein (α-syn) in the brain is a core feature of Parkinson disease (PD) and leads to microglial activation, production of inflammatory cytokines and chemokines, T-cell infiltration, and neurodegeneration. Here, we have used both an in vivo mouse model induced by viral overexpression of α-syn as well as in vitro systems to study the role of the MHCII complex in α-syn-induced neuroinflammation and neurodegeneration. We find that in vivo, expression of full-length human α-syn causes striking induction of MHCII expression by microglia, while knock-out of MHCII prevents α-syn-induced microglial activation, antigen presentation, IgG deposition, and the degeneration of dopaminergic neurons. In vitro, treatment of microglia with aggregated α-syn leads to activation of antigen processing and presentation of antigen sufficient to drive CD4 T-cell proliferation and to trigger cytokine release. These results indicate a central role for microglial MHCII in the activation of both the innate and adaptive immune responses to α-syn in PD and suggest that the MHCII signaling complex may be a target of neuroprotective therapies for the disease.

    Topics: alpha-Synuclein; Animals; Animals, Newborn; CD4-Positive T-Lymphocytes; Cell Proliferation; Cells, Cultured; Dopaminergic Neurons; Genes, MHC Class II; Humans; Male; Mice; Mice, Inbred C57BL; Mice, Knockout; Microglia; Nerve Degeneration; Parkinson Disease

2013
Telmisartan attenuates MPTP induced dopaminergic degeneration and motor dysfunction through regulation of α-synuclein and neurotrophic factors (BDNF and GDNF) expression in C57BL/6J mice.
    Neuropharmacology, 2013, Volume: 73

    Telmisartan (TEL), an angiotensin type 1 receptor (AT1R) antagonist, has been reported to exert neuroprotective effect in animal models of Parkinson's disease (PD). However, its effect on motor functions, mutant protein α-synuclein (SYN) and neurotrophic factors (BDNF and GDNF) expression and their interrelation in PD has not yet been elucidated. In the present study, the effect of TEL on 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) induced motor dysfunctions and dopaminergic degeneration was ascertained through investigating the alterations in protein expression of dopamine transporter (DAT), tyrosine hydroxylase (TH) and SYN in C57BL/6J mouse. Further, the role of TEL on the gene expression of neurotrophic factors such as BDNF and GDNF and protein expression of vesicular monoamine transporter 2 (VMAT2) and Glial fibrillary acidic proteins (GFAP) were studied. In TEL treated mouse, strong negative correlation was observed between motor function and SYN, while a strong positive correlation was noted with BDNF and GDNF expression. TEL caused down-regulation of SYN, GFAP and up-regulation of DAT, TH, VAMT2, BDNF and GDNF expressions. Present data suggest that brain renin angiotensin system (RAS) plays a crucial role in motor function and in the regulation of key proteins such as SYN, BDNF and GDNF, DAT, TH, VMAT2 and GFAP in Parkinsonism. In conclusion, the present study shows that angiotensin type 1 receptor antagonists can ameliorate motor dysfunction and act as potential neuroprotective agent in the management of Parkinsonism.

    Topics: alpha-Synuclein; Angiotensin II Type 1 Receptor Blockers; Animals; Behavior, Animal; Benzimidazoles; Benzoates; Brain-Derived Neurotrophic Factor; Corpus Striatum; Dopamine; Dopamine Plasma Membrane Transport Proteins; Gait; Gene Expression; Glial Cell Line-Derived Neurotrophic Factor; Glial Fibrillary Acidic Protein; Male; Mice; Mice, Inbred C57BL; Nerve Degeneration; Neuroprotective Agents; Parkinsonian Disorders; Substantia Nigra; Telmisartan; Tyrosine 3-Monooxygenase; Vesicular Monoamine Transport Proteins

2013
Boosting chaperone-mediated autophagy in vivo mitigates α-synuclein-induced neurodegeneration.
    Brain : a journal of neurology, 2013, Volume: 136, Issue:Pt 7

    α-Synuclein levels are critical to Parkinson's disease pathogenesis. Wild-type α-synuclein is degraded partly by chaperone-mediated autophagy, and aberrant α-synuclein may act as an inhibitor of the pathway. To address whether the induction of chaperone-mediated autophagy may represent a potential therapy against α-synuclein-induced neurotoxicity, we overexpressed lysosomal-associated membrane protein 2a, the rate-limiting step of chaperone-mediated autophagy, in human neuroblastoma SH-SY5Y cells, rat primary cortical neurons in vitro, and nigral dopaminergic neurons in vivo. Overexpression of the lysosomal-associated membrane protein 2a in cellular systems led to upregulation of chaperone-mediated autophagy, decreased α-synuclein turnover, and selective protection against adenoviral-mediated wild-type α-synuclein neurotoxicity. Protection was observed even when the steady-state levels of α-synuclein were unchanged, suggesting that it occurred through the attenuation of α-synuclein-mediated dysfunction of chaperone-mediated autophagy. Overexpression of the lysosomal receptor through the nigral injection of recombinant adeno-associated virus vectors effectively ameliorated α-synuclein-induced dopaminergic neurodegeneration by increasing the survival of neurons located in the substantia nigra as well as the axon terminals located in the striatum, which was associated with a reduction in total α-synuclein levels and related aberrant species. We conclude that induction of chaperone-mediated autophagy may provide a novel therapeutic strategy in Parkinson's disease and related synucleinopathies through two different mechanisms: amelioration of dysfunction of chaperone-mediated autophagy and lowering of α-synuclein levels.

    Topics: alpha-Synuclein; Amphetamine; Analysis of Variance; Animals; Apomorphine; Autophagy; Cells, Cultured; Cerebral Cortex; Corpus Striatum; Dependovirus; Dopamine; Embryo, Mammalian; Enzyme Inhibitors; Female; Gene Expression Regulation; Genetic Vectors; Green Fluorescent Proteins; Hemagglutinins; Humans; Lysosomal Membrane Proteins; Lysosomal-Associated Membrane Protein 2; Macrolides; Mice; Molecular Chaperones; Motor Activity; Nerve Degeneration; Neuroblastoma; Neurons; Rats; Transfection; Tyrosine 3-Monooxygenase

2013
Parkinson disease affects peripheral sensory nerves in the pharynx.
    Journal of neuropathology and experimental neurology, 2013, Volume: 72, Issue:7

    Dysphagia is very common in patients with Parkinson disease (PD) and often leads to aspiration pneumonia, the most common cause of death in PD. Current therapies are largely ineffective for dysphagia. Because pharyngeal sensation normally triggers the swallowing reflex, we examined pharyngeal sensory nerves in PD patients for Lewy pathology.Sensory nerves supplying the pharynx were excised from autopsied pharynges obtained from patients with clinically diagnosed and neuropathologically confirmed PD (n = 10) and healthy age-matched controls (n = 4). We examined the glossopharyngeal nerve (cranial nerve IX), the pharyngeal sensory branch of the vagus nerve (PSB-X), and the internal superior laryngeal nerve (ISLN) innervating the laryngopharynx. Immunohistochemistry for phosphorylated α-synuclein was used to detect Lewy pathology. Axonal α-synuclein aggregates in the pharyngeal sensory nerves were identified in all of the PD subjects but not in the controls. The density of α-synuclein-positive lesions was greater in PD patients with dysphagia versus those without dysphagia. In addition, α-synuclein-immunoreactive nerve fibers in the ISLN were much more abundant than those in cranial nerve IX and PSB-X. These findings suggest that pharyngeal sensory nerves are directly affected by pathologic processes in PD. These abnormalities may decrease pharyngeal sensation, thereby impairing swallowing and airway protective reflexes and contributing to dysphagia and aspiration.

    Topics: Aged; Aged, 80 and over; alpha-Synuclein; Autonomic Pathways; Deglutition Disorders; Female; Glossopharyngeal Nerve; Humans; Laryngeal Nerves; Male; Nerve Degeneration; Parkinson Disease; Pharynx; Vagus Nerve

2013
Exosomes of BV-2 cells induced by alpha-synuclein: important mediator of neurodegeneration in PD.
    Neuroscience letters, 2013, Aug-26, Volume: 548

    Parkinson's disease (PD) is a progressive neurodegenerative disease. Alpha-synuclein aggregation, which can activate microglia to enhance its dopaminergic neurotoxicity, plays a central role in the progression of PD. However the mechanism is still unclear. To investigate how alpha-synuclein affects the neuron, exosomes were derived from alpha-synuclein treated mouse microglia cell line BV-2 cells by differential centrifugation and ultracentrifugation. We found that alpha-synuclein can induce an increase of exosomal secretion by microglia. These activated exosomes expressed a high level of MHC class II molecules and membrane TNF-α. In addition, the activated exosomes cause increased apoptosis. Exosomes secreted from activated microglias might be important mediator of alpha-synuclein-induced neurodegeneration in PD.

    Topics: alpha-Synuclein; Animals; Apoptosis; Cell Line; Exosomes; Mice; Microglia; Nerve Degeneration; Parkinson Disease

2013
Chronic overload of SEPT4, a parkin substrate that aggregates in Parkinson's disease, causes behavioral alterations but not neurodegeneration in mice.
    Molecular brain, 2013, Aug-11, Volume: 6

    In autosomal recessive early-onset Parkinsonism (PARK2), the pathogenetic process from the loss of function of a ubiquitin ligase parkin to the death of dopamine neurons remains unclear. A dominant hypothesis attributes the neurotoxicity to accumulated substrates that are exempt from parkin-mediated degradation. Parkin substrates include two septins; SEPT4/CDCrel-2 which coaggregates with α-synuclein as Lewy bodies in Parkinson's disease, and its closest homolog SEPT5/CDCrel-1/PNUTL1 whose overload with viral vector can rapidly eliminate dopamine neurons in rats. However, chronic effects of pan-neural overload of septins have never been examined in mammals. To address this, we established a line of transgenic mice that express the largest gene product SEPT4(54kDa) via the prion promoter in the entire brain.. Histological examination and biochemical quantification of SEPT4-associated proteins including α-synuclein and the dopamine transporter in the nigrostriatal dopamine neurons found no significant difference between Sept4(Tg/+) and wild-type littermates. Thus, the hypothetical pathogenicity by the chronic overload of SEPT4 alone, if any, is insufficient to trigger neurodegenerative process in the mouse brain. Intriguingly, however, a systematic battery of behavioral tests revealed unexpected abnormalities in Sept4(Tg/+) mice that include consistent attenuation of voluntary activities in distinct behavioral paradigms and altered social behaviors.. Together, these data indicate that septin dysregulations commonly found in postmortem human brains with Parkinson's disease, schizophrenia and bipolar disorders may be responsible for a subset of behavioral abnormalities in the patients.

    Topics: alpha-Synuclein; Animals; Behavior, Animal; Circadian Rhythm; Dopamine; Dopamine Plasma Membrane Transport Proteins; Exploratory Behavior; Humans; Methamphetamine; Mice; Mice, Inbred C57BL; Mice, Transgenic; Neostriatum; Nerve Degeneration; Parkinson Disease; Peptides; Protein Structure, Quaternary; Rats; Septins; Solubility; Substrate Specificity; Tyrosine 3-Monooxygenase; Ubiquitin-Protein Ligases

2013
Parkin depletion delays motor decline dose-dependently without overtly affecting neuropathology in α-synuclein transgenic mice.
    BMC neuroscience, 2013, Nov-05, Volume: 14

    Mutations of the gene encoding the major component of Lewy bodies (LB), α-synuclein (α-syn), cause autosomal dominant forms of Parkinson's disease (PD), whereas loss-of-function mutations of the gene encoding the multifunctional E3 ubiquitin-protein ligase Parkin account for autosomal recessive forms of the disease. Parkin overproduction protects against α-syn-dependent neurodegeneration in various in vitro and in vivo models, but it remains unclear whether this process is affected by Parkin deficiency. We addressed this issue, by carrying out more detailed analyses of transgenic mice overproducing the A30P variant of human α-syn (hA30Pα-syn) and with two, one or no parkin knockout alleles.. Longitudinal behavioral follow-up of these mice indicated that Parkin depletion delayed disease-predictive sensorimotor impairment due to α-syn accumulation, in a dose-dependent fashion. At the end stage of the disease, neuronal deposits containing fibrillar α-syn species phosphorylated at S129 (PS129α-syn) were the predominant neuropathological feature in hA30Pα-syn mice, regardless of their parkin expression. Some of these deposits colocalized with the LB markers ubiquitin and α-syn truncated at D135 (α-synD135), indicating that PS129α-syn is subjected to secondary posttranslational modification (PTM); these features were not significantly affected by parkin dysfunction.. These findings suggest that Parkin deficiency acts as a protective modifier in α-syn-dependent neurodegeneration, without overtly affecting the composition and characteristics of α-syn deposits in end-stage disease.

    Topics: alpha-Synuclein; Animals; Blotting, Western; Brain; Disease Models, Animal; Fluorescent Antibody Technique; Humans; Mass Spectrometry; Mice; Mice, Inbred C57BL; Mice, Knockout; Mice, Transgenic; Microscopy, Electron, Transmission; Motor Skills; Nerve Degeneration; Parkinson Disease; Ubiquitin-Protein Ligases

2013
rAAV2/7 vector-mediated overexpression of alpha-synuclein in mouse substantia nigra induces protein aggregation and progressive dose-dependent neurodegeneration.
    Molecular neurodegeneration, 2013, Nov-25, Volume: 8

    Alpha-synuclein is a key protein implicated in the pathogenesis of Parkinson's disease (PD). It is the main component of the Lewy bodies, a cardinal neuropathological feature in the disease. In addition, whole locus multiplications and point mutations in the gene coding for alpha-synuclein lead to autosomal dominant monogenic PD. Over the past decade, research on PD has impelled the development of new animal models based on alpha-synuclein. In this context, transgenic mouse lines have failed to reproduce several hallmarks of PD, especially the strong and progressive dopaminergic neurodegeneration over time that occurs in the patients. In contrast, viral vector-based models in rats and non-human primates display prominent, although highly variable, nigral dopaminergic neuron loss. However, the few studies available on viral vector-mediated overexpression of alpha-synuclein in mice report a weak neurodegenerative process and no clear Lewy body-like pathology. To address this issue, we performed a comprehensive comparative study of alpha-synuclein overexpression by means of recombinant adeno-associated viral vectors serotype 2/7 (rAAV2/7) at different doses in adult mouse substantia nigra.. We noted a significant and dose-dependent alpha-synucleinopathy over time upon nigral viral vector-mediated alpha-synuclein overexpression. We obtained a strong, progressive and dose-dependent loss of dopaminergic neurons in the substantia nigra, reaching a maximum of 82% after 8 weeks. This effect correlated with a reduction in tyrosine hydroxylase immunoreactivity in the striatum. Moreover, behavioural analysis revealed significant motor impairments from 12 weeks after injection on. In addition, we detected the presence of alpha-synuclein-positive aggregates in the remaining surviving neurons. When comparing wild-type to mutant A53T alpha-synuclein at the same vector dose, both induced a similar degree of cell death. These data were supported by a biochemical analysis that showed a net increase in soluble and insoluble alpha-synuclein expression over time to the same extent for both alpha-synuclein variants.. In conclusion, our in vivo data provide evidence that strong and significant alpha-synuclein-induced neuropathology and progressive dopaminergic neurodegeneration can be achieved in mouse brain by means of rAAV2/7.

    Topics: alpha-Synuclein; Animals; Blotting, Western; Dependovirus; Disease Models, Animal; Female; Genetic Vectors; Humans; Mice; Mice, Inbred C57BL; Mice, Transgenic; Microscopy, Confocal; Nerve Degeneration; Neurons; Parkinson Disease; Substantia Nigra; Transduction, Genetic; Transgenes

2013
Potential neuroprotective effect of ibuprofen, insights from the mice model of Parkinson's disease.
    Pharmacological reports : PR, 2013, Volume: 65, Issue:5

    Parkinson's disease (PD) is one of the most common neurodegenerative diseases. An inflammatory reaction seems to be involved in the pathological process in PD. Prospective clinical studies with various nonsteroidal anti-inflammatory drugs (NSAIDs) have shown that ibuprofen decreases the risk of PD. In the present study we investigated the influence of ibuprofen on dopaminergic neuron injury in the mice model of PD.. Twelve-month-old male C57Bl mice were injected with MPTP together with various doses of ibuprofen (10, 30 or 50 mg/kg), administered 1 h before MPTP injection for 7 consecutive days. Evaluation concerned dopamine content in the striatum, tyrosine hydroxylase (TH) protein and α-synuclein expression measured 7 and 21 days post MPTP administration (dpa).. MPTP caused injury to dopaminergic neuron endings in the striatum: dopamine content decreased by about 0% 7 dpa and by 85% 21 dpa; TH protein expression diminished by 21% 7 dpa; α-synuclein level decreased by 10 and 26% 7 and 21 dpa, respectively. Ibuprofen administration to mice treated with MPTP significantly increased the level of dopamine in the striatum 7 and 21 dpa. It also prevented TH protein decrease and increased α-synuclein level 21 dpa.. Ibuprofen was shown to protect neurons against MPTP-induced injury in the striatum. The possible mechanism of the neuroprotective effect of ibuprofen might be associated with decreased dopamine turnover and cyclooxygenases inhibition resulting in lower reactive oxygen species formation.

    Topics: alpha-Synuclein; Animals; Anti-Inflammatory Agents, Non-Steroidal; Basal Ganglia; Cytoprotection; Disease Models, Animal; Dopamine; Dose-Response Relationship, Drug; Ibuprofen; Male; Mice; Mice, Inbred C57BL; Nerve Degeneration; Neurons; Neuroprotective Agents; Parkinsonian Disorders; Time Factors; Tyrosine 3-Monooxygenase

2013
Alpha-synuclein pathology and axonal degeneration of the peripheral motor nerves innervating pharyngeal muscles in Parkinson disease.
    Journal of neuropathology and experimental neurology, 2013, Volume: 72, Issue:2

    Parkinson disease (PD) is a neurodegenerative disease primarily characterized by cardinal motor manifestations and CNS pathology. Current drug therapies can often stabilize these cardinal motor symptoms, and attention has shifted to the other motor and nonmotor symptoms of PD that are resistant to drug therapy. Dysphagia in PD is perhaps the most important drug-resistant symptom because it leads to aspiration and pneumonia, the leading cause of death. Here, we present direct evidence for degeneration of the pharyngeal motor nerves in PD. We examined the cervical vagal nerve (cranial nerve X), pharyngeal branch of nerve X, and pharyngeal plexus innervating the pharyngeal muscles in 14 postmortem specimens, that is, from 10 patients with PD and 4 age-matched control subjects. Synucleinopathy in the pharyngeal nerves was detected using an immunohistochemical method for phosphorylated α-synuclein. Alpha-synuclein aggregates were revealed in nerve X and the pharyngeal branch of nerve X, and immunoreactive intramuscular nerve twigs and axon terminals within the neuromuscular junctions were identified in all of the PD patients but in none of the controls. These findings indicate that the motor nervous system of the pharynx is involved in the pathologic process of PD. Notably, PD patients who have had dysphagia had a higher density of α-synuclein aggregates in the pharyngeal nerves than those without dysphagia. These findings indicate that motor involvement of the pharynx in PD is one of the factors leading to oropharyngeal dysphagia commonly seen in PD patients.

    Topics: Acetylcholinesterase; Aged; Aged, 80 and over; alpha-Synuclein; Axons; Case-Control Studies; Female; Humans; Male; Nerve Degeneration; Neurologic Examination; Neuromuscular Junction; Parkinson Disease; Pharyngeal Muscles; Silver Staining; Vagus Nerve

2013
Parkinson disease: from pathology to molecular disease mechanisms.
    Free radical biology & medicine, 2013, Volume: 62

    Parkinson disease (PD) is a complex neurodegenerative disorder with both motor and nonmotor symptoms owing to a spreading process of neuronal loss in the brain. At present, only symptomatic treatment exists and nothing can be done to halt the degenerative process, as its cause remains unclear. Risk factors such as aging, genetic susceptibility, and environmental factors all play a role in the onset of the pathogenic process but how these interlink to cause neuronal loss is not known. There have been major advances in the understanding of mechanisms that contribute to nigral dopaminergic cell death, including mitochondrial dysfunction, oxidative stress, altered protein handling, and inflammation. However, it is not known if the same processes are responsible for neuronal loss in nondopaminergic brain regions. Many of the known mechanisms of cell death are mirrored in toxin-based models of PD, but neuronal loss is rapid and not progressive and limited to dopaminergic cells, and drugs that protect against toxin-induced cell death have not translated into neuroprotective therapies in humans. Gene mutations identified in rare familial forms of PD encode proteins whose functions overlap widely with the known molecular pathways in sporadic disease and these have again expanded our knowledge of the neurodegenerative process but again have so far failed to yield effective models of sporadic disease when translated into animals. We seem to be missing some key parts of the jigsaw, the trigger event starting many years earlier in the disease process, and what we are looking at now is merely part of a downstream process that is the end stage of neuronal death.

    Topics: alpha-Synuclein; Animals; Disease Models, Animal; Genetic Predisposition to Disease; Humans; Mitochondria; Mutation; Nerve Degeneration; Oxidative Stress; Parkinson Disease; Substantia Nigra

2013
Extensive distribution of glial cytoplasmic inclusions in an autopsied case of multiple system atrophy with a prolonged 18-year clinical course.
    Neuropathology : official journal of the Japanese Society of Neuropathology, 2012, Volume: 32, Issue:1

    We describe herein an autopsied case of multiple system atrophy (MSA) with prolonged clinical course of 18 years, and evaluate the extent of neurodegeneration and glial cytoplasmic inclusions (GCIs) in the entire brain of this rare case. A 64-year-old woman presented with typical neurological symptoms and imaging features of MSA. Thereafter, she became bedridden, and breathing was assisted through a tracheostomy for 12 years. She died at the age of 82 after 18 years from the initial symptom. Post mortem examination revealed severe neurodegeneration in the inferior olive, pontine nuclei, substantia nigra, locus ceruleus, putamen and cerebellum. Notably, phosphorylated α-synuclein (p-α-syn)-positive GCIs were found in these areas, but their number was very low. In contrast, the density of GCIs was much higher in such regions as the tectum/tegmentum of the brainstem, pyramidal tracts, neocortices and limbic system, which usually contain a small number of GCIs. Another constituent of GCIs, ubiquitin (Ub) and Ub-associated autophagy substrate p62, were also positive in some GCIs, and distribution of Ub/p62 immunoreactivity was proportionate to that of p-α-syn+ GCIs despite the very long duration of the disease. Furthermore, this case had complicated hypoxic encephalopathy, but p-α-syn+ GCIs were also found in the damaged white matter, indicating the contribution of α-syncleinopathy as well as hypoxic effect to the secondary myelin and axonal loss in the white matter. Together, this rare case suggests the contribution of the disease duration to the prevalence of GCIs, and the possible involvement of the limbic system in extensive-stage disease.

    Topics: Age of Onset; Aged, 80 and over; alpha-Synuclein; Brain; Female; Humans; Inclusion Bodies; Middle Aged; Multiple System Atrophy; Nerve Degeneration; Neuroglia; Ubiquitin

2012
Melatonin reduces the expression of alpha-synuclein in the dopamine containing neuronal regions of amphetamine-treated postnatal rats.
    Journal of pineal research, 2012, Volume: 52, Issue:1

    Alpha-synuclein (α-syn) is a neuronal protein that is involved in various degenerative disorders such as Parkinson's disease. It is found in the presynaptic terminals and perinuclear zones of many brain regions. Amphetamine (AMPH), a psychostimulant drug abused progressively more commonly in recent years, has been known to induce neurotoxicity in the central dopaminergic pathway, which is associated with increased oxidative stress. Recently, AMPH has been shown to significantly increase the level of α-syn in dopaminergic neuroblastoma cell cultures. Melatonin is recognized as an antioxidant for the nervous system. This study tested whether melatonin can attenuate the effect of AMPH on the expression of α-syn in the dopaminergic pathway of the neonatal rat. Four-day old postnatal rats (P4) were injected subcutaneously with either AMPH (increasing dose, 5-10 mg/kg daily) alone or AMPH with melatonin (2 mg/kg) daily at 10:00 AM for 7 consecutive days. As determined using Western blot, the level of α-syn was significantly increased in the substantia nigra, dorsal striatum, nucleus accumbens, and prefrontal cortex of the AMPH-treated group, while melatonin treatment either prior to AMPH or alone decreased the accumulation of the protein to 77%, 96%, 78%, and 77% of the control value, respectively. Furthermore, an immunofluorescent study showed that the α-syn-immunoreactivity increased noticeably in the nuclei of cell bodies and nerve terminals of the AMPH-treated group. Again, melatonin lowered this immunoreactivity. These results indicate that melatonin has a direct or indirect effect in reducing the expression of α-syn in the postnatal rat. The exact mechanism of this mitigation should be further investigated.

    Topics: alpha-Synuclein; Amphetamine; Analysis of Variance; Animals; Animals, Newborn; Brain; Dopamine; Melatonin; Nerve Degeneration; Neuroprotective Agents; Rats; Rats, Wistar

2012
Inclusion of a portion of the native SNCA 3'UTR reduces toxicity of human S129A SNCA on striatal-projecting dopamine neurons in rat substantia nigra.
    Developmental neurobiology, 2012, Volume: 72, Issue:6

    Experimental models of Parkinson's disease (PD) created by aberrant expression of the alpha-synuclein (SNCA) coding region have been reported. However, noncoding regions function in normal physiology and recent in vitro studies have shown that microRNAs-7 and -153 regulate SNCA expression by binding the 3'UTR. Here, effects of different hSNCA forms were examined in vivo. Adult, male rats were injected into one substantia nigra (SN) with AAV-wtSNCA, AAV-S129A hSNCA, or AAV-S129D hSNCA either with or without a portion of the native 3'UTR. DA neurons in SN that maintained striatal (ST) projections at the end of treatment were retrogradely labeled by bilateral ST fluorogold (FG) injections and FG-positive DA neurons in SN were counted. At 5 weeks, hSNCA coding vectors reduced numbers of FG-positive neurons in injected SN compared with uninjected SN (wtSNCA, p = 0.05; S129A/D hSNCA, p = 0.01). At 7 and 9 weeks, wtSNCA- and S129D hSNCA-treated rats exhibited recovery, but S129A hSNCA-injected rats did not (p = 0.01). In contrast, numbers of FG-positive neurons were unaffected by hSNCA expression when the 3'UTR was included. When FG-positive neurons were expressed as the ratio of numbers in injected to uninjected sides, the S129A hSNCA coding vector resulted in the highest decrease at 9 weeks versus wtSNCA (p = 0.05) or S129D hSNCA (p = 0.01). Inclusion of the 3'UTR resulted in no significant differences in FG-positive neuron ratios. These data suggest that inclusion of the 3'UTR protects against S129A hSNCA-induced loss of nigrostriatal-projecting DA neurons in vivo and that mis-regulation of hSNCA expression and function at noncoding regions contribute to PD pathogenesis.

    Topics: 3' Untranslated Regions; alpha-Synuclein; Animals; Corpus Striatum; Disease Models, Animal; Dopaminergic Neurons; Genetic Therapy; Male; Nerve Degeneration; Neural Pathways; Parkinson Disease; Phosphorylation; Rats; Rats, Sprague-Dawley; Substantia Nigra

2012
Myeloperoxidase inhibition ameliorates multiple system atrophy-like degeneration in a transgenic mouse model.
    Neurotoxicity research, 2012, Volume: 21, Issue:4

    Multiple system atrophy (MSA) is a rare and fatal α-synucleinopathy characterized by a distinctive oligodendrogliopathy with glial cytoplasmic inclusions and associated neuronal multisystem degeneration. The majority of patients presents with a rapidly progressive parkinsonian disorder and atypical features such as early autonomic failure and cerebellar ataxia. We have previously reported that complete MSA pathology can be modeled in transgenic mice overexpressing oligodendroglial α-synuclein under conditions of oxidative stress induced by 3-nitropropionic acid (3-NP) including striatonigral degeneration, olivopontocerebellar atrophy, astrogliosis, and microglial activation. Here, we show that myeloperoxidase (MPO), a key enzyme involved in the production of reactive oxygen species by phagocytic cells, is expressed in both human and mouse MSA brains. We also demonstrate that in the MSA mouse model, MPO inhibition reduces motor impairment and rescues vulnerable neurons in striatum, substantia nigra pars compacta, cerebellar cortex, pontine nuclei, and inferior olives. MPO inhibition is associated with suppression of microglial activation but does not affect 3-NP induced astrogliosis in the same regions. Finally, MPO inhibition results in reduced intracellular aggregates of α-synuclein. This study suggests that MPO inhibition may represent a novel candidate treatment strategy against MSA-like neurodegeneration acting through its anti-inflammatory and anti-oxidative properties.

    Topics: Aged; alpha-Synuclein; Animals; Brain; Disease Models, Animal; Dose-Response Relationship, Drug; Enzyme Inhibitors; Gliosis; Humans; Male; Mice; Mice, Transgenic; Microglia; Middle Aged; Motor Activity; Multiple System Atrophy; Nerve Degeneration; Peroxidase; Pyrimidinones; Pyrroles

2012
Sustained expression of PGC-1α in the rat nigrostriatal system selectively impairs dopaminergic function.
    Human molecular genetics, 2012, Apr-15, Volume: 21, Issue:8

    Mitochondrial dysfunction and oxidative stress have been implicated in the etiology of Parkinson's disease. Therefore, pathways controlling mitochondrial activity rapidly emerge as potential therapeutic targets. Here, we explore the neuronal response to prolonged overexpression of peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α), a transcriptional regulator of mitochondrial function, both in vitro and in vivo. In neuronal primary cultures from the ventral midbrain, PGC-1α induces mitochondrial biogenesis and increases basal respiration. Over time, we observe an increasing proportion of the oxygen consumed by neurons which are dedicated to adenosine triphosphate production. In parallel to enhanced oxidative phosphorylation, PGC-1α progressively leads to a decrease in mitochondrial polarization. In the adult rat nigrostriatal system, adeno-associated virus (AAV)-mediated overexpression of PGC-1α induces the selective loss of dopaminergic markers and increases dopamine (DA) catabolism, leading to a reduction in striatal DA content. In addition, PGC-1α prevents the labeling of nigral neurons following striatal injection of the fluorogold retrograde tracer. When PGC-1α is expressed at higher levels following intranigral AAV injection, it leads to overt degeneration of dopaminergic neurons. Finally, PGC-1α overexpression does not prevent nigrostriatal degeneration in pathologic conditions induced by α-synuclein overexpression. Overall, we find that lasting overexpression of PGC-1α leads to major alterations in the metabolic activity of neuronal cells which dramatically impair dopaminergic function in vivo. These results highlight the central role of PGC-1α in the function and survival of dopaminergic neurons and the critical need for maintaining physiological levels of PGC-1α activity.

    Topics: Adenosine Triphosphate; alpha-Synuclein; Animals; Cell Respiration; Cell Survival; Cells, Cultured; Corpus Striatum; Dependovirus; Dopamine; Dopaminergic Neurons; Female; Gene Expression Regulation; Genetic Vectors; Humans; Membrane Potential, Mitochondrial; Mice; Mitochondria; Nerve Degeneration; Neurons; Oxidative Phosphorylation; Oxygen Consumption; Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha; Rats; Rats, Sprague-Dawley; Substantia Nigra; Trans-Activators; Transcription Factors

2012
Fluoxetine ameliorates behavioral and neuropathological deficits in a transgenic model mouse of α-synucleinopathy.
    Experimental neurology, 2012, Volume: 234, Issue:2

    The term α-synucleinopathies refers to a group of age-related neurological disorders including Parkinson's disease (PD), Dementia with Lewy Bodies (DLB) and Multiple System Atrophy (MSA) that display an abnormal accumulation of alpha-synuclein (α-syn). In contrast to the neuronal α-syn accumulation observed in PD and DLB, MSA is characterized by a widespread oligodendrocytic α-syn accumulation. Transgenic mice expressing human α-syn under the oligodendrocyte-specific myelin basic protein promoter (MBP1-hαsyn tg mice) model many of the behavioral and neuropathological alterations observed in MSA. Fluoxetine, a selective serotonin reuptake inhibitor, has been shown to be protective in toxin-induced models of PD, however its effects in an in vivo transgenic model of α-synucleinopathy remain unclear. In this context, this study examined the effect of fluoxetine in the MBP1-hαsyn tg mice, a model of MSA. Fluoxetine administration ameliorated motor deficits in the MBP1-hαsyn tg mice, with a concomitant decrease in neurodegenerative pathology in the basal ganglia, neocortex and hippocampus. Fluoxetine administration also increased levels of the neurotrophic factors, GDNF (glial-derived neurotrophic factor) and BDNF (brain-derived neurotrophic factor) in the MBP1-hαsyn tg mice compared to vehicle-treated tg mice. This fluoxetine-induced increase in GDNF and BDNF protein levels was accompanied by activation of the ERK signaling pathway. The effects of fluoxetine administration on myelin and serotonin markers were also examined. Collectively these results indicate that fluoxetine may represent a novel therapeutic intervention for MSA and other neurodegenerative disorders.

    Topics: alpha-Synuclein; Animals; Behavior, Animal; Brain; Fluoxetine; Glial Cell Line-Derived Neurotrophic Factor; Mice; Mice, Transgenic; Motor Skills; Nerve Degeneration; Neurons; Selective Serotonin Reuptake Inhibitors

2012
Emerging neurotoxic mechanisms in environmental factors-induced neurodegeneration.
    Neurotoxicology, 2012, Volume: 33, Issue:4

    Exposure to environmental neurotoxic metals, pesticides and other chemicals is increasingly recognized as a key risk factor in the pathogenesis of chronic neurodegenerative disorders such as Parkinson's and Alzheimer's diseases. Oxidative stress and apoptosis have been actively investigated as neurotoxic mechanisms over the past two decades, resulting in a greater understanding of neurotoxic processes. Nevertheless, emerging evidence indicates that epigenetic changes, protein aggregation and autophagy are important cellular and molecular correlates of neurodegenerative diseases resulting from chronic neurotoxic chemical exposure. During the Joint Conference of the 13th International Neurotoxicology Association and the 11th International Symposium on Neurobehavioral Methods and Effects in Occupational and Environmental Health, the recent progress made toward understanding epigenetic mechanisms, protein aggregation, autophagy, and deregulated kinase activation following neurotoxic chemical exposure and the relevance to neurodegenerative conditions were one of the themes of the symposium. Dr. Anumantha G. Kanthasamy described the role of acetylation of histones and non-histone proteins in neurotoxicant-induced neurodegenerative processes in the nigral dopaminergic neuronal system. Dr. Arthi Kanthasamy illustrated the role of autophagy as a key determinant in cell death events during neurotoxic insults. Dr. Ajay Rana provided evidence for posttranslational modification of α-synuclein protein by the Mixed Linage Kinase (MLK) group of kinases to initiate protein aggregation in cell culture and animal models of Parkinson's disease. These presentations outlined emerging cutting edge mechanisms that might set the stage for future mechanistic investigations into new frontiers of molecular neurotoxicology. This report summarizes the views of symposium participants, with emphasis on future directions for study of environmentally and occupationally linked chronic neurodegenerative diseases.

    Topics: alpha-Synuclein; Animals; Disease Models, Animal; Dopaminergic Neurons; Environmental Exposure; Environmental Pollutants; Epigenesis, Genetic; Gene Expression Regulation; Gene-Environment Interaction; Genetic Predisposition to Disease; Humans; Nerve Degeneration; Nervous System; Neurodegenerative Diseases; Parkinsonian Disorders; Proteasome Endopeptidase Complex; Protein Kinases; Risk Assessment; Risk Factors; Signal Transduction

2012
Comparison of the behavioural and histological characteristics of the 6-OHDA and α-synuclein rat models of Parkinson's disease.
    Experimental neurology, 2012, Volume: 235, Issue:1

    Development of relevant models of Parkinson's disease (PD) is essential for a better understanding of the pathological processes underlying the human disease and for the evaluation of promising targets for therapeutic intervention. To date, most pre-clinical studies have been performed in the well-established rodent and non-human primate models using injection of 6-hydroxydopamine (6-OHDA) or 1-methyl-4-phenyl-1,2,3,6-tetrahydroxypyridine (MPTP). Overexpression of the disease-causing protein α-synuclein (α-syn), using adeno-associated viral (AAV) vectors, has provided a novel model that recapitulates many features of the human disease. In the present study we compared the AAV-α-syn rat model with models where the nigro-striatal pathway is lesioned by injection of 6-OHDA in the striatum (partial lesion) or the medial forebrain bundle (full lesion). Examination of the behavioural changes over time revealed a different progression and magnitude of the motor impairment. Interestingly, dopamine (DA) neuron loss is prominent in both the toxin and the AAV-α-syn models. However, α-syn overexpressing animals were seen to exhibit less cell and terminal loss for an equivalent level of motor abnormalities. Prominent and persistent axonal pathology is only observed in the α-syn rat model. We suggest that, while neuronal and terminal loss mainly accounts for the behavioural impairment in the toxin-based model, similar motor deficits result from the combination of cell death and dysfunction of the remaining nigro-striatal neurons in the AAV-α-syn model. While the two models have been developed to mimic DA neuron deficiency, they differ in their temporal and neuropathological characteristics, and replicate different aspects of the pathophysiology of the human disease. This study suggests that the AAV-α-syn model replicates the human pathology more closely than either of the other two 6-OHDA lesion models.

    Topics: alpha-Synuclein; Animals; Behavior, Animal; Corpus Striatum; Disease Models, Animal; Female; Medial Forebrain Bundle; Motor Activity; Nerve Degeneration; Neurons; Oxidopamine; Parkinson Disease, Secondary; Rats; Rats, Sprague-Dawley; Substantia Nigra

2012
A proteomic analysis of MCLR-induced neurotoxicity: implications for Alzheimer's disease.
    Toxicological sciences : an official journal of the Society of Toxicology, 2012, Volume: 127, Issue:2

    Cyanobacteria-derived microcystin-leucine-arginine (MCLR), commonly characterized as a hepatotoxin, has recently been found to show neurotoxicity, but the exact mechanism is still unknown. To further our understanding of the neurotoxic effects of MCLR and the mechanisms behind it, we used two-dimensional gel electrophoresis and mass spectrometry analysis to identify global protein profiles associated with MCLR-induced neurotoxicity. MCLR-treated hippocampi showed alterations in proteins involved in cytoskeleton, neurodegenerative disease, oxidative stress, apoptosis, and energy metabolism. After validation by Western blot and quantitative real-time PCR, the expressions of three proteins related to neurodegenerative disease, septin 5, α-internexin, and α-synuclein, were identified to be altered by MCLR exposure. Based on our proteomic analysis that MCLR toxicity might be linked to neurodegeneration, we examined the activity of serine/threonine-specific protein phosphatases (PPs), which are markers of neurodegenerative disease. MCLR was found to induce inhibition of PPs and abnormal hyperphosphorylation of the neuronal microtubule-associated protein tau. This was found to lead to impairment of learning and memory, accompanied by severe histological damage and neuronal apoptosis in the hippocampal CA1 regions of rats. Our results support the hypothesis that MCLR could induce neurotoxic effects, the reason for which could be attributed to the disruption of the cytoskeleton, oxidative stress, and inhibition of PPs in the hippocampus. Moreover, MCLR was found to induce tau hyperphosphorylation, spatial memory impairment, neuronal degenerative changes, and apoptosis, suggesting that this cyanotoxin may contribute to Alzheimer's disease in humans.

    Topics: alpha-Synuclein; Alzheimer Disease; Animals; Apoptosis; Blotting, Western; Cell Cycle Proteins; Dose-Response Relationship, Drug; Electrophoresis, Gel, Two-Dimensional; Hippocampus; Intermediate Filament Proteins; Male; Memory; Microcystins; Nerve Degeneration; Nerve Tissue Proteins; Neurotoxicity Syndromes; Phosphoprotein Phosphatases; Phosphorylation; Proteomics; Rats; Rats, Wistar; Real-Time Polymerase Chain Reaction; Reproducibility of Results; Septins; Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization; tau Proteins; Time Factors

2012
Lysosomal dysfunction in neurodegeneration: the role of ATP13A2/PARK9.
    Autophagy, 2012, Volume: 8, Issue:6

    Neuronal homeostasis and survival critically depend on an efficient autophagy-lysosomal degradation pathway, especially since neurons cannot reduce the concentration of misfolded proteins and damaged organelles by cell division. While increasing evidence implicates lysosomal dysfunction in the pathogenesis of neurodegenerative disorders, the molecular underpinnings of the role of lysosomes in neurodegeneration remain largely unknown. To this end, studies of neurodegenerative disorders caused by mutations in lysosomal proteins offer an opportunity to elucidate such mechanisms and potentially identify specific therapeutic targets. One of these disorders is Kufor-Rakeb syndrome, caused by mutations in the lysosomal protein ATP13A2/PARK9 and characterized by early-onset Parkinsonism, pyramidal degeneration and dementia. We found that loss of ATP13A2 function results in impaired lysosomal function and, consequently, accumulation of SNCA/α-synuclein and neurotoxicity. Our results suggest that targeting of ATP13A2 to lysosomes to enhance lysosomal function may result in neuroprotection in Kufor-Rakeb syndrome. From a broader perspective, these findings, together with other recent studies of lysosomal dysfunction in neurodegeneration, suggest that strategies to upregulate lysosomal function in neurons represent a promising therapeutic approach for neurodegenerative disorders.

    Topics: alpha-Synuclein; Animals; Caenorhabditis elegans; Glucosylceramidase; Humans; Lysosomes; Mice; Nerve Degeneration; Proton-Translocating ATPases

2012
Loss of spinal motor neurons and alteration of alpha-synuclein immunostaining in MPTP induced Parkinsonism in mice.
    Journal of chemical neuroanatomy, 2012, Volume: 44, Issue:2

    1-Methyl, 4-phenyl, 1,2,3,6-tetrahydropiridine (MPTP) is a neurotoxin, widely used to produce experimental models of Parkinson Disease in rodents and primates. Although dopaminergic neurons are the most sensitive to MPTP neurotoxicity, different neuronal subtypes are affected. Among these, recent studies indicate that MPTP may produce pathological effects on spinal neurons. In fact, MPTP activates apoptotic proteins within the spinal cord and in particular within the motor neurons, suggesting commonalities between Parkinson Disease and Amyotrophic Lateral Sclerosis. In order to assess this point, in the present study we measured whether MPTP produces motor neurons loss. We chose a dose of MPTP (20 mg/kg × 3, 2 h apart), which in C57BL/6N mice was able to induce a massive nigrostriatal damage. Since both Parkinson Disease and Amyotrophic Lateral Sclerosis are characterized by altered alpha-synuclein immunostaining, this protein was also evaluated within spinal motor neurons, following MPTP administration. Three different monoclonal antibodies, recognizing distinct epitopes in the sequence of alpha-synuclein were used. Severe dopaminergic cell loss was quantified by stereology within the substantia nigra pars compacta, along with marked decrease of striatal tyrosine hydroxylase densitometry. The same doses of MPTP also caused a significant motor neuron loss in the spinal cord (roughly 30%). Spared motor neurons appeared often dysmorphic and vacuolated and possessed altered alpha-synuclein immunostaining. This latter finding extended to other cell types of the spinal cord. These data indicate that MPTP, apart from being a dopaminergic neurotoxin, produces also motor neuron death, thus bridging experimental Parkinsonism and motor neuron disease.

    Topics: 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine; alpha-Synuclein; Animals; Apoptosis; Cell Count; Corpus Striatum; Dopaminergic Neurons; Humans; Immunohistochemistry; Mice; Mice, Inbred C57BL; Motor Neurons; MPTP Poisoning; Nerve Degeneration; Spinal Cord; Stereotaxic Techniques; Substantia Nigra; Tyrosine 3-Monooxygenase

2012
Identification of novel ATP13A2 interactors and their role in α-synuclein misfolding and toxicity.
    Human molecular genetics, 2012, Sep-01, Volume: 21, Issue:17

    Lysosomes are responsible for degradation and recycling of bulky cell material, including accumulated misfolded proteins and dysfunctional organelles. Increasing evidence implicates lysosomal dysfunction in several neurodegenerative disorders, including Parkinson's disease and related synucleinopathies, which are characterized by the accumulation of α-synuclein (α-syn) in Lewy bodies. Studies of lysosomal proteins linked to neurodegenerative disorders present an opportunity to uncover specific molecular mechanisms and pathways that contribute to neurodegeneration. Loss-of-function mutations in a lysosomal protein, ATP13A2 (PARK9), cause Kufor-Rakeb syndrome that is characterized by early-onset parkinsonism, pyramidal degeneration and dementia. While loss of ATP13A2 function plays a role in α-syn misfolding and toxicity, the normal function of ATP13A2 in the brain remains largely unknown. Here, we performed a screen to identify ATP13A2 interacting partners, as a first step toward elucidating its function. Utilizing a split-ubiquitin membrane yeast two-hybrid system that was developed to identify interacting partners of full-length integral membrane proteins, we identified 43 novel interactors that primarily implicate ATP13A2 in cellular processes such as endoplasmic reticulum (ER) translocation, ER-to-Golgi trafficking and vesicular transport and fusion. We showed that a subset of these interactors modified α-syn aggregation and α-syn-mediated degeneration of dopaminergic neurons in Caenorhabditis elegans, further suggesting that ATP13A2 and α-syn are functionally linked in neurodegeneration. These results implicate ATP13A2 in vesicular trafficking and provide a platform for further studies of ATP13A2 in neurodegeneration.

    Topics: alpha-Synuclein; Animals; Caenorhabditis elegans; Dopaminergic Neurons; Gene Knockdown Techniques; HEK293 Cells; Humans; Nerve Degeneration; Protein Binding; Protein Folding; Proton-Translocating ATPases; Reproducibility of Results; Two-Hybrid System Techniques

2012
Disrupted autophagy leads to dopaminergic axon and dendrite degeneration and promotes presynaptic accumulation of α-synuclein and LRRK2 in the brain.
    The Journal of neuroscience : the official journal of the Society for Neuroscience, 2012, May-30, Volume: 32, Issue:22

    Parkinson's disease (PD) is characterized pathologically by the formation of ubiquitin and α-synuclein (α-syn)-containing inclusions (Lewy bodies), dystrophic dopamine (DA) terminals, and degeneration of midbrain DA neurons. The precise molecular mechanisms underlying these pathological features remain elusive. Accumulating evidence has implicated dysfunctional autophagy, the cell self-digestion and neuroprotective pathway, as one of the pathogenic systems contributing to the development of idiopathic PD. Here we characterize autophagy-deficient mouse models and provide in vivo evidence for the potential role that impaired autophagy plays in pathogenesis associated with PD. Cell-specific deletion of essential autophagy gene Atg7 in midbrain DA neurons causes delayed neurodegeneration, accompanied by late-onset locomotor deficits. In contrast, Atg7-deficient DA neurons in the midbrain exhibit early dendritic and axonal dystrophy, reduced striatal dopamine content, and the formation of somatic and dendritic ubiquitinated inclusions in DA neurons. Furthermore, whole-brain-specific loss of Atg7 leads to presynaptic accumulation of α-syn and LRRK2 proteins, which are encoded by two autosomal dominantly inherited PD-related genes. Our results suggest that disrupted autophagy may be associated with enhanced levels of endogenous α-syn and LRRK2 proteins in vivo. Our findings implicate dysfunctional autophagy as one of the failing cellular mechanisms involved in the pathogenesis of idiopathic PD.

    Topics: alpha-Synuclein; Animals; Autophagy; Autophagy-Related Protein 5; Autophagy-Related Protein 7; Brain; Cells, Cultured; Chromatography, High Pressure Liquid; Dendrites; Disease Models, Animal; Dopamine; Dopaminergic Neurons; Embryo, Mammalian; Ephrin-B1; Fibroblasts; Gene Expression Regulation; Inclusion Bodies; Intermediate Filament Proteins; Mice; Mice, Transgenic; Microtubule-Associated Proteins; Motor Activity; Movement Disorders; Nerve Degeneration; Nerve Tissue Proteins; Nestin; Presynaptic Terminals; Tyrosine 3-Monooxygenase; Ubiquitin

2012
Conditional expression of Parkinson's disease-related mutant α-synuclein in the midbrain dopaminergic neurons causes progressive neurodegeneration and degradation of transcription factor nuclear receptor related 1.
    The Journal of neuroscience : the official journal of the Society for Neuroscience, 2012, Jul-04, Volume: 32, Issue:27

    α-Synuclein (α-syn) plays a prominent role in the degeneration of midbrain dopaminergic (mDA) neurons in Parkinson's disease (PD). However, only a few studies on α-syn have been performed in the mDA neurons in vivo, which may be attributed to a lack of α-syn transgenic mice that develop PD-like severe degeneration of mDA neurons. To gain mechanistic insights into the α-syn-induced mDA neurodegeneration, we generated a new line of tetracycline-regulated inducible transgenic mice that overexpressed the PD-related α-syn A53T missense mutation in the mDA neurons. Here we show that the mutant mice developed profound motor disabilities and robust mDA neurodegeneration, resembling some key motor and pathological phenotypes of PD. We also systematically examined the subcellular abnormalities that appeared in the mDA neurons of mutant mice and observed a profound decrease of dopamine release, the fragmentation of Golgi apparatus, and the impairments of autophagy/lysosome degradation pathways in these neurons. To further understand the specific molecular events leading to the α-syn-dependent degeneration of mDA neurons, we found that overexpression of α-syn promoted a proteasome-dependent degradation of nuclear receptor-related 1 protein (Nurr1), whereas inhibition of Nurr1 degradation ameliorated the α-syn-induced loss of mDA neurons. Given that Nurr1 plays an essential role in maintaining the normal function and survival of mDA neurons, our studies suggest that the α-syn-mediated suppression of Nurr1 protein expression may contribute to the preferential vulnerability of mDA neurons in the pathogenesis of PD.

    Topics: alpha-Synuclein; Animals; Animals, Newborn; Disease Models, Animal; Disease Progression; Dopaminergic Neurons; Female; HEK293 Cells; Humans; Male; Mesencephalon; Mice; Mice, Inbred C57BL; Mice, Transgenic; Mutation, Missense; Nerve Degeneration; Nuclear Receptor Subfamily 4, Group A, Member 2; Parkinsonian Disorders; Primary Cell Culture

2012
In vivo alterations in calcium buffering capacity in transgenic mouse model of synucleinopathy.
    The Journal of neuroscience : the official journal of the Society for Neuroscience, 2012, Jul-18, Volume: 32, Issue:29

    Abnormal accumulation of α-synuclein is centrally involved in the pathogenesis of many disorders with Parkinsonism and dementia. Previous in vitro studies suggest that α-synuclein dysregulates intracellular calcium. However, it is unclear whether these alterations occur in vivo. For this reason, we investigated calcium dynamics in transgenic mice expressing human WT α-synuclein using two-photon microscopy. We imaged spontaneous and stimulus-induced neuronal activity in the barrel cortex. Transgenic mice exhibited augmented, long-lasting calcium transients characterized by considerable deviation from the exponential decay. The most evident pathology was observed in response to a repetitive stimulation in which subsequent stimuli were presented before relaxation of calcium signal to the baseline. These alterations were detected in the absence of significant increase in neuronal spiking response compared with age-matched controls, supporting the possibility that α-synuclein promoted alterations in calcium dynamics via interference with intracellular buffering mechanisms. The characteristic shape of calcium decay and augmented response during repetitive stimulation can serve as in vivo imaging biomarkers in this model of neurodegeneration, to monitor progression of the disease and screen candidate treatment strategies.

    Topics: alpha-Synuclein; Animals; Brain; Brain Diseases; Calcium; Disease Models, Animal; Female; Mice; Mice, Transgenic; Nerve Degeneration; Neurons

2012
α-Synuclein disrupts stress signaling by inhibiting polo-like kinase Cdc5/Plk2.
    Proceedings of the National Academy of Sciences of the United States of America, 2012, Oct-02, Volume: 109, Issue:40

    Parkinson disease (PD) results from the slow, progressive loss of dopaminergic neurons in the substantia nigra. Alterations in α-synuclein (aSyn), such as mutations or multiplications of the gene, are thought to trigger this degeneration. Here, we show that aSyn disrupts mitogen-activated protein kinase (MAPK)-controlled stress signaling in yeast and human cells, which results in inefficient cell protective responses and cell death. aSyn is a substrate of the yeast (and human) polo-like kinase Cdc5 (Plk2), and elevated levels of aSyn prevent Cdc5 from maintaining a normal level of GTP-bound Rho1, which is an essential GTPase that regulates stress signaling. The nine N-terminal amino acids of aSyn are essential for the interaction with polo-like kinases. The results support a unique mechanism of PD pathology.

    Topics: alpha-Synuclein; Analysis of Variance; beta-Galactosidase; Blotting, Western; Bridged Bicyclo Compounds, Heterocyclic; Cell Line, Tumor; Humans; Microscopy, Fluorescence; Mitogen-Activated Protein Kinases; Nerve Degeneration; Parkinson Disease; Protein Serine-Threonine Kinases; Signal Transduction; Thiazolidines; Yeasts

2012
Antibody-aided clearance of extracellular α-synuclein prevents cell-to-cell aggregate transmission.
    The Journal of neuroscience : the official journal of the Society for Neuroscience, 2012, Sep-26, Volume: 32, Issue:39

    Abnormal deposition and intercellular propagation of α-synuclein plays a central role in the pathogenesis of disorders such as Parkinson's Disease (PD) and dementia with Lewy bodies (DLB). Previous studies demonstrated that immunization against α-synuclein resulted in reduced α-synuclein accumulation and synaptic loss in a transgenic (tg) mouse model, highlighting the potential for immunotherapy. However, the mechanism by which immunization prevents synucleinopathy-associated deficits remains unknown. Here, we show that antibodies against α-synuclein specifically target and aid in clearance of extracellular α-synuclein proteins by microglia, thereby preventing their actions on neighboring cells. Antibody-assisted clearance occurs mainly in microglia through the Fcγ receptor, and not in neuronal cells or astrocytes. Stereotaxic administration of antibody into the brains of α-synuclein tg mice prevented neuron-to-astroglia transmission of α-synuclein and led to increased localization of α-synuclein and the antibody in microglia. Furthermore, passive immunization with α-synuclein antibody reduced neuronal and glial accumulation of α-synuclein and ameliorated neurodegeneration and behavioral deficits associated with α-synuclein overexpression. These findings provide an underlying mechanistic basis for immunotherapy for PD/DLB and suggest extracellular forms of α-synuclein as potential therapeutic targets.

    Topics: alpha-Synuclein; Amyloid; Analysis of Variance; Animals; Antibodies; Antigens, CD; Astrocytes; Brain; Calcium-Binding Proteins; Cathepsin D; Caveolin 1; Cell Communication; Cell Line; Chromatography, Gel; Culture Media, Conditioned; Cytokines; Disease Models, Animal; Extracellular Space; Humans; Immunization, Passive; Lewy Body Disease; Mice; Mice, Transgenic; Microfilament Proteins; Microscopy, Electron, Transmission; Nerve Degeneration; Neuroglia; Phosphopyruvate Hydratase; Platelet-Derived Growth Factor; Synaptic Transmission

2012
Increased iron levels correlate with the selective nigral dopaminergic neuron degeneration in Parkinson's disease.
    Journal of neural transmission (Vienna, Austria : 1996), 2011, Volume: 118, Issue:3

    The staging of Lewy-related pathology in sporadic Parkinson's disease (PD) reveals that many brain nuclei are affected in PD during different stages, except the ventral tegmental area (VTA), which is close related to the substantia nigra (SN) and enriched in dopamine (DA) neurons. Why DA neurons are selectively degenerated in the SN of PD is far from known. In the present study, we observed that the number of tyrosine hydroxylase immunoreactive neurons decreased and iron-staining positive cells increased in the SN, but not in the VTA, in the chronic 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated PD mice. Increased expression of divalent metal transporter 1 and decreased expression of ferroportin 1 might associate with this increased nigral iron levels. Lipofuscin granular aggregations and upregulation of alpha-synuclein (α-synuclein) were also observed only in the SN. These results suggest that increased iron levels associate with the selective degeneration of DA neurons in the SN. The intracellular regulation mechanisms for the iron transporters may be different in the SN and VTA under the same conditions. Moreover, the lipofuscin granular aggregations and upregulation of α-synuclein were also involved in the selective degeneration of dopaminergic neurons in the SN.

    Topics: alpha-Synuclein; Animals; Cell Count; Dopamine; Immunohistochemistry; Iron; Male; Mice; Mice, Inbred C57BL; MPTP Poisoning; Nerve Degeneration; Neurons; Parkinson Disease, Secondary; Random Allocation; Substantia Nigra; Tyrosine 3-Monooxygenase; Ventral Tegmental Area

2011
Alpha-synuclein loss in spinal muscular atrophy.
    Journal of molecular neuroscience : MN, 2011, Volume: 43, Issue:3

    Spinal muscular atrophy, the most prevalent hereditary motor neuron disease, is caused by mutations in the survival motor neuron (SMN) 1 gene. A significant reduction in the encoded SMN protein leads to the degeneration of motor neurons. However, the molecular events leading to this process are not well understood. The present study uses a previously developed neuronal cell culture model of spinal muscular atrophy for a multiplex transcriptome analysis. Furthermore, gene expression analysis was performed on in vitro cell cultures, as well as tissue samples of spinal muscular atrophy patients and transgenic mice. RNA and subsequent Western blot protein analyses suggest that low SMN levels are associated with significantly lower alpha-synuclein expression. Examination of two genes related to vesicular transport showed a similar though less dramatic decrease in expression. The 140-amino acid protein alpha-synuclein, dominant mutations of which have previously been associated with an autosomal dominant form of Parkinson's disease, is strongly expressed in select neurons of the brain. Although not well understood, the physiologic functions of alpha-synuclein have been linked to synaptic vesicular neurotransmitter release and neuroprotection, suggesting a possible contribution to Smn-deficient motor neuron pathology. Furthermore, alpha-synuclein may be a genetic modifier or biomarker of spinal muscular atrophy.

    Topics: alpha-Synuclein; Animals; Cells, Cultured; Fibroblasts; Humans; Mice; Mice, Transgenic; Motor Neurons; Muscular Atrophy, Spinal; Nerve Degeneration; SMN Complex Proteins

2011
Tau-positive glial cytoplasmic granules in multiple system atrophy.
    Neuropathology : official journal of the Japanese Society of Neuropathology, 2011, Volume: 31, Issue:3

    Multiple system atrophy (MSA) is a sporadic neurodegenerative disease that is pathologically characterized by the filamentous aggregation of α-synuclein. We report a case of MSA showing unusual neuropathological findings and review six autopsied cases of MSA. The patient progressively developed parkinsonism and ataxia for the 9 years prior to her death at the age of 72 years. Neuropathological examinations revealed neuronal loss restricted to the olivopontocerebellar and striatonigral region, which was more severe in the putamen. Staining with anti-α-synuclein antibody demonstrated widespread occurrence of glial cytoplasmic inclusions, which mainly accumulated in oligodendroglial cells and corresponded closely to the degree of disease progression. In addition, tau-positive granules were detected within the glial cytoplasm in the neurodegenerative region, which was especially prominent in the putamen and internal capsule. Tau accumulation was also clearly recognized by staining with specific antibodies against three-repeat or four-repeat tau. The glia that demonstrated deposition of tau-positive granules were distinguished from α-synuclein-positive oligodendroglia by double immunohistochemical staining. These characteristic glial accumulations of tau were also present in all six cases of MSA. These results indicate that tau-positive granules in glia are common findings in MSA and that tau aggregation might be another pathway to neurodegeneration in MSA.

    Topics: Aged; alpha-Synuclein; Cytoplasmic Granules; Fatal Outcome; Female; Humans; Male; Middle Aged; Multiple System Atrophy; Nerve Degeneration; Neuroglia; Olivary Nucleus; Substantia Nigra; tau Proteins

2011
Glutathione depletion and overproduction both initiate degeneration of nigral dopaminergic neurons.
    Acta neuropathologica, 2011, Volume: 121, Issue:4

    Parkinson's disease is a neurodegenerative disorder characterized by severe motor deficits mainly due to degeneration of dopaminergic neurons in the substantia nigra. Decreased levels of the cell's most important anti-oxidant, glutathione, have been detected in nigral neurons of Parkinson patients, but it is unknown if they are the cause or merely the consequence of the disease. To elucidate if glutathione depletion causes selective degeneration of nigral dopaminergic neurons, we down-regulated glutathione synthesis in different brain areas of adult rats by a viral vector-based RNAi approach. Decreased glutathione synthesis resulted in progressive degeneration of nigral dopaminergic neurons, while extra-nigral and striatal neurons were significantly less vulnerable. Degeneration of dopaminergic neurons was accompanied by progressive protein aggregate formation and functional motor deficits and was partially rescued by α-synuclein. That the survival of nigral dopaminergic neurons depends on the precise control of glutathione levels was further demonstrated by significant degeneration induced through moderate overproduction of glutathione. Over-expression of either of the two subunits of glutamate-cysteine ligase induced aberrant glutathiolation of cellular proteins and significant degeneration of dopaminergic neurons. Thus, while glutathione depletion was demonstrated to be a selective trigger for dopaminergic neuron degeneration, a glutathione replacement approach as a potential treatment option for Parkinson's patients must be considered with great care. In conclusion, our data demonstrate that survival of nigral dopaminergic neurons crucially depends on a tight regulation of their glutathione levels and that the depleted glutathione content detected in the brains of Parkinson's disease patients can be a causative insult for neuronal degeneration.

    Topics: alpha-Synuclein; Analysis of Variance; Animals; Apomorphine; Catalytic Domain; Cell Survival; Cysteine; Disease Models, Animal; Dopamine; Dopamine Agonists; Female; Gene Expression Regulation; Gliosis; Glutathione; Glutathione Reductase; Green Fluorescent Proteins; Movement; Nerve Degeneration; Rats; Rats, Wistar; RNA, Small Interfering; Sensation Disorders; Stereotyped Behavior; Substantia Nigra; Time Factors; Transduction, Genetic; Tyrosine 3-Monooxygenase; Vesicular Monoamine Transport Proteins

2011
Cerebrospinal fluid from patients with multiple system atrophy promotes in vitro α-synuclein fibril formation.
    Neuroscience letters, 2011, Mar-10, Volume: 491, Issue:1

    The aggregation of α-synuclein (αS) in the central nervous system (CNS) is the hallmark of multiple system atrophy (MSA) and Lewy body diseases including Parkinson's disease (PD) and dementia with Lewy bodies (DLB) (α-synucleinopathies). To test the hypothesis that patients with α-synucleinopathies have a CNS environment favorable for αS aggregation, we examined the influence of cerebrospinal fluid (CSF) from patients with MSA (n=20), DLB (n=8), and PD (n=10) on in vitro αS fibril (fαS) formation at pH 7.5 and 37°C using fluorescence spectroscopy with thioflavin S, compared with those with hereditary spinocerebellar ataxia (hSCA) (n=16), and tension-type headache (n=7). CSF from MSA patients (MSA-CSF) promoted fαS formation more strongly than PD-, hSCA-, or headache-CSF. By electron microscopic analyses, the width of fαS formed in MSA-CSF was significantly greater than others. MSA may have a CSF environment particularly favorable for fαS formation.

    Topics: Adult; Aged; Aged, 80 and over; alpha-Synuclein; Cerebrospinal Fluid; Female; Humans; Male; Middle Aged; Multiple System Atrophy; Nerve Degeneration; Neurofibrils; Spinocerebellar Ataxias; Tension-Type Headache

2011
Neuroinflammation and α-synuclein dysfunction potentiate each other, driving chronic progression of neurodegeneration in a mouse model of Parkinson's disease.
    Environmental health perspectives, 2011, Volume: 119, Issue:6

    Mechanisms whereby gene-environment interactions mediate chronic, progressive neurodegenerative processes in Parkinson's disease (PD)-the second most common neurodegenerative disease-remain elusive.. We created a two-hit [neuroinflammation and mutant α-synuclein (α-syn) overexpression] animal model to investigate mechanisms through which mutant α-syn and inflammation work in concert to mediate chronic PD neurodegeneration.. We used an intraperitoneal injection of the inflammogen lipopolysaccharide (LPS; 3 × 106 EU/kg) to initiate systemic and brain inflammation in wild-type (WT) mice and transgenic (Tg) mice overexpressing human A53T mutant α-syn. We then evaluated nigral dopaminergic neurodegeneration, α-syn pathology, and neuroinflammation.. After LPS injection, both WT and Tg mice initially displayed indistinguishable acute neuroinflammation; however, only Tg mice developed persistent neuroinflammation, chronic progressive degeneration of the nigrostriatal dopamine pathway, accumulation of aggregated, nitrated α-syn, and formation of Lewy body-like inclusions in nigral neurons. Further mechanistic studies indicated that 4-week infusion of two inhibitors of inducible nitric oxide synthase and NADPH oxidase, major free radical-generating enzymes in activated microglia, blocked nigral α-syn pathology and neurodegeneration in LPS-injected Tg mice.. Microglia-derived oxidative stress bridged neuroinflammation and α-syn pathogenic alteration in mediating chronic PD progression. Our two-hit animal model involving both a genetic lesion and an environmental trigger reproduced key features of PD and demonstrated synergistic effects of genetic predisposition and environmental exposures in the development of PD. The chronic progressive nature of dopaminergic neurodegeneration, which is absent in most existing PD models, makes this new model invaluable for the study of mechanisms of PD progression.

    Topics: alpha-Synuclein; Amidines; Animals; Animals, Genetically Modified; Benzylamines; Disease Models, Animal; Injections, Intraperitoneal; Lewy Bodies; Lipopolysaccharides; Mice; NADPH Oxidases; Nerve Degeneration; Neurodegenerative Diseases; Nitric Oxide Synthase Type II; Onium Compounds; Oxidative Stress; Parkinson Disease; Substantia Nigra

2011
Chronic exposure to manganese decreases striatal dopamine turnover in human alpha-synuclein transgenic mice.
    Neuroscience, 2011, Apr-28, Volume: 180

    Interaction of genetic and environmental factors is likely involved in Parkinson's disease (PD). Mutations and multiplications of alpha-synuclein (α-syn) cause familial PD, and chronic manganese (Mn) exposure can produce an encephalopathy with signs of parkinsonism. We exposed male transgenic C57BL/6J mice expressing human α-syn or the A53T/A30P doubly mutated human α-syn under the tyrosine hydroxylase promoter and non-transgenic littermates to MnCl₂-enriched (1%) or control food, starting at the age of 4 months. Locomotor activity was increased by Mn without significant effect of the transgenes. Mice were sacrificed at the age of 7 or 20 months. Striatal Mn was significantly increased about three-fold in those exposed to MnCl₂. The number of tyrosine hydroxylase positive substantia nigra compacta neurons was significantly reduced in 20 months old mice (-10%), but Mn or transgenes were ineffective (three-way ANOVA with the factors gene, Mn and age). In 7 months old mice, striatal homovanillic acid (HVA)/dopamine (DA) ratios and aspartate levels were significantly increased in control mice with human α-syn as compared to non-transgenic controls (+17 and +11%, respectively); after Mn exposure both parameters were significantly reduced (-16 and -13%, respectively) in human α-syn mice, but unchanged in non-transgenic animals and mice with mutated α-syn (two-way ANOVA with factors gene and Mn). None of the parameters were changed in the 20 months old mice. Single HVA/DA ratios and single aspartate levels significantly correlated across all treatment groups suggesting a causal relationship between the rate of striatal DA metabolism and aspartate release. In conclusion, under our experimental conditions, Mn and human α-syn, wild-type and doubly mutated, did not interact to induce PD-like neurodegenerative changes. However, Mn significantly and selectively interacted with human wild-type α-syn on indices of striatal DA neurotransmission, the neurotransmitter most relevant to PD.

    Topics: alpha-Synuclein; Animals; Blotting, Western; Chlorides; Chromatography, High Pressure Liquid; Corpus Striatum; Dopamine; Humans; Immunohistochemistry; Manganese Compounds; Mice; Mice, Inbred C57BL; Mice, Transgenic; Motor Activity; Nerve Degeneration; Parkinsonian Disorders; Synaptic Transmission

2011
Progressive neurodegeneration or endogenous compensation in an animal model of Parkinson's disease produced by decreasing doses of alpha-synuclein.
    PloS one, 2011, Mar-07, Volume: 6, Issue:3

    The pathological hallmarks of Parkinson's disease (PD) are degeneration of dopamine (DA) neurons of the substantia nigra (SN) and the presence of alpha-synuclein (α-syn)-rich Lewy bodies in DA cells that remain. To model these aspects of the disease, we previously showed that high titer (5.1×10exp12 gp/ml) AAV1/2 driven expression of A53T α-syn in the SN of rats caused nigrostriatal pathology including a loss of DA neurons, but also with toxicity in the GFP control group. In the current study, we evaluate the effects of two lower titers by dilution of the vector (1∶3 [1.7×10exp12] and 1∶10 [5.1×10exp11]) to define a concentration that produced pathology specific for α-syn. In GFP and empty vector groups there were no behavioural or post-mortem changes at 3 or 6 weeks post-administration at either vector dose. Dilution of the AAV1/2 A53T α-syn (1:3) produced significant paw use asymmetry, reductions in striatal tyrosine hydroxylase (TH), and increases in DA turnover at 3 weeks in the absence of overt pathology. By 6 weeks greater evidence of pathology was observed and included, reductions in SN DA neurons, striatal DA, TH and DA-transporter, along with a sustained behavioural deficit. In contrast, the 1:10 AAV1/2 A53T α-syn treated animals showed normalization between 3 and 6 weeks in paw use asymmetry, reductions in striatal TH, and increased DA turnover. Progression of dopaminergic deficits using the 1:3 titer of AAV1/2 A53Tα-syn provides a platform for evaluating treatments directed at preventing and/or reversing synucleinopathy. Use of the 1:10 titer of AAV1/2 A53T α-syn provides an opportunity to study mechanisms of endogenous compensation. Furthermore, these data highlight the need to characterize the titer of vector being utilized, when using AAV to express pathogenic proteins and model disease process, to avoid producing non-specific effects.

    Topics: alpha-Synuclein; Animals; Axons; Cell Count; Dependovirus; Disease Models, Animal; Dopamine Plasma Membrane Transport Proteins; Female; Forelimb; Genetic Therapy; Genetic Vectors; Neostriatum; Nerve Degeneration; Parkinson Disease; Prosencephalon; Protein Transport; Rats; Rats, Sprague-Dawley; Substantia Nigra; Transgenes; Tyrosine 3-Monooxygenase

2011
Passive immunization reduces behavioral and neuropathological deficits in an alpha-synuclein transgenic model of Lewy body disease.
    PloS one, 2011, Apr-29, Volume: 6, Issue:4

    Dementia with Lewy bodies (DLB) and Parkinson's Disease (PD) are common causes of motor and cognitive deficits and are associated with the abnormal accumulation of alpha-synuclein (α-syn). This study investigated whether passive immunization with a novel monoclonal α-syn antibody (9E4) against the C-terminus (CT) of α-syn was able to cross into the CNS and ameliorate the deficits associated with α-syn accumulation. In this study we demonstrate that 9E4 was effective at reducing behavioral deficits in the water maze, moreover, immunization with 9E4 reduced the accumulation of calpain-cleaved α-syn in axons and synapses and the associated neurodegenerative deficits. In vivo studies demonstrated that 9E4 traffics into the CNS, binds to cells that display α-syn accumulation and promotes α-syn clearance via the lysosomal pathway. These results suggest that passive immunization with monoclonal antibodies against the CT of α-syn may be of therapeutic relevance in patients with PD and DLB.

    Topics: alpha-Synuclein; Animals; Antibodies, Monoclonal; Behavior, Animal; Cell Line, Tumor; Disease Models, Animal; Immunization, Passive; Immunohistochemistry; Lewy Body Disease; Lysosomes; Maze Learning; Mice; Mice, Transgenic; Nerve Degeneration; Rats

2011
Synphilin suppresses α-synuclein neurotoxicity in a Parkinson's disease Drosophila model.
    Genesis (New York, N.Y. : 2000), 2011, Volume: 49, Issue:5

    Parkinson's disease (PD) is the second most common neurodegenerative disorder in humans. It affects 1% of the population over 65-years old. Its causes are environmental and genetic. As the world population ages, there is an urgent need for better and more detailed animal models for this kind of disease. In this work we show that the use of transgenic Drosophila is comparable to more complicated and costly animal models such as mice. The Drosophila model behaves very similar to the equivalent transgenic mice model. We show that both Synphilin-1 and α-synuclein are toxic by themselves, but when co-expressed, they suppress their toxicity reciprocally. Importantly, the symptoms induced in the fly can be treated and partially reverted using standard PD pharmacological treatments. This work showcases Drosophila as a detailed and multifaceted model for Parkinson's disease, providing a convenient platform in which to study and find new genetic modifiers of PD. genesis 49:392-402, 2011.

    Topics: alpha-Synuclein; Animals; Animals, Genetically Modified; Antiparkinson Agents; Blotting, Western; Carbidopa; Carrier Proteins; Disease Models, Animal; Drosophila; Female; Humans; Kaplan-Meier Estimate; Levodopa; Male; Motor Activity; Nerve Degeneration; Nerve Tissue Proteins; Neurotoxicity Syndromes; Parkinson Disease; Reverse Transcriptase Polymerase Chain Reaction

2011
Melittin restores proteasome function in an animal model of ALS.
    Journal of neuroinflammation, 2011, Jun-20, Volume: 8

    Amyotrophic lateral sclerosis (ALS) is a paralyzing disorder characterized by the progressive degeneration and death of motor neurons and occurs both as a sporadic and familial disease. Mutant SOD1 (mtSOD1) in motor neurons induces vulnerability to the disease through protein misfolding, mitochondrial dysfunction, oxidative damage, cytoskeletal abnormalities, defective axonal transport- and growth factor signaling, excitotoxicity, and neuro-inflammation.Melittin is a 26 amino acid protein and is one of the components of bee venom which is used in traditional Chinese medicine to inhibit of cancer cell proliferation and is known to have anti-inflammatory and anti-arthritic effects.The purpose of the present study was to determine if melittin could suppress motor neuron loss and protein misfolding in the hSOD1G93A mouse, which is commonly used as a model for inherited ALS. Meltittin was injected at the 'ZuSanLi' (ST36) acupuncture point in the hSOD1G93A animal model. Melittin-treated animals showed a decrease in the number of microglia and in the expression level of phospho-p38 in the spinal cord and brainstem. Interestingly, melittin treatment in symptomatic ALS animals improved motor function and reduced the level of neuron death in the spinal cord when compared to the control group. Furthermore, we found increased of α-synuclein modifications, such as phosphorylation or nitration, in both the brainstem and spinal cord in hSOD1G93A mice. However, melittin treatment reduced α-synuclein misfolding and restored the proteasomal activity in the brainstem and spinal cord of symptomatic hSOD1G93A transgenic mice.Our research suggests a potential functional link between melittin and the inhibition of neuroinflammation in an ALS animal model.

    Topics: alpha-Synuclein; Amyotrophic Lateral Sclerosis; Animals; Behavior, Animal; Brain Stem; Cell Death; Disease Models, Animal; Humans; Male; Melitten; Mice; Mice, Transgenic; Motor Activity; Nerve Degeneration; Proteasome Endopeptidase Complex; Protein Folding; Rotarod Performance Test; Spinal Cord; Superoxide Dismutase; Survival Rate

2011
Pros and cons of a prion-like pathogenesis in Parkinson's disease.
    BMC neurology, 2011, Jun-20, Volume: 11

    Parkinson's disease (PD) is a slowly progressive neurodegenerative disorder which affects widespread areas of the brainstem, basal ganglia and cerebral cortex. A number of proteins are known to accumulate in parkinsonian brains including ubiquitin and α-synuclein. Prion diseases are sporadic, genetic or infectious disorders with various clinical and histopathological features caused by prion proteins as infectious proteinaceous particles transmitting a misfolded protein configuration through brain tissue. The most important form is Creutzfeldt-Jakob disease which is associated with a self-propagating pathological precursor form of the prion protein that is physiologically widely distributed in the central nervous system.. It has recently been found that α-synuclein may behave similarly to the prion precursor and propagate between cells. The post-mortem proof of α-synuclein containing Lewy bodies in embryonic dopamine cells transplants in PD patient suggests that the misfolded protein might be transmitted from the diseased host to donor neurons reminiscent of prion behavior. The involvement of the basal ganglia and brainstem in the degenerative process are other congruencies between Parkinson's and Creutzfeldt-Jakob disease. However, a number of issues advise caution before categorizing Parkinson's disease as a prion disorder, because clinical appearance, brain imaging, cerebrospinal fluid and neuropathological findings exhibit fundamental differences between both disease entities. Most of all, infectiousness, a crucial hallmark of prion diseases, has never been observed in PD so far. Moreover, the cellular propagation of the prion protein has not been clearly defined and it is, therefore, difficult to assess the molecular similarities between the two disease entities.. At the current state of knowledge, the molecular pathways of transmissible pathogenic proteins are not yet fully understood. Their exact involvement in the pathophysiology of prion disorders and neurodegenerative diseases has to be further investigated in order to elucidate a possible overlap between both disease categories that are currently regarded as distinct entities.

    Topics: alpha-Synuclein; Humans; Nerve Degeneration; Parkinson Disease; Prions

2011
Genetic activation of Nrf2 signaling is sufficient to ameliorate neurodegenerative phenotypes in a Drosophila model of Parkinson's disease.
    Disease models & mechanisms, 2011, Volume: 4, Issue:5

    Parkinson's disease (PD) is the most common neurodegenerative movement disorder. Oxidative stress has been associated with the etiology of both sporadic and monogenic forms of PD. The transcription factor Nrf2, a conserved global regulator of cellular antioxidant responses, has been implicated in neuroprotection against PD pathology. However, direct evidence that upregulation of the Nrf2 pathway is sufficient to confer neuroprotection in genetic models of PD is lacking. Expression of the PD-linked gene encoding α-synuclein in dopaminergic neurons of Drosophila results in decreased locomotor activity and selective neuron loss in a progressive age-dependent manner, providing a genetically accessible model of PD. Here we show that upregulation of the Nrf2 pathway by overexpressing Nrf2 or its DNA-binding dimerization partner, Maf-S, restores the locomotor activity of α-synuclein-expressing flies. Similar benefits are observed upon RNA-interference-mediated downregulation of the prime Nrf2 inhibitor, Keap1, as well as in conditions of keap1 heterozygosity. Consistently, the α-synuclein-induced dopaminergic neuron loss is suppressed by Maf-S overexpression or keap1 heterozygosity. Our data validate the sustained upregulation of the Nrf2 pathway as a neuroprotective strategy against PD. This model provides a genetically accessible in vivo system in which to evaluate the potential of additional Nrf2 pathway components and regulators as therapeutic targets.

    Topics: alpha-Synuclein; Animals; Disease Models, Animal; Dopaminergic Neurons; Drosophila melanogaster; Drosophila Proteins; Intracellular Signaling Peptides and Proteins; Kelch-Like ECH-Associated Protein 1; Locomotion; Nerve Degeneration; NF-E2-Related Factor 2; Parkinson Disease; Phenotype; Signal Transduction; Transcriptional Activation; Transgenes

2011
α-Syn suppression reverses synaptic and memory defects in a mouse model of dementia with Lewy bodies.
    The Journal of neuroscience : the official journal of the Society for Neuroscience, 2011, Jul-06, Volume: 31, Issue:27

    Abnormally accumulated α-synuclein (α-syn) is a pathological hallmark of Lewy body-related disorders such as Parkinson's disease (PD) and dementia with Lewy body disease (DLB). However, it is not well understood whether and how abnormal accumulation of α-syn leads to cognitive impairment or dementia in PD and DLB. Furthermore, it is not known whether targeted removal of α-syn pathology can reverse cognitive decline. Here, we found that the distribution of α-syn pathology in an inducible α-syn transgenic mouse model recapitulates that in human DLB. Abnormal accumulation of α-syn in the limbic system, particularly in the hippocampus, correlated with memory impairment and led to structural synaptic deficits. Furthermore, when α-syn expression was suppressed, we observed partial clearing of pre-existing α-syn pathology and reversal of structural synaptic defects, resulting in an improvement in memory function.

    Topics: Acoustic Stimulation; Age Factors; alpha-Synuclein; Analysis of Variance; Animals; Animals, Newborn; Brain; Calcium-Calmodulin-Dependent Protein Kinase Type 2; Conditioning, Classical; Cues; Disease Models, Animal; Disease Progression; Embryo, Mammalian; Fear; Female; Gene Expression Regulation; Glial Fibrillary Acidic Protein; Gliosis; Humans; In Vitro Techniques; Indoles; Lewy Body Disease; Male; Memory Disorders; Mice; Mice, Inbred C57BL; Mice, Transgenic; Mutation; Nerve Degeneration; Nerve Tissue Proteins; Serine; Synapses

2011
Parkin-mediated protection of dopaminergic neurons in a chronic MPTP-minipump mouse model of Parkinson disease.
    Journal of neuropathology and experimental neurology, 2011, Volume: 70, Issue:8

    Loss-of-function mutations in the ubiquitin ligase parkin are the major cause of recessively inherited early-onset Parkinson disease (PD). Impairment of parkin activity caused by nitrosative or dopamine-related modifications may also be responsible for the loss of dopaminergic (DA) neurons in sporadic PD. Previous studies have shown that viral vector-mediated delivery of parkin prevented DA neurodegeneration in several animal models, but little is known about the neuroprotective actions of parkin in vivo. Here, we investigated mechanisms of neuroprotection of overexpressed parkin in a modified long-term mouse model of PD using osmotic minipump administration of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Recombinant adeno-associated viral vector-mediated intranigral delivery of parkin prevented motor deficits and DA cell loss in the mice. Ser129-phosphorylated α-synuclein-immunoreactive cells were increased in the substantia nigra of parkin-treated mice. Moreover, delivery of parkin alleviated the MPTP-induced decrease of the active phosphorylated form of Akt. On the other hand, upregulation of p53 and mitochondrial alterations induced by chronic MPTP administration were barely suppressed by parkin. These results suggest that the neuroprotective actions of parkin may be impaired in severe PD.

    Topics: 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine; alpha-Synuclein; Analysis of Variance; Animals; bcl-2-Associated X Protein; Cell Count; Chromatography, High Pressure Liquid; Corpus Striatum; Disease Models, Animal; Dopamine; Gene Expression Regulation; Genetic Vectors; Green Fluorescent Proteins; Humans; Male; Mesencephalon; Mice; Mice, Inbred C57BL; Microinjections; Motor Activity; MPTP Poisoning; Nerve Degeneration; Neurons; Neurotoxins; Oncogene Protein v-akt; Rotarod Performance Test; Signal Transduction; Statistics as Topic; Transduction, Genetic; Tumor Suppressor Protein p53; Ubiquitin-Protein Ligases

2011
Measurement of proteasomal dysfunction in cell models of dopaminergic degeneration.
    Methods in molecular biology (Clifton, N.J.), 2011, Volume: 758

    Parkinson's disease (PD) is the second most common neurodegenerative diseases, which occurs in both inheritable and sporadic forms. The interplay of the genetic mutations and environmental exposure to disease risk factors contributes to the pathogenic events leading to the demise of dopaminergic neurons in PD. Proteasome is one of the major proteolytic machinery responsible for degrading unwanted and damaged intracellular proteins. Emerging evidence implicates the incomplete proteolysis by ubiquitin-proteasome system (UPS) in PD pathogenesis. Proteasome inhibition recapitulates some of the key features of PD in vivo and in vitro. Varieties of dopaminergic neurotoxins emerge to inhibit proteasomal function. Given that some PD-related gene mutations impair proteolytic function of UPS, it has been well-accepted that both genetic and environmental factors may conspire to compromise the UPS in the initiation and progression of the disease. The enzymatic assays for the proteasomal activities with fluorogenic substrates and western blot analysis of ubiquitinated proteins provide an entry point to determine UPS function in the process of dopaminergic degeneration.

    Topics: alpha-Synuclein; Animals; Blotting, Western; Cell Extracts; Cell Line; Cells, Cultured; Dopaminergic Neurons; Electrophoresis, Polyacrylamide Gel; Enzyme Assays; Microscopy, Confocal; Nerve Degeneration; Parkinson Disease; Proteasome Endopeptidase Complex; Protein Transport; Rats; Ubiquitin; Ubiquitination

2011
α-Synuclein deficiency and efferent nerve degeneration in the mouse cochlea: a possible cause of early-onset presbycusis.
    Neuroscience research, 2011, Volume: 71, Issue:3

    Efferent nerves under the outer hair cells (OHCs) play a role in the protection of these cells from loud stimuli. Previously, we showed that cochlear α-synuclein expression is localized to efferent auditory synapses at the base of the OHCs. To prove our hypothesis that α-synuclein deficiency and efferent auditory deficit might be a cause of hearing loss, we compared the morphology of efferent nerve endings and α-synuclein expression within the cochleae of two mouse models of presbycusis.. Comparative animal study of presbycusis.. The C57BL/6J(C57) mouse strain, a well-known model of early-onset hearing loss, and the CBA mouse strain, a model of relatively late-onset hearing loss, were examined. Auditory brainstem responses and distortion product otoacoustic emissions were recorded, and cochlear morphology with efferent nerve ending was compared. Western blotting was used to examine α-synuclein expression in the cochlea.. Compared with CBA mice, C57 mice showed earlier onset high-frequency hearing loss and decreased function in OHCs, especially within high-frequency regions. C57 mice demonstrated more severe pathologic changes within the cochlea, particularly within the basal turn, than CBA mice of the same age. Weaker α-synuclein and synaptophysin expression in the efferent nerve endings and cochlear homogenates in C57 mice was observed.. Our results support the hypothesis that efferent nerve degeneration, possibly due to differential α-synuclein expression, is a potential cause of early-onset presbycusis. Further studies at the cellular level are necessary to verify our results.

    Topics: Age of Onset; alpha-Synuclein; Animals; Cochlea; Disease Models, Animal; Disease Progression; Efferent Pathways; Hair Cells, Auditory, Outer; Male; Mice; Mice, Inbred C57BL; Mice, Inbred CBA; Nerve Degeneration; Presbycusis

2011
Apolipoprotein E and LRP1 Increase Early in Parkinson's Disease Pathogenesis.
    The American journal of pathology, 2011, Volume: 179, Issue:5

    Parkinson's disease (PD) is characterized by α-synuclein-containing Lewy bodies (LBs) and loss of melanized neurons in the substantia nigra (SN). Recently, a link between apolipoprotein E (ApoE) expression, α-synuclein aggregation, and neurodegeneration was suggested. Here, we report on ApoE expression appearing in melanized neurons of the SN and in LBs in both PD and incidental LB disease cases. Interestingly, increased expression of the low-density lipoprotein receptor-related protein 1 (the receptor for ApoE) was also observed in incidental LB disease and PD. Our data suggest that alterations in lipoprotein homeostasis/signaling in melanized neurons of the SN are an early event during PD pathogenesis.

    Topics: Adult; Aged; Aged, 80 and over; alpha-Synuclein; Apolipoproteins E; Cadaver; Female; Homeostasis; Humans; Immunohistochemistry; Lewy Bodies; Low Density Lipoprotein Receptor-Related Protein-1; Male; Melanins; Middle Aged; Nerve Degeneration; Neurons; Parkinson Disease; Substantia Nigra

2011
Exogenous α-synuclein fibrils induce Lewy body pathology leading to synaptic dysfunction and neuron death.
    Neuron, 2011, Oct-06, Volume: 72, Issue:1

    Inclusions composed of α-synuclein (α-syn), i.e., Lewy bodies (LBs) and Lewy neurites (LNs), define synucleinopathies including Parkinson's disease (PD) and dementia with Lewy bodies (DLB). Here, we demonstrate that preformed fibrils generated from full-length and truncated recombinant α-syn enter primary neurons, probably by adsorptive-mediated endocytosis, and promote recruitment of soluble endogenous α-syn into insoluble PD-like LBs and LNs. Remarkably, endogenous α-syn was sufficient for formation of these aggregates, and overexpression of wild-type or mutant α-syn was not required. LN-like pathology first developed in axons and propagated to form LB-like inclusions in perikarya. Accumulation of pathologic α-syn led to selective decreases in synaptic proteins, progressive impairments in neuronal excitability and connectivity, and, eventually, neuron death. Thus, our data contribute important insights into the etiology and pathogenesis of PD-like α-syn inclusions and their impact on neuronal functions, and they provide a model for discovering therapeutics targeting pathologic α-syn-mediated neurodegeneration.

    Topics: alpha-Synuclein; Animals; Axonal Transport; Cell Death; Endocytosis; Hippocampus; Humans; Lewy Bodies; Mice; Mice, Inbred C57BL; Nerve Degeneration; Nerve Tissue Proteins; Neurites; Neurons; Primary Cell Culture; Synapses; Voltage-Sensitive Dye Imaging

2011
Differential regional expression patterns of α-synuclein, TNF-α, and IL-1β; and variable status of dopaminergic neurotoxicity in mouse brain after Paraquat treatment.
    Journal of neuroinflammation, 2011, Nov-24, Volume: 8

    Paraquat (1, 1-dimethyl-4, 4-bipyridium dichloride; PQ) causes neurotoxicity, especially dopaminergic neurotoxicity, and is a supposed risk factor for Parkinson's disease (PD). However, the cellular and molecular mechanisms of PQ-induced neurodegeneration are far from clear. Previous studies have shown that PQ induces neuroinflammation and dopaminergic cell loss, but the prime cause of those events is still in debate.. We examined the neuropathological effects of PQ not only in substantia nigra (SN) but also in frontal cortex (FC) and hippocampus of the progressive mouse (adult Swiss albino) model of PD-like neurodegeneration, using immunohistochemistry, western blots, and histological and biochemical analyses.. PQ caused differential patterns of changes in cellular morphology and expression of proteins related to PD and neuroinflammation in the three regions examined (SN, FC and hippocampus). Coincident with behavioral impairment and brain-specific ROS generation, there was differential immunolocalization and decreased expression levels of tyrosine hydroxylase (TH) in the three regions, whereas α-synuclein immunopositivity increased in hippocampus, increased in FC and decreased in SN. PQ-induced neuroinflammation was characterized by area-specific changes in localization and appearances of microglial cells with or without activation and increment in expression patterns of tumor necrosis factor-α in the three regions of mouse brain. Expression of interleukin-1β was increased in FC and hippocampus but not significantly changed in SN.. The present study demonstrates that PQ induces ROS production and differential α-synuclein expression that promotes neuroinflammation in microglia-dependent or -independent manners, and produces different patterns of dopaminergic neurotoxicity in three different regions of mouse brain.

    Topics: alpha-Synuclein; alpha-Tocopherol; Animals; Antioxidants; Behavior, Animal; Catalase; DNA-Binding Proteins; Dopaminergic Neurons; Dose-Response Relationship, Drug; Frontal Lobe; Glutathione Transferase; Herbicides; Hippocampus; Humans; Interleukin-1beta; Male; Mice; Nerve Degeneration; Nerve Tissue Proteins; Nuclear Proteins; Paraquat; Random Allocation; Reactive Oxygen Species; Substantia Nigra; Superoxide Dismutase; Survival Rate; Tumor Necrosis Factor-alpha; Tyrosine 3-Monooxygenase

2011
[Multiple system atrophy - synuclein and neuronal degeneration].
    Rinsho shinkeigaku = Clinical neurology, 2011, Volume: 51, Issue:11

    Multiple system atrophy (MSA) is a sporadic neurodegenerative disorder that encompasses olivopontocerebellar atrophy (OPCA), striatonigral degeneration (SND) and Shy-Drager syndrome (SDS). The histopathological hallmarks are α-synuclein (AS) positive glial cytoplasmic inclusions (GCIs) in oligodendroglias. AS aggregation is also found in glial nuclear inclusions (GNIs), neuronal cytoplasmic inclusions (NCIs), neuronal nuclear inclusions (NNIs) and dystrophic neurties. Reviewing the pathological features of 102 MSA cases, OPCA-type was relatively more frequent and SND-type was less frequent in Japanese MSA cases, which suggested different phenotypic pattern of MSA might exist between races, compared to the relatively high frequency of SND-type in western countries. In early stage of MSA, NNIs, NCIs and diffuse homogenous stain of AS in neuronal nuclei and cytoplasm were observed in various vulnerable lesions including the pontine nuclei, putamen, substantia nigra, locus ceruleus, inferior olivary nucleus, intermediolateral column of thoracic cord, lower motor neurons and cortical pyramidal neurons, in additions to GCIs. These findings indicated that the primary nonfibrillar and fibrillar AS aggregation also occurred in neurons. Therefore both the direct involvement of neurons themselves and the oligodendroglia-myelin-axon mechanism may synergistically accelerate the degenerative process of MSA.

    Topics: Aged; Aged, 80 and over; alpha-Synuclein; Female; Histocytochemistry; Humans; Male; Middle Aged; Multiple System Atrophy; Nerve Degeneration; Oligodendroglia

2011
VPS41, a protein involved in lysosomal trafficking, is protective in Caenorhabditis elegans and mammalian cellular models of Parkinson's disease.
    Neurobiology of disease, 2010, Volume: 37, Issue:2

    VPS41 is a protein identified as a potential therapeutic target for Parkinson's disease (PD) as a result of a high-throughput RNAi screen in Caenorhabditis elegans. VPS41 has a plausible mechanistic link to the pathogenesis of PD, as in yeast it is known to participate in trafficking of proteins to the lysosomal system and several recent lines of evidence have pointed to the importance of lysosomal system dysfunction in the neurotoxicity of alpha-synuclein (alpha-syn). We found that expression of the human form of VPS41 (hVPS41) prevents dopamine (DA) neuron loss induced by alpha-syn overexpression and 6-hydroxydopamine (6-OHDA) neurotoxicity in C. elegans. In SH-SY5Y neuroblastoma cell lines stably transfected with hVPS41, we determined that presence of this protein conferred protection against the neurotoxins 6-OHDA and rotenone. Overexpression of hVPS41 did not alter the mitochondrial membrane depolarization induced by these neurotoxins. hVPS41 did, however, block downstream events in the apoptotic cascade including activation of caspase-9 and caspase-3, and PARP cleavage. We also observed that hVPS41 reduced the accumulation of insoluble high-molecular weight forms of alpha-syn in SH-SY5Y cells after treatment with rotenone. These data show that hVPS41 is protective against both alpha-syn and neurotoxic-mediated injury in invertebrate and cellular models of PD. These protective functions may be related to enhanced clearance of misfolded or aggregated protein, including alpha-syn. Our studies indicate that hVPS41 may be a useful target for developing therapeutic strategies for human PD.

    Topics: alpha-Synuclein; Animals; Apoptosis; Caenorhabditis elegans; Caspases; Cell Line; Cytoprotection; Dopamine; Humans; Lysosomes; Nerve Degeneration; Neurons; Neurotoxins; Parkinson Disease; Protein Transport; Saccharomyces cerevisiae Proteins; Uncoupling Agents; Vesicular Transport Proteins

2010
Degeneration of dopaminergic neurons induced by thrombin injection in the substantia nigra of the rat is enhanced by dexamethasone: role of monoamine oxidase enzyme.
    Neurotoxicology, 2010, Volume: 31, Issue:1

    Anti-inflammatory strategies receive growing attention for their potential to prevent pathological deterioration in disorders such as Parkinson's disease, which is accompanied by inflammatory reactions that might play a critical role in the degeneration of nigral dopaminergic neurons. We investigated the influence of dexamethasone - a potent synthetic member of the glucocorticoids class of steroid hormones that acts as an anti-inflammatory - on the degeneration of the dopaminergic neurons of rats observed after intranigral injection of thrombin, a serine protease that induces inflammation through microglia proliferation and activation. We evaluated tyrosine hydroxylase (TH)-positive neurons as well as astroglial and microglial populations; dexamethasone prevented the loss of astrocytes but was unable to stop microglial proliferation induced by thrombin. Moreover, dexamethasone produced alterations in the levels of nexin and the thrombin receptor PAR-1, and facilitated accumulation of alpha-synuclein induced by thrombin in dopaminergic neurons. Dexamethasone increased oxidative stress and expression of monoamine oxidase A and B, along with changes on different MAP kinases related to degenerative processes, resulting in a bigger loss of dopaminergic neurons after intranigral injection of thrombin in dexamethasone-treated animals. It is interesting to ascertain that inhibition of monoamine oxidase by tranylcypromine prevented neurodegeneration of dopaminergic neurons, thus suggesting that the deleterious effects of dexamethasone might be mediated by monoamine oxidase.

    Topics: alpha-Synuclein; Analysis of Variance; Animals; Dexamethasone; Drug Synergism; Female; Glial Fibrillary Acidic Protein; Glucocorticoids; Histocompatibility Antigens; Hydrazines; In Situ Nick-End Labeling; Monoamine Oxidase; Monoamine Oxidase Inhibitors; Nerve Degeneration; Neuroglia; Neurons; Nitric Oxide Synthase; Oncogene Protein v-akt; Rats; Rats, Wistar; Receptors, Thrombin; Spectrophotometry; Substantia Nigra; Sulfonamides; Thrombin; Time Factors; Tranylcypromine; Tyrosine 3-Monooxygenase

2010
Dysfunctional mitochondria uphold calpain activation: contribution to Parkinson's disease pathology.
    Neurobiology of disease, 2010, Volume: 37, Issue:3

    Calpain is a ubiquitous calcium-sensitive protease that is essential for normal physiologic neuronal function. However, mitochondrial-mediated-calcium homeostasis alterations may lead to its pathologic activation that jeopardizes neuronal structure and function. Here, we provide evidence to support a role for the involvement of calpain 1 in mitochondrial-induced neurodegeneration in a Parkinson's disease (PD) cellular model. We show that dysfunctional mitochondria increases cytosolic calcium, thereby, inducing calpain activation. Interestingly, its inhibition significantly attenuated the accumulation of alpha-synuclein oligomers and contributed to an increase of insoluble alpha-synuclein aggregates, known to be cytoprotective. Moreover, our data corroborate that calpain-1 overactivation in our mitochondrial-deficient cells promote caspase-3 activation. Overall, our findings further clarify the crucial role of dysfunctional mitochondria in the control of molecular mechanisms occurring in PD brain cells, providing a potentially novel correlation between the degradation of calpain substrates suggesting a putative role of calpain and calpain inhibition as a therapeutic tool in PD.

    Topics: alpha-Synuclein; Calcium; Calcium Signaling; Calpain; Caspase 3; Cell Line, Transformed; Enzyme Activation; Humans; Inclusion Bodies; Mitochondria; Mitochondrial Diseases; Models, Biological; Nerve Degeneration; Parkinson Disease

2010
Vulnerability of peripheral catecholaminergic neurons to MPTP is not regulated by alpha-synuclein.
    Neurobiology of disease, 2010, Volume: 38, Issue:1

    Although generally considered a prototypical movement disorder, Parkinson's disease is commonly associated with a broad-spectrum of non-motor symptoms, including autonomic dysfunctions caused by significant alterations in catecholaminergic neurons of the peripheral sympathetic nervous system. Here we present evidence that alpha-synuclein is highly expressed by sympathetic ganglion neurons throughout embryonic and postnatal life and that it is found in tyrosine hydroxylase-positive sympathetic fibers innervating the heart of adult mice. However, mice deficient in alpha-synuclein do not exhibit any apparent alterations in sympathetic development. Sympathetic neurons isolated from mouse embryos and early postnatal mice are sensitive to the parkinsonian drug MPTP/MPP(+) and intoxication requires entry of the neurotoxin through the noradrenaline transporter. Furthermore, recovery of noradrenaline from cardiac sympathetic fibers is reduced in adult mice treated with MPTP systemically. However, MPP(+)-induced sympathetic neuron loss in vitro or MPTP-induced cardiac noradrenaline depletion in vivo is not modified in mice lacking alpha-synuclein. This is in clear contrast with the observation that dopaminergic neurons of the central nervous system are significantly less vulnerable to MPTP/MPP(+) in the absence of alpha-synuclein, suggesting different actions of this molecule in central and peripheral catecholaminergic neurons.

    Topics: 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine; 1-Methyl-4-phenylpyridinium; alpha-Synuclein; Animals; Catecholamines; Cell Death; Cells, Cultured; Ganglia, Sympathetic; Mice; Mice, Mutant Strains; Nerve Degeneration; Neurons; Neurotoxins; Parkinsonian Disorders; Sympathetic Fibers, Postganglionic; Tyrosine 3-Monooxygenase

2010
Inhibition of FK506 binding proteins reduces alpha-synuclein aggregation and Parkinson's disease-like pathology.
    The Journal of neuroscience : the official journal of the Society for Neuroscience, 2010, Feb-17, Volume: 30, Issue:7

    alpha-Synuclein (alpha-SYN) is a key player in the pathogenesis of Parkinson's disease (PD). In pathological conditions, the protein is present in a fibrillar, aggregated form inside cytoplasmic inclusions called Lewy bodies. Members of the FK506 binding protein (FKBP) family are peptidyl-prolyl isomerases that were shown recently to accelerate the aggregation of alpha-SYN in vitro. We now established a neuronal cell culture model for synucleinopathy based on oxidative stress-induced alpha-SYN aggregation and apoptosis. Using high-content analysis, we examined the role of FKBPs in aggregation and apoptotic cell death. FK506, a specific inhibitor of this family of proteins, inhibited alpha-SYN aggregation and neuronal cell death in this synucleinopathy model dose dependently. Knockdown of FKBP12 or FKBP52 reduced the number of alpha-SYN aggregates and protected against cell death, whereas overexpression of FKBP12 or FKBP52 accelerated both aggregation of alpha-SYN and cell death. Thus, FK506 likely targets FKBP members in the cell culture model. Furthermore, oral administration of FK506 after viral vector-mediated overexpression of alpha-SYN in adult mouse brain significantly reduced alpha-SYN aggregate formation and neuronal cell death. Our data explain previously described neuroregenerative and neuroprotective effects of immunophilin ligands and validate FKBPs as a novel drug target for the causative treatment of PD.

    Topics: Age Factors; alpha-Synuclein; Animals; Apoptosis; Cells, Cultured; Corpus Striatum; Disease Models, Animal; Humans; Indoles; Intermediate Filament Proteins; Luciferases; Mice; Mice, Inbred C57BL; Mice, Transgenic; Mutation; Nerve Degeneration; Neuroblastoma; Parkinson Disease; RNA, Small Interfering; Tacrolimus Binding Protein 1A; Tacrolimus Binding Proteins; Time Factors; Transfection

2010
Selective molecular alterations in the autophagy pathway in patients with Lewy body disease and in models of alpha-synucleinopathy.
    PloS one, 2010, Feb-19, Volume: 5, Issue:2

    Lewy body disease is a heterogeneous group of neurodegenerative disorders characterized by alpha-synuclein accumulation that includes dementia with Lewy bodies (DLB) and Parkinson's Disease (PD). Recent evidence suggests that impairment of lysosomal pathways (i.e. autophagy) involved in alpha-synuclein clearance might play an important role. For this reason, we sought to examine the expression levels of members of the autophagy pathway in brains of patients with DLB and Alzheimer's Disease (AD) and in alpha-synuclein transgenic mice.. By immunoblot analysis, compared to controls and AD, in DLB cases levels of mTor were elevated and Atg7 were reduced. Levels of other components of the autophagy pathway such as Atg5, Atg10, Atg12 and Beclin-1 were not different in DLB compared to controls. In DLB brains, mTor was more abundant in neurons displaying alpha-synuclein accumulation. These neurons also showed abnormal expression of lysosomal markers such as LC3, and ultrastructural analysis revealed the presence of abundant and abnormal autophagosomes. Similar alterations were observed in the brains of alpha-synuclein transgenic mice. Intra-cerebral infusion of rapamycin, an inhibitor of mTor, or injection of a lentiviral vector expressing Atg7 resulted in reduced accumulation of alpha-synuclein in transgenic mice and amelioration of associated neurodegenerative alterations.. This study supports the notion that defects in the autophagy pathway and more specifically in mTor and Atg7 are associated with neurodegeneration in DLB cases and alpha-synuclein transgenic models and supports the possibility that modulators of the autophagy pathway might have potential therapeutic effects.

    Topics: Aged; Aged, 80 and over; alpha-Synuclein; Alzheimer Disease; Animals; Autophagy; Autophagy-Related Protein 7; Blotting, Western; Brain; Cell Line; Cell Line, Tumor; Disease Models, Animal; Female; Humans; Lewy Body Disease; Male; Mice; Mice, Transgenic; Microscopy, Confocal; Microscopy, Electron; Nerve Degeneration; Pyramidal Cells; RNA Interference; Signal Transduction; Ubiquitin-Activating Enzymes

2010
Alpha-synuclein disrupted dopamine homeostasis leads to dopaminergic neuron degeneration in Caenorhabditis elegans.
    PloS one, 2010, Feb-19, Volume: 5, Issue:2

    Disruption of dopamine homeostasis may lead to dopaminergic neuron degeneration, a proposed explanation for the specific vulnerability of dopaminergic neurons in Parkinson's disease. While expression of human alpha-synuclein in C. elegans results in dopaminergic neuron degeneration, the effects of alpha-synuclein on dopamine homeostasis and its contribution to dopaminergic neuron degeneration in C. elegans have not been reported. Here, we examined the effects of alpha-synuclein overexpression on worm dopamine homeostasis. We found that alpha-synuclein expression results in upregulation of dopamine synthesis and content, and redistribution of dopaminergic synaptic vesicles, which significantly contribute to dopaminergic neuron degeneration. These results provide in vivo evidence supporting a critical role for dopamine homeostasis in supporting dopaminergic neuron integrity.

    Topics: 3,4-Dihydroxyphenylacetic Acid; alpha-Synuclein; Animals; Animals, Genetically Modified; Caenorhabditis elegans; Dopamine; Female; Green Fluorescent Proteins; Homeostasis; Humans; Male; Microscopy, Confocal; Motor Activity; Nerve Degeneration; Synaptic Vesicles; Vesicular Monoamine Transport Proteins

2010
Synphilin-1 attenuates neuronal degeneration in the A53T alpha-synuclein transgenic mouse model.
    Human molecular genetics, 2010, Jun-01, Volume: 19, Issue:11

    Genetic alterations in alpha-synuclein cause autosomal dominant familial Parkinsonism and may contribute to sporadic Parkinson's disease (PD). Synphilin-1 is an alpha-synuclein-interacting protein, with implications in PD pathogenesis related to protein aggregation. Currently, the in vivo role of synphilin-1 in alpha-synuclein-linked pathogenesis is not fully understood. Using the mouse prion protein promoter, we generated synphilin-1 transgenic mice, which did not display PD-like phenotypes. However, synphilin-1/A53T alpha-synuclein double-transgenic mice survived longer than A53T alpha-synuclein single-transgenic mice. There were attenuated A53T alpha-synuclein-induced motor abnormalities and decreased astroglial reaction and neuronal degeneration in brains in double-transgenic mice. Overexpression of synphilin-1 decreased caspase-3 activation, increased beclin-1 and LC3 II expression and promoted formation of aggresome-like structures, suggesting that synphilin-1 alters multiple cellular pathways to protect against neuronal degeneration. These studies demonstrate that synphilin-1 can diminish the severity of alpha-synucleinopathy and play a neuroprotective role against A53T alpha-synuclein toxicity in vivo.

    Topics: alpha-Synuclein; Analysis of Variance; Animals; Apoptosis Regulatory Proteins; Beclin-1; Brain; Carrier Proteins; Caspase 3; Immunoblotting; Immunohistochemistry; Intracellular Signaling Peptides and Proteins; Lewy Bodies; Mice; Mice, Transgenic; Mutation, Missense; Nerve Degeneration; Nerve Tissue Proteins; Parkinson Disease

2010
Upregulation of alpha-synuclein expression in the rat cerebellum in experimental hepatic encephalopathy.
    Neuropathology and applied neurobiology, 2010, Volume: 36, Issue:5

    The overexpression of alpha-synuclein has been associated with neurodegenerative diseases, especially when the protein aggregates to form insoluble structures. The present study examined the effect of chronic hyperammonaemia on alpha-synuclein expression in the rat cerebellum following portacaval anastomosis (PCA).. Immunohistochemical and western blot determinations were performed 1 month and 6 months after the PCA procedure.. A time-dependent increase in alpha-synuclein expression was seen in the cerebellar grey matter compared with the controls. At 1 month post PCA, alpha-synuclein-immunopositive material was observed in the molecular layer, while the Purkinje cells showed weak alpha-synuclein expression, and alpha-synuclein aggregates were observed throughout the granular layer. At 6 months post PCA, alpha-synuclein expression was significantly increased compared with the controls. alpha-synuclein-immunostained astroglial cells were also found; the Bergmann glial cells showed alpha-synuclein-positive processes in the molecular layer of PCA-exposed rats, and in the granular layer, perivascular astrocytes showed intense alpha-synuclein immunoreactivity, as indicated by colocalization of alpha-synuclein with glial fibrillary acidic protein (GFAP). In addition, ubiquitin-immunoreactive inclusions were present in PCA-exposed rats, although they did not colocalize with alpha-synuclein. Western blotting performed at 6 months post PCA showed a reduction in the level of soluble alpha-synuclein compared with 1 month post PCA and the controls; this reduction was concomitant with an increase in the insoluble form of alpha-synuclein.. Although the precise mechanism by which alpha-synuclein aggregates in PCA-treated rats remains unknown, the present data suggest an important role for this protein in the onset and progression of hepatic encephalopathy, probably via its expression in astroglial cells.

    Topics: alpha-Synuclein; Animals; Astrocytes; Blotting, Western; Cerebellum; Chronic Disease; Disease Models, Animal; Disease Progression; Hepatic Encephalopathy; Hyperammonemia; Immunohistochemistry; Male; Nerve Degeneration; Rats; Rats, Sprague-Dawley; Up-Regulation

2010
Astrocytic expression of Parkinson's disease-related A53T alpha-synuclein causes neurodegeneration in mice.
    Molecular brain, 2010, Apr-21, Volume: 3

    Parkinson's disease (PD) is the most common movement disorder. While neuronal deposition of alpha-synuclein serves as a pathological hallmark of PD and Dementia with Lewy Bodies, alpha-synuclein-positive protein aggregates are also present in astrocytes. The pathological consequence of astrocytic accumulation of alpha-synuclein, however, is unclear.. Here we show that PD-related A53T mutant alpha-synuclein, when selectively expressed in astrocytes, induced rapidly progressed paralysis in mice. Increasing accumulation of alpha-synuclein aggregates was found in presymptomatic and symptomatic mouse brains and correlated with the expansion of reactive astrogliosis. The normal function of astrocytes was compromised as evidenced by cerebral microhemorrhage and down-regulation of astrocytic glutamate transporters, which also led to increased inflammatory responses and microglial activation. Interestingly, the activation of microglia was mainly detected in the midbrain, brainstem and spinal cord, where a significant loss of dopaminergic and motor neurons was observed. Consistent with the activation of microglia, the expression level of cyclooxygenase 1 (COX-1) was significantly up-regulated in the brain of symptomatic mice and in cultured microglia treated with conditioned medium derived from astrocytes over-expressing A53T alpha-synuclein. Consequently, the suppression of COX-1 activities extended the survival of mutant mice, suggesting that excess inflammatory responses elicited by reactive astrocytes may contribute to the degeneration of neurons.. Our findings demonstrate a critical involvement of astrocytic alpha-synuclein in initiating the non-cell autonomous killing of neurons, suggesting the viability of reactive astrocytes and microglia as potential therapeutic targets for PD and other neurodegenerative diseases.

    Topics: alpha-Synuclein; Animals; Astrocytes; Brain; Humans; Mice; Mice, Inbred C57BL; Mice, Transgenic; Mutation; Nerve Degeneration; Neuropsychological Tests; Parkinson Disease; Survival Rate

2010
Drosophila histone deacetylase 6 protects dopaminergic neurons against {alpha}-synuclein toxicity by promoting inclusion formation.
    Molecular biology of the cell, 2010, Jul-01, Volume: 21, Issue:13

    Parkinson's disease (PD) is associated with progressive degeneration of dopaminergic (DA) neurons. We report for the first time that the Drosophila histone deacetylase 6 (dHDAC6) plays a critical role in the protection of DA neurons and the formation of alpha-synuclein inclusions by using a Drosophila PD model constructed by ectopic expression of human alpha-synuclein. Depletion of dHDAC6 significantly enhances the effects caused by ectopic expression of alpha-synuclein, namely, loss of DA neurons, retinal degeneration, and locomotor dysfunction. Expression of alpha-synuclein in the DA neurons leads to fewer inclusions in the brains of dHDAC6 mutant flies than in wild-type flies. Conversely, overexpression of dHDAC6 is able to suppress the alpha-synuclein-induced DA neuron loss and retinal degeneration and promote inclusion formation. Furthermore, mutation of dHDAC6 reinforces the accumulation of oligomers that are suggested to be a toxic form of alpha-synuclein. We propose that alpha-synuclein inclusion formation in the presence of dHDAC6 protects DA neurons from being damaged by oligomers, which may uncover a common mechanism for synucleinopathies.

    Topics: alpha-Synuclein; Animals; Animals, Genetically Modified; Behavior, Animal; Brain; Disease Models, Animal; Dopamine; Drosophila melanogaster; Drosophila Proteins; Histone Deacetylase 6; Histone Deacetylases; Humans; Inclusion Bodies; Male; Motor Activity; Mutation; Nerve Degeneration; Neurons; Parkinson Disease; Retina

2010
Neurodegeneration in a transgenic mouse model of multiple system atrophy is associated with altered expression of oligodendroglial-derived neurotrophic factors.
    The Journal of neuroscience : the official journal of the Society for Neuroscience, 2010, May-05, Volume: 30, Issue:18

    Multiple system atrophy (MSA) is a neurodegenerative disorder characterized by striatonigral degeneration and olivo-pontocerebellar atrophy. Neuronal degeneration is accompanied by primarily oligodendrocytic accumulation of alpha-synuclein (alphasyn) as opposed to the neuronal inclusions more commonly found in other alpha-synucleinopathies such as Parkinson's disease. It is unclear how alphasyn accumulation in oligodendrocytes may lead to the extensive neurodegeneration observed in MSA; we hypothesize that the altered expression of oligodendrocyte-derived neurotrophic factors by alphasyn may be involved. In this context, the expression of a number neurotrophic factors reportedly expressed by oligodendrocytes [glial-derived neurotrophic factor (GDNF), brain-derived neurotrophic factor (BDNF), and insulin-like growth factor 1 (IGF-1), as well as basic fibroblast growth factor 2 (bFGF2), reportedly astrocyte derived] were examined in transgenic mouse models expressing human alphasyn (halphasyn) under the control of either neuronal (PDGFbeta or mThy1) or oligodendrocytic (MBP) promoters. Although protein levels of BDNF and IGF-1 were altered in all the alphasyn transgenic mice regardless of promoter type, a specific decrease in GDNF protein expression was observed in the MBP-halphasyn transgenic mice. Intracerebroventricular infusion of GDNF improved behavioral deficits and ameliorated neurodegenerative pathology in the MBP-halphasyn transgenic mice. Consistent with the studies in the MBP-halphasyn transgenic mice, analysis of GDNF expression levels in human MSA samples demonstrated a decrease in the white frontal cortex and to a lesser degree in the cerebellum compared with controls. These results suggest a mechanism in which alphasyn expression in oligodendrocytes impacts on the trophic support provided by these cells for neurons, perhaps contributing to neurodegeneration.

    Topics: Aged; alpha-Synuclein; Animals; Behavior, Animal; Brain; Disease Models, Animal; Female; Glial Cell Line-Derived Neurotrophic Factor; Humans; Injections, Intraventricular; Male; Mice; Mice, Knockout; Mice, Transgenic; Multiple System Atrophy; Nerve Degeneration; Nerve Fibers, Myelinated; Nerve Growth Factors; Neurons; Oligodendroglia; Promoter Regions, Genetic

2010
Over-expression of alpha-synuclein in the nervous system enhances axonal degeneration after peripheral nerve lesion in a transgenic mouse strain.
    Journal of neurochemistry, 2010, Volume: 114, Issue:4

    Wallerian degeneration in peripheral nerves occurs after a traumatic insult when the distal nerve part degenerates while peripheral macrophages enter the nerve stump and remove the accruing debris by phagozytosis. We used an experimental model to investigate the effect of either the absence or over-expression of alpha-synuclein (alpha-syn) after transecting the sciatic nerves of mice. alpha-Synuclein is a major component of Lewy bodies and its aggregation results in a premature destruction of nerve cells. It has also been found present in different peripheral nerves but its role in the axon remains still unclear. Following sciatic nerve transection in different mouse strains, we investigated the numbers of invading macrophages, the amounts of remaining myelin and axons 6 days after injury. All mice showed clear signs of Wallerian degeneration, but transgenic mice expressing human wild-type alpha-syn showed lower numbers of invading macrophages, less preserved myelin and significantly lower numbers of preserved axons in comparison with either knockout mice or a mouse strain with a spontaneous deletion of alpha-syn. The use of protein aggregation filtration blots and paraffin-embedded tissue blots displayed depositions of alpha-syn aggregates within sciatic nerve axons of transgenic mice. Thicker myelin sheaths and higher numbers of mitochondria were detected in old alpha-syn transgenic mice. In a human sural nerve, alpha-syn could also be identified within axons. Thus, alpha-syn and its aggregates are not only a component of Lewy bodies and synapses but also of axons and these aggregates might interfere with axonal transport. alpha-Synuclein transgenic mice represent an appropriate model for investigations on axonal transport in neurodegenerative diseases.

    Topics: alpha-Synuclein; Animals; Axonal Transport; Axons; Cell Movement; Disease Models, Animal; Macrophages; Mice; Mice, Inbred C57BL; Mice, Knockout; Mice, Transgenic; Myelin Sheath; Nerve Degeneration; Peripheral Nerves; Sciatic Neuropathy; Wallerian Degeneration

2010
Neuropathological asymmetry in argyrophilic grain disease.
    Journal of neuropathology and experimental neurology, 2010, Volume: 69, Issue:7

    The presence of argyrophilic grains in the neuropil is associated with a form of dementia. We investigated morphological asymmetry in 653 consecutive autopsy patients from a general geriatric hospital (age [mean +/- SD] = 81.1 +/- 8.9 years), focusing on those from patients with advanced argyrophilic grain disease. Paraffin sections of the bilateral posterior hippocampi were immunostained with anti-phosphorylated tau and anti-4-repeat tau antibodies and by the Gallyas-Braak method. In a side-to-side comparison, asymmetry was defined when either the extent or the density of argyrophilic grains was different. Of the 653 subjects, 65 (10%) had Stage 3 argyrophilic grain disease, and 59 (90.8%) showed histopathological asymmetry. Antemortem computed tomographic images (n = 24), magnetic resonance imaging scans (n = 8), and combined computed tomographic and magnetic resonance images (n = 15) were available; images from 20 of the 47 subjects showed asymmetry that correlated with the histopathological asymmetry. Cerebral cortical asymmetry consistent with the histopathology was also visible in N-isopropyl-123I-p-iodoamphetamine single photon emission computed tomographic images from 6 patients and 18F-labeled fluorodeoxyglucose positron emission tomographic images from 2 patients. Thus, asymmetric involvement of the medial temporal lobe in patients with advanced argyrophilic grain disease may represent a diagnostic feature and contribute to distinguishing dementia with grains from Alzheimer disease.

    Topics: Aged; Aged, 80 and over; alpha-Synuclein; Alzheimer Disease; Amyloid beta-Peptides; Apolipoprotein E4; Cognition Disorders; Dementia; Female; Hippocampus; Humans; Magnetic Resonance Imaging; Male; Mental Status Schedule; Middle Aged; Nerve Degeneration; Neurofibrillary Tangles; Neuropil; Retrospective Studies; Statistics, Nonparametric; tau Proteins; Tomography, X-Ray Computed

2010
Neuroscience and heart-brain medicine: the year in review.
    Cleveland Clinic journal of medicine, 2010, Volume: 77 Suppl 3

    Important recent publications in the area of neuroscience and heart-brain medicine center largely around three topics: (1) mechanisms of cardiac sympathetic denervation in Parkinson disease, (2) cytoplasmic monoamine metabolites as autotoxins, and (3) the validity of power spectral analysis of heart rate variability to indicate cardiac sympathetic tone. Findings by Orimo et al support a centripetal, retrograde pathogenetic process involving alpha-synuclein deposition and degeneration of cardiac noradrenergic neurons in Parkinson disease. Several studies suggest that processes increasing cytoplasmic monoamines lead to neuronal loss from auto-oxidation or enzymatic oxidation. Lack of correlation between commonly used indices from power spectral analysis of heart rate variability and cardiac norepinephrine spillover casts doubt on the validity of power spectral analysis to indicate cardiac sympathetic tone.

    Topics: alpha-Synuclein; Brain; Catecholamines; Free Radicals; Heart; Heart Rate; Humans; Lewy Body Disease; Myocardium; Nerve Degeneration; Neurons; Norepinephrine; Parkinson Disease; Sympathectomy; Sympathetic Nervous System

2010
α-Synuclein expression in rat substantia nigra suppresses phospholipase D2 toxicity and nigral neurodegeneration.
    Molecular therapy : the journal of the American Society of Gene Therapy, 2010, Volume: 18, Issue:10

    We present genetic evidence that an in vivo role of α-synuclein (α-syn) is to inhibit phospholipase D2 (PLD2), an enzyme that is believed to participate in vesicle trafficking, membrane signaling, and both endo- and exocytosis. Overexpression of PLD2 in rat substantia nigra pars compacta (SNc) caused severe neurodegeneration of dopamine (DA) neurons, loss of striatal DA, and an associated ipsilateral amphetamine-induced rotational asymmetry. Coexpression of human wild type α-syn suppressed PLD2 neurodegeneration, DA loss, and amphetamine-induced rotational asymmetry. However, an α-syn mutant defective for inhibition of PLD2 in vitro also failed to inhibit PLD toxicity in vivo. Further, reduction of PLD2 activity in SNc, either by siRNA knockdown of PLD2 or overexpression of α-syn, both produced an unusual contralateral amphetamine-induced rotational asymmetry, opposite to that seen with overexpression of PLD2, suggesting that PLD2 and α-syn were both involved in DA release or reuptake. Finally, α-syn coimmunoprecipitated with PLD2 from extracts prepared from striatal tissues. Taken together, our data demonstrate that α-syn is an inhibitor of PLD2 in vivo, and confirm earlier reports that α-syn inhibits PLD2 in vitro. Our data also demonstrate that it is possible to use viral-mediated gene transfer to study gene interactions in vivo.

    Topics: alpha-Synuclein; Animals; Dependovirus; Dopamine; Genetic Vectors; Immunoblotting; Immunohistochemistry; Microscopy, Confocal; Nerve Degeneration; Phospholipase D; Plasmids; Rats; Reverse Transcriptase Polymerase Chain Reaction; Substantia Nigra

2010
Alpha-synuclein promotes SNARE-complex assembly in vivo and in vitro.
    Science (New York, N.Y.), 2010, Sep-24, Volume: 329, Issue:5999

    Presynaptic nerve terminals release neurotransmitters repeatedly, often at high frequency, and in relative isolation from neuronal cell bodies. Repeated release requires cycles of soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE)-complex assembly and disassembly, with continuous generation of reactive SNARE-protein intermediates. Although many forms of neurodegeneration initiate presynaptically, only few pathogenic mechanisms are known, and the functions of presynaptic proteins linked to neurodegeneration, such as α-synuclein, remain unclear. Here, we show that maintenance of continuous presynaptic SNARE-complex assembly required a nonclassical chaperone activity mediated by synucleins. Specifically, α-synuclein directly bound to the SNARE-protein synaptobrevin-2/vesicle-associated membrane protein 2 (VAMP2) and promoted SNARE-complex assembly. Moreover, triple-knockout mice lacking synucleins developed age-dependent neurological impairments, exhibited decreased SNARE-complex assembly, and died prematurely. Thus, synucleins may function to sustain normal SNARE-complex assembly in a presynaptic terminal during aging.

    Topics: Aging; alpha-Synuclein; Animals; Cell Line; Cells, Cultured; HSP40 Heat-Shock Proteins; Humans; Membrane Fusion; Membrane Proteins; Mice; Mice, Knockout; Mice, Transgenic; Nerve Degeneration; Neurons; Presynaptic Terminals; Protein Binding; Rats; Recombinant Fusion Proteins; SNARE Proteins; Vesicle-Associated Membrane Protein 2

2010
Synergistic effects of environmental risk factors and gene mutations in Parkinson's disease accelerate age-related neurodegeneration.
    Journal of neurochemistry, 2010, Volume: 115, Issue:6

    As Parkinson's disease appears to be a multifactoral disorder, the use of animal models to investigate combined effects of genetic and environmental risk factors are of great importance especially in the context of aging which is the single major risk factor for the disorder. Here, we assessed the combined effects of neonatal iron feeding and environmental paraquat exposure on age-related nigrostriatal degeneration in transgenic mice expressing the A53T familial mutant form of human α-synuclein within these neurons. We report here that A53T α-synuclein mice exhibit greater susceptibility to paraquat. Increased oral intake of iron in the neonatal period leads to a progressive age-related enhancement of dopaminergic neurodegeneration associated with paraquat neurotoxicity. Furthermore, neurodegeneration associated with these combined genetic and environmental risk factors could be attenuated by systemic treatment with the bioavailable antioxidant compound EUK-189. These data suggest that environmental factors previously identified as contributors to neurodegeneration associated with sporadic Parkinson's disease may also be candidates for observed variations in symptoms and disease progression in monogenic forms and that this may mechanistically involve increased levels of oxidatively-induced post-translational nitration of α-synuclein.

    Topics: Aging; alpha-Synuclein; Animals; Cell Line; Cells, Cultured; Disease Progression; Environmental Exposure; Humans; Iron; Mice; Mice, Transgenic; Mutation; Nerve Degeneration; Paraquat; Parkinson Disease; Rats; Risk Factors

2010
A stable proportion of Lewy body bearing neurons in the substantia nigra suggests a model in which the Lewy body causes neuronal death.
    Neurobiology of aging, 2010, Volume: 31, Issue:1

    Lewy bodies in Parkinson disease could be innocent bystanders or active agents responsible for neuronal death. Eighteen elderly patients with a Parkinson syndrome were studied prospectively and selected postmortem on the presence of Lewy bodies (14 cases with Parkinson disease, four with dementia with Lewy bodies). Information on disease duration was available in 17 cases. While akinesia and rigidity were linked with the neuronal loss, the percentages of Lewy body bearing neurons and of alpha-synuclein immunoreactive neurons in the substantia nigra were not correlated with the symptoms or the disease duration, and appeared stable, involving 3.6% of the neurons on average. Such stability indicated that, during the whole course of the disease, the destruction of the Lewy bodies was equal to their production. In the model that is proposed here, the Lewy bodies are eliminated when the neurons that bear them die. With the hypothesis that neuronal death is directly related to Lewy bodies, it is possible to estimate their life span, which was calculated to be 6.2 months (15.9 months for any type of alpha-synuclein inclusion).

    Topics: Age of Onset; alpha-Synuclein; Cell Count; Cell Death; Disease Progression; Humans; Lewy Bodies; Lewy Body Disease; Nerve Degeneration; Neurons; Parkinson Disease; Parkinsonian Disorders; Prospective Studies; Substantia Nigra; Time Factors

2010
Common key-signals in learning and neurodegeneration: focus on excito-amino acids, beta-amyloid peptides and alpha-synuclein.
    Journal of neural transmission (Vienna, Austria : 1996), 2009, Volume: 116, Issue:8

    In this paper a hypothesis that some special signals ("key-signals" excito-amino acids, beta-amyloid peptides and alpha-synuclein) are not only involved in information handling by the neuronal circuits, but also trigger out substantial structural and/or functional changes in the Central Nervous System (CNS) is introduced. This forces the neuronal circuits to move from one stable state towards a new state, but in doing so these signals became potentially dangerous. Several mechanisms are put in action to protect neurons and glial cells from these potentially harmful signals. However, in agreement with the Red Queen Theory of Ageing (Agnati et al. in Acta Physiol Scand 145:301-309, 1992), it is proposed that during ageing these neuroprotective processes become less effective while, in the meantime, a shortage of brain plasticity occurs together with an increased need of plasticity for repairing the wear and tear of the CNS. The paper presents findings supporting the concept that such key-signals in instances such as ageing may favour neurodegenerative processes in an attempt of maximizing neuronal plasticity.

    Topics: alpha-Synuclein; Amyloid beta-Peptides; Animals; Brain; Dopamine; Glutamic Acid; Homocysteine; Humans; Learning; Models, Neurological; Nerve Degeneration; Neurodegenerative Diseases; Neuronal Plasticity; Neurons; Protein Conformation; Protein Multimerization; Signal Transduction

2009
Effects of UCH-L1 on alpha-synuclein over-expression mouse model of Parkinson's disease.
    Journal of neurochemistry, 2009, Volume: 108, Issue:4

    The rare inherited form of Parkinson's disease (PD), PARK5, is caused by a missense mutation in ubiquitin carboxy-terminal hydrolase-L1 (UCH-L1) gene, resulting in Ile93Met substitution in its gene product (UCH-L1(Ile93Met)). PARK5 is inherited in an autosomal-dominant mode, but whether the Ile93Met mutation gives rise to a gain-of-toxic-function or loss-of-function of UCH-L1 protein remains controversial. Here, we investigated the selective vulnerabilities of dopaminergic (DA) neurons in UCH-L1-transgenic (Tg) and spontaneous UCH-L1-null gracile axonal dystrophy mice to an important PD-causing insult, abnormal accumulation of alpha-synuclein (alphaSyn). Immunohistochemistry of midbrain sections of a patient with sporadic PD showed alphaSyn- and UCH-L1-double-positive Lewy bodies in nigral DA neurons, suggesting physical and/or functional interaction between the two proteins in human PD brain. Recombinant adeno-associated viral vector-mediated over-expression of alphaSyn for 4 weeks significantly enhanced the loss of nigral DA cell bodies in UCH-L1(Ile93Met)-Tg mice, but had weak effects in age-matched UCH-L1(wild-type)-Tg mice and non-Tg littermates. In contrast, the extent of alphaSyn-induced DA cell loss in gracile axonal dystrophy mice was not significantly different from wild-type littermates at 13-weeks post-injection. Our results support the hypothesis that PARK5 is caused by a gain-of-toxic-function of UCH-L1(Ile93Met) mutant, and suggest that regulation of UCH-L1 in nigral DA cells could be a future target for treatment of PD.

    Topics: Aged; alpha-Synuclein; Animals; Brain Chemistry; Cell Death; Disease Models, Animal; Dopamine; Female; Genetic Predisposition to Disease; Humans; Male; Mice; Mice, Inbred C57BL; Mice, Knockout; Mice, Transgenic; Mutation; Nerve Degeneration; Neurons; Parkinson Disease; Substantia Nigra; Ubiquitin Thiolesterase

2009
Association of alpha-synuclein immunoreactivity with inflammatory activity in multiple sclerosis lesions.
    Journal of neuropathology and experimental neurology, 2009, Volume: 68, Issue:2

    Multiple sclerosis (MS) has neurodegenerative features including neuronal and axonal loss and widespread atrophy of the brain and spinal cord. The cause of this neurodegeneration has been largely attributed to inflammation, but other mechanisms, including those associated with classic neurodegenerative diseases such as the alpha-synucleinopathies, might also be involved in MS pathogenesis. In this study, 96 brain lesions containing varying degrees of inflammatory activity from 12 autopsied MS cases were compared with corresponding regions from 6 neuropathologically normal controls; 2 cerebral biopsy lesions from an MS patient were also studied. We found alpha-synuclein immunoreactivity in the cytoplasm of cells in MS lesions with inflammatory activity but not in control samples. alpha-Synuclein-immunoreactive cells were identified in active (15/15 lesions in the brainstem, 9/13 in cerebral hemispheres) and chronic active (14/15 in the brainstem, 12/22 in cerebral hemispheres) lesions but were absent in chronic inactive lesions (0/31); the greater immunoreactivity in brainstem compared with cerebral hemisphere lesions was significant (p < 0.05). Double-immunofluorescence staining revealed localization of alpha-synuclein immunoreactivity mostly in neurons, microglia/macrophages, and oligodendrocytes, and only rarely in astrocytes. The results suggest that alpha-synuclein expression regulated by inflammatory signals may contribute to neurodegenerative processes in MS lesions.

    Topics: Adult; Aged; alpha-Synuclein; Biomarkers; Brain Stem; Cerebral Infarction; Cerebrum; Encephalitis; Female; Humans; Macrophages; Male; Microglia; Middle Aged; Multiple Sclerosis; Nerve Degeneration; Neuroglia; Neurons; Oligodendroglia; Pyramidal Tracts; Wallerian Degeneration

2009
The ALS/PDC syndrome of Guam and the cycad hypothesis.
    Neurology, 2009, Feb-03, Volume: 72, Issue:5

    Topics: alpha-Synuclein; Amino Acids, Diamino; Amyotrophic Lateral Sclerosis; Animals; Bacterial Toxins; Causality; Cyanobacteria Toxins; Cycas; Dementia; Disease Models, Animal; Epidemiologic Research Design; Epidemiologic Studies; Flour; Guam; Hazardous Substances; Humans; Incidence; Indonesia; Japan; Marine Toxins; Methylazoxymethanol Acetate; Microcystins; Nerve Degeneration; Parkinsonian Disorders; Plant Extracts; Syndrome; Tauopathies

2009
Bacterial artificial chromosome transgenic mice expressing a truncated mutant parkin exhibit age-dependent hypokinetic motor deficits, dopaminergic neuron degeneration, and accumulation of proteinase K-resistant alpha-synuclein.
    The Journal of neuroscience : the official journal of the Society for Neuroscience, 2009, Feb-18, Volume: 29, Issue:7

    Recessive mutations in parkin are the most common cause of familial early-onset Parkinson's disease (PD). Recent studies suggest that certain parkin mutants may exert dominant toxic effects to cultured cells and such dominant toxicity can lead to progressive dopaminergic (DA) neuron degeneration in Drosophila. To explore whether mutant parkin could exert similar pathogenic effects to mammalian DA neurons in vivo, we developed a BAC (bacterial artificial chromosome) transgenic mouse model expressing a C-terminal truncated human mutant parkin (Parkin-Q311X) in DA neurons driven by a dopamine transporter promoter. Parkin-Q311X mice exhibit multiple late-onset and progressive hypokinetic motor deficits. Stereological analyses reveal that the mutant mice develop age-dependent DA neuron degeneration in substantia nigra accompanied by a significant loss of DA neuron terminals in the striatum. Neurochemical analyses reveal a significant reduction of the striatal dopamine level in mutant mice, which is significantly correlated with their hypokinetic motor deficits. Finally, mutant Parkin-Q311X mice, but not wild-type controls, exhibit age-dependent accumulation of proteinase K-resistant endogenous alpha-synuclein in substantia nigra and colocalized with 3-nitrotyrosine, a marker for oxidative protein damage. Hence, our study provides the first mammalian genetic evidence that dominant toxicity of a parkin mutant is sufficient to elicit age-dependent hypokinetic motor deficits and DA neuron loss in vivo, and uncovers a causal relationship between dominant parkin toxicity and progressive alpha-synuclein accumulation in DA neurons. Our study underscores the need to further explore the putative link between parkin dominant toxicity and PD.

    Topics: Aging; alpha-Synuclein; Animals; Chromosomes, Artificial, Bacterial; Corpus Striatum; Disease Models, Animal; Dopamine; Endopeptidase K; Genetic Vectors; Humans; Mice; Mice, Transgenic; Movement Disorders; Mutation; Nerve Degeneration; Parkinson Disease; Protein Structure, Tertiary; Substantia Nigra; Transfection; Ubiquitin-Protein Ligases

2009
Unique copper-induced oligomers mediate alpha-synuclein toxicity.
    FASEB journal : official publication of the Federation of American Societies for Experimental Biology, 2009, Volume: 23, Issue:8

    Parkinson's disease and a number of other neurodegenerative diseases have been linked to either genetic mutations in the alpha-synuclein gene or show evidence of aggregates of the alpha-synuclein protein, sometimes in the form of Lewy bodies. There currently is no clear evidence of a distinct neurotoxic species of alpha-synuclein to explain the death of neurons in these diseases. We undertook to assess the toxicity of alpha-synuclein via exogenous application in cell culture. Initially, we showed that only aggregated alpha-synuclein is neurotoxic and requires the presence copper but not iron. Other members of the synuclein family showed no toxicity in any form and inherited point mutations did not alter the effective toxic concentration of alpha-synuclein. Through protein fractionation techniques, we were able to isolate an oligomeric species responsible for the toxicity of alpha-synuclein. This oligomeric species has a unique stellate appearance under EM and again, requires association with copper to induce cell death. The results allow us to suggest that the toxic species of alpha-synuclein in vivo could possibly be these stellate oligomers and not fibrils. Our data provide a link between the recently noted association of copper and alpha-synuclein and a potential role for the combination in causing neurodegeneration.

    Topics: alpha-Synuclein; Cell Death; Cell Line; Copper; Humans; In Vitro Techniques; Lewy Bodies; Microscopy, Electron, Transmission; Nerve Degeneration; Parkinson Disease; Point Mutation; Protein Structure, Quaternary; Recombinant Proteins

2009
A chemical chaperone, sodium 4-phenylbutyric acid, attenuates the pathogenic potency in human alpha-synuclein A30P + A53T transgenic mice.
    Parkinsonism & related disorders, 2009, Volume: 15, Issue:9

    Aggregation and cytotoxicity of misfolded alpha-synuclein are postulated to be crucial in the disease processes of Parkinson's disease (PD) and other synucleinopathies. Mutations in the alpha-synuclein gene in some pedigrees of familial PD have been reported. The mutant alpha-synuclein has been reported to form fibrillar aggregates resulting in biochemical abnormalities that are responsible for the onset of familial PD. Thus, any agent that effectively prevents the development of misfolded and aggregated alpha-synuclein would be a disease modifying therapeutic candidate. We examined the efficacy of sodium 4-phenylbutyric acid (PBA), one of the chemical chaperons, in transgenic (Tg) mice overexpressing human alpha-synuclein containing a double mutation (A30P + A53T). To evaluate the therapeutic efficacy, bradykinesia and motor coordination were assessed using a pole test and a rotarod treadmill task, respectively. After PBA treatment, these motor deteriorations gradually improved. In immunohistochemical examinations, both a loss of tyrosine hydroxylase-positive neurons and an increase of phosphorylated alpha-synuclein in the substantia nigra were inhibited, resulting in no depletion of the striatal dopamine content. These data suggest that PBA might be one of the therapeutic reagents for neurodegenerative disorders.

    Topics: alpha-Synuclein; Animals; Blotting, Western; Brain; Dopamine; Humans; Immunohistochemistry; Mice; Mice, Transgenic; Molecular Chaperones; Mutation; Nerve Degeneration; Neurons; Neuroprotective Agents; Phenylbutyrates

2009
A highly reproducible rotenone model of Parkinson's disease.
    Neurobiology of disease, 2009, Volume: 34, Issue:2

    The systemic rotenone model of Parkinson's disease (PD) accurately replicates many aspects of the pathology of human PD and has provided insights into the pathogenesis of PD. The major limitation of the rotenone model has been its variability, both in terms of the percentage of animals that develop a clear-cut nigrostriatal lesion and the extent of that lesion. The goal here was to develop an improved and highly reproducible rotenone model of PD. In these studies, male Lewis rats in three age groups (3, 7 or 12-14 months) were administered rotenone (2.75 or 3.0 mg/kg/day) in a specialized vehicle by daily intraperitoneal injection. All rotenone-treated animals developed bradykinesia, postural instability, and/or rigidity, which were reversed by apomorphine, consistent with a lesion of the nigrostriatal dopamine system. Animals were sacrificed when the PD phenotype became debilitating. Rotenone treatment caused a 45% loss of tyrosine hydroxylase-positive substantia nigra neurons and a commensurate loss of striatal dopamine. Additionally, in rotenone-treated animals, alpha-synuclein and poly-ubiquitin positive aggregates were observed in dopamine neurons of the substantia nigra. In summary, this version of the rotenone model is highly reproducible and may provide an excellent tool to test new neuroprotective strategies.

    Topics: alpha-Synuclein; Animals; Disease Models, Animal; Dopamine; Dyskinesia, Drug-Induced; Hypokinesia; Injections, Intraperitoneal; Male; Muscle Rigidity; Nerve Degeneration; Neurons; Neurotoxins; Parkinsonian Disorders; Rats; Rats, Inbred Lew; Reproducibility of Results; Rotenone; Substantia Nigra; Tyrosine 3-Monooxygenase; Ubiquitins; Uncoupling Agents

2009
Unified staging system for Lewy body disorders: correlation with nigrostriatal degeneration, cognitive impairment and motor dysfunction.
    Acta neuropathologica, 2009, Volume: 117, Issue:6

    The two current major staging systems in use for Lewy body disorders fail to classify up to 50% of subjects. Both systems do not allow for large numbers of subjects who have Lewy-type alpha-synucleinopathy (LTS) confined to the olfactory bulb or who pass through a limbic-predominant pathway that at least initially bypasses the brainstem. The results of the current study, based on examination of a standard set of ten brain regions from 417 subjects stained immunohistochemically for alpha-synuclein, suggest a new staging system that, in this study, allows for the classification of all subjects with Lewy body disorders. The autopsied subjects included elderly subjects with Parkinson's disease, dementia with Lewy bodies, incidental Lewy body disease and Alzheimer's disease with Lewy bodies, as well as comparison groups without Lewy bodies. All subjects were classifiable into one of the following stages: I. Olfactory Bulb Only; IIa Brainstem Predominant; IIb Limbic Predominant; III Brainstem and Limbic; IV Neocortical. Progression of subjects through these stages was accompanied by a generally stepwise worsening in terms of striatal tyrosine hydroxylase concentration, substantia nigra pigmented neuron loss score, Mini Mental State Examination score and score on the Unified Parkinson's Disease Rating Scale Part 3. Additionally, there were significant correlations between these measures and LTS density scores. It is suggested that the proposed staging system would improve on its predecessors by allowing classification of a much greater proportion of cases.

    Topics: Aged; Aged, 80 and over; Aging; alpha-Synuclein; Alzheimer Disease; Apolipoproteins E; Brain; Cognition Disorders; Diagnosis, Differential; Female; Humans; Immunohistochemistry; Lewy Body Disease; Male; Movement Disorders; Nerve Degeneration; Neurodegenerative Diseases; Parkinson Disease; Severity of Illness Index

2009
Absence of alpha-synuclein pathology in postencephalitic parkinsonism.
    Acta neuropathologica, 2009, Volume: 118, Issue:3

    Postencephalitic parkinsonism (PEP), a chronic complication of encephalitis lethargica, is a tauopathy characterized by multisystem neuronal loss and gliosis with widespread neurofibrillary lesions composed of both 3- and 4-repeat (3R and 4R) tau isoforms. Previous immunohistochemical studies in a small number of PEP cases demonstrated absence of Lewy bodies as well as the lack of other alpha-synuclein pathology, classifying PEP as a "pure" tauopathy. Neuropathologic examination of 10 brains with clinico-pathologically verified PEP confirmed widespread neurodegeneration in subcortical and brainstem areas associated with multifocal neurofibrillary pathology comprising both 3R and 4R tau. Very rare beta-amyloid deposits were observed in two elderly patients, while Lewy bodies and neurites or any other alpha-synuclein deposits were completely absent. The causes and molecular background of total absence of alpha-synuclein pathology in PEP, in contrast to most other tauopathies, remain as unknown as the pathogenesis of PEP.

    Topics: Adult; alpha-Synuclein; Amyloid beta-Peptides; Brain; Brain Stem; Female; Humans; Immunohistochemistry; Male; Middle Aged; Nerve Degeneration; Neurons; Parkinson Disease, Postencephalitic; Tauopathies

2009
Mitochondrial inhibitor 3-nitroproprionic acid enhances oxidative modification of alpha-synuclein in a transgenic mouse model of multiple system atrophy.
    Journal of neuroscience research, 2009, Volume: 87, Issue:12

    Multiple system atrophy (MSA) is a progressive neurodegenerative disease characterized by autonomic failure, parkinsonism, cerebellar ataxia, and oligodendrocytic accumulation of alpha-synuclein (alphasyn). Oxidative stress has been linked to neuronal death in MSA and the mitochondrial toxin 3-nitropropionic acid (3NP) is known to enhance the motor deficits and neurodegeneration in transgenic mice models of MSA. However, the effect of 3NP administration on alphasyn itself has not been studied. In this context, we examined the neuropathological effects of 3NP administration in alphasyn transgenic mice expressing human alphasyn (halphasyn) under the control of the myelin basic protein (MBP) promoter and the effect of this administration on posttranslational modifications of alphasyn, on levels of total alphasyn, and on its solubility. We demonstrate that 3NP administration altered levels of nitrated and oxidized alphasyn in the MBP-halphasyn tg while not affecting global levels of phosphorylated or total alphasyn. 3NP administration also exaggerated neurological deficits in the MBP-halphasyn tg mice, resulting in widespread neuronal degeneration and behavioral impairment.

    Topics: alpha-Synuclein; Animals; Brain; Convulsants; Disease Models, Animal; Mice; Mice, Transgenic; Mitochondria; Multiple System Atrophy; Myelin Basic Protein; Nerve Degeneration; Nitrates; Nitro Compounds; Oxidative Stress; Promoter Regions, Genetic; Propionates

2009
Striatal neuroinflammation promotes Parkinsonism in rats.
    PloS one, 2009, Volume: 4, Issue:5

    Sporadic Parkinson's disease (PD) is a progressive neurodegenerative disorder with unknown cause, but it has been suggested that neuroinflammation may play a role in pathogenesis of the disease. Neuroinflammatory component in process of PD neurodegeneration was proposed by postmortem, epidemiological and animal model studies. However, it remains unclear how neuroinflammatory factors contribute to dopaminergic neuronal death in PD.. In this study, we analyzed the relationship among inducible nitric oxide synthase (iNOS)-derived NO, mitochondrial dysfunction and dopaminergic neurodegeneration to examine the possibility that microglial neuroinflammation may induce dopaminergic neuronal loss in the substantia nigra. Unilateral injection of lipopolysaccharide (LPS) into the striatum of rat was followed by immunocytochemical, histological, neurochemical and biochemical analyses. In addition, behavioral assessments including cylinder test and amphetamine-induced rotational behavior test were employed to validate ipsilateral damage to the dopamine nigrostriatal pathway. LPS injection caused progressive degeneration of the dopamine nigrostriatal system, which was accompanied by motor impairments including asymmetric usage of forelimbs and amphetamine-induced turning behavior in animals. Interestingly, some of the remaining nigral dopaminergic neurons had intracytoplasmic accumulation of alpha-synuclein and ubiquitin. Furthermore, defect in the mitochondrial respiratory chain, and extensive S-nitrosylation/nitration of mitochondrial complex I were detected prior to the dopaminergic neuronal loss. The mitochondrial injury was prevented by treatment with L-N(6)-(l-iminoethyl)-lysine, an iNOS inhibitor, suggesting that iNOS-derived NO is associated with the mitochondrial impairment.. These results implicate neuroinflammation-induced S-nitrosylation/nitration of mitochondrial complex I in mitochondrial malfunction and subsequent degeneration of the nigral dopamine neurons.

    Topics: alpha-Synuclein; Animals; Behavior, Animal; Cytoplasm; Disease Progression; Dopamine; Drug Administration Routes; Electron Transport Complex I; Inflammation; Lipopolysaccharides; Male; Mitochondria; Neostriatum; Nerve Degeneration; Neurons; Nitrosation; Parkinsonian Disorders; Rats; Rats, Sprague-Dawley; Ubiquitin

2009
The role of ubiquitin linkages on alpha-synuclein induced-toxicity in a Drosophila model of Parkinson's disease.
    Journal of neurochemistry, 2009, Volume: 110, Issue:1

    Parkinson's disease (PD) is a common movement disorder marked by the loss of dopaminergic (DA) neurons in the brain stem and the presence of intraneuronal inclusions designated as Lewy bodies (LB). The cause of neurodegeneration in PD is not clear, but it has been suggested that protein misfolding and aggregation contribute significantly to the development of the disease. Misfolded and aggregated proteins are cleared by ubiquitin proteasomal system (UPS) and autophagy lysosomal pathway (ALP). Recent studies suggested that different types of ubiquitin linkages can modulate these two pathways in the process of protein degradation. In this study, we found that co-expression of ubiquitin can rescue neurons from alpha-syn-induced neurotoxicity in a Drosophila model of PD. This neuroprotection is dependent on the formation of lysine 48 polyubiquitin linkage which is known to target protein degradation via the proteasome. Consistent with our results that we observed in vivo, we found that ubiquitin co-expression in the cell can facilitate cellular protein degradation by the proteasome in a lysine 48 polyubiquitin-dependent manner. Taken together, these results suggest that facilitation of proteasomal protein degradation can be a potential therapeutic approach for PD.

    Topics: alpha-Synuclein; Animals; Autophagy; Cell Line, Tumor; Cells, Cultured; Cytoprotection; Disease Models, Animal; Drosophila melanogaster; Humans; Lewy Bodies; Lysine; Lysosomes; Nerve Degeneration; Neurons; Parkinson Disease; Polymers; Proteasome Endopeptidase Complex; Protein Folding; Substantia Nigra; Ubiquitin; Ubiquitination

2009
Differences between subacute and chronic MPTP mice models: investigation of dopaminergic neuronal degeneration and alpha-synuclein inclusions.
    Journal of neurochemistry, 2009, Volume: 109, Issue:5

    Animal models are invaluable tools to study neurodegenerative disorders but a general consensus on the most accurate rodent model of Parkinson's disease has not been reached. Here, we examined how different methods of MPTP administration influence the degeneration of the dopaminergic (DA) system. Adult male C57BL/6 mice were treated with the same cumulative dose of MPTP following four distinct procedures: (i) subacute i.p. injections; (ii) 28-day chronic s.c. infusion; (iii) 28-day chronic i.p. infusion; and (iv) 14-day chronic i.p. infusion. Subacute MPTP treatment significantly affected all aspects of the DA system within the nigral and striatal territories. In contrast, the 28-day chronic s.c. infusion did not significantly alter any components of the DA system. The 28- and 14-day chronic i.p. infusions induced loss of tyrosine hydroxylase (TH)-positive cells correlated with a decrease in Nurr1 mRNA levels, but no significant decrease in the density of TH striatal fibers. Importantly, however, only the 14-day chronic MPTP i.p. infusion protocol promoted the formation of neuronal inclusions as noted by the expression of alpha-synuclein protein within the cytoplasm of TH nigral neurons. Overall, we found that the 14-day chronic MPTP i.p. infusion reproduces more accurately the pathological characteristics of early stage Parkinson's disease.

    Topics: 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine; alpha-Synuclein; Animals; Corpus Striatum; Disease Models, Animal; DNA-Binding Proteins; Dopamine; Drug Administration Routes; Drug Administration Schedule; Exploratory Behavior; Intranuclear Inclusion Bodies; Mice; Mice, Inbred C57BL; MPTP Poisoning; Nerve Degeneration; Nuclear Receptor Subfamily 4, Group A, Member 2; Substantia Nigra; Time Factors; Transcription Factors; Tyrosine 3-Monooxygenase

2009
Subchronic infusion of the product of inflammation prostaglandin J2 models sporadic Parkinson's disease in mice.
    Journal of neuroinflammation, 2009, Jul-25, Volume: 6

    Chronic neuroinflammation is implicated in Parkinson's disease (PD). Inflammation involves the activation of microglia and astrocytes that release high levels of prostaglandins. There is a profound gap in our understanding of how cyclooxygenases and their prostaglandin products redirect cellular events to promote PD neurodegeneration. The major prostaglandin in the mammalian brain is prostaglandin D2, which readily undergoes spontaneous dehydration to generate the bioactive cyclopentenone prostaglandins of the J2 series. These J2 prostaglandins are highly reactive and neurotoxic products of inflammation shown in cellular models to impair the ubiquitin/proteasome pathway and cause the accumulation of ubiquitinated proteins. PD is a disorder that exhibits accumulation of ubiquitinated proteins in neuronal inclusions (Lewy bodies). The role of J2 prostaglandins in promoting PD neurodegeneration has not been investigated under in vivo conditions.. We addressed the neurodegenerative and behavioral effects of the administration of prostaglandin J2 (PGJ2) simultaneously into the substantia nigra/striatum of adult male FVB mice by subchronic microinjections. One group received unilateral injections of DMSO (vehicle, n = 6) and three groups received PGJ2 [3.4 microg or 6.7 microg (n = 6 per group) or 16.7 microg (n = 5)] per injection. Immunohistochemical and behavioral analyses were applied to assess the effects of the subchronic PGJ2 microinfusions.. Immunohistochemical analysis demonstrated a PGJ2 dose-dependent significant and selective loss of dopaminergic neurons in the substantia nigra while the GABAergic neurons were spared. PGJ2 also triggered formation of aggregates immunoreactive for ubiquitin and alpha-synuclein in the spared dopaminergic neurons. Moreover, PGJ2 infusion caused a massive microglia and astrocyte activation that could initiate a deleterious cascade leading to self-sustained progressive neurodegeneration. The PGJ2-treated mice also exhibited locomotor and posture impairment.. Our studies establish the first model of inflammation in which administration of an endogenous highly reactive product of inflammation, PGJ2, recapitulates key aspects of PD. Our novel PGJ2-induced PD model strongly supports the view that localized and chronic production of highly reactive and neurotoxic prostaglandins, such as PGJ2, in the CNS could be an integral component of inflammation triggered by insults evoked by physical, chemical or microbial stimuli and thus establishes a link between neuroinflammation and PD neurodegeneration.

    Topics: alpha-Synuclein; Animals; Cell Death; Disease Models, Animal; Dopamine; Dose-Response Relationship, Drug; Drug Administration Schedule; Encephalitis; Gliosis; Immunohistochemistry; Inclusion Bodies; Inflammation Mediators; Male; Mice; Microinjections; Movement Disorders; Nerve Degeneration; Neurons; Parkinsonian Disorders; Prostaglandin D2; Substantia Nigra

2009
Neuroprotective effect of the antiparkinsonian drug pramipexole against nigrostriatal dopaminergic degeneration in rotenone-treated mice.
    Neurochemistry international, 2009, Volume: 55, Issue:8

    Pramipexole, an agonist for dopamine (DA) D2/D3-receptors, has been used to treat both early and advanced Parkinson's disease (PD). In this study, we examined the effect of pramipexole on DA neurons in a PD model of C57BL/6 mice, which were treated with rotenone (30 mg/kg, p.o.) daily for 28 days. Pramipexole (1 mg/kg, i.p.) was injected daily 30 min before each oral administration of rotenone. Chronic oral administration of rotenone caused a loss of DA neurons in the substantia nigra pars compacta (SNpc), motor deficits and the up-regulation of alpha-synuclein immunoreactivity in some surviving DA neurons. Pramipexole inhibited rotenone-induced DA neuronal death and motor deficits, and reduced immunoreactivity for alpha-synuclein. In addition, pramipexole inhibited the in vitro oligomerization of human wild-type alpha-synuclein by H(2)O(2)plus cytochrome c. To examine the neuroprotective effect of pramipexole against oxidative stress, we used a DJ-1-knockdown SH-SY5Y cell line and electron spin resonance (ESR) spectrometry. Simultaneous treatment with H(2)O(2) and pramipexole resulted in the significant protection of DJ-1-knockdown cells against cell death in a concentration-dependent manner. A high concentration of pramipexole directly scavenged hydroxyl radical (*OH) generated from H(2)O(2) and Fe(2+). Furthermore, pramipexole increased Bcl-2 immunoreactivity in DA neurons in the SNpc. These results suggest that pramipexole may protect DA neurons against exposure to rotenone by chronic oral administration, and this effect is mediated by multiple functions including scavenging of *OH and induction of Bcl-2 protein.

    Topics: alpha-Synuclein; Animals; Antiparkinson Agents; Apoptosis; Benzothiazoles; Cell Line, Tumor; Corpus Striatum; Cytochromes c; Dopamine; Dose-Response Relationship, Drug; Humans; Hydrogen Peroxide; Hydroxyl Radical; Male; Mice; Mice, Inbred C57BL; Nerve Degeneration; Neural Pathways; Neurotoxins; Oxidative Stress; Parkinsonian Disorders; Pramipexole; Proto-Oncogene Proteins c-bcl-2; Rotenone; Substantia Nigra; Uncoupling Agents

2009
Parkinsonism in hiv-infected patients on highly active antiretroviral therapy.
    Neurology, 2009, Aug-04, Volume: 73, Issue:5

    Topics: Adult; Age of Onset; Aging; AIDS Dementia Complex; alpha-Synuclein; Antiretroviral Therapy, Highly Active; Causality; Disease Progression; Humans; Male; Middle Aged; Mitochondrial Diseases; Nerve Degeneration; Neurons; Parkinsonian Disorders; RNA, Viral; Substantia Nigra; Ubiquitination; Viral Load

2009
Methylation status and neurodegenerative markers in Parkinson disease.
    Clinical chemistry, 2009, Volume: 55, Issue:10

    Increased concentrations of plasma total homocysteine (tHcy) have been associated with age-related diseases, including dementia, stroke, and Parkinson disease (PD). Methylation status might link Hcy metabolism to neurodegenerative proteins in patients with PD.. We tested blood samples from 87 patients with PD (median age 68 years; 35 men) for tHcy, methylmalonic acid (MMA), vitamin B(12), vitamin B(6), folate, S-adenosyl methionine (SAM), S-adenosyl homocysteine (SAH), and amyloid-beta(1-42). We collected citrate blood from a subset of 45 patients to prepare platelet-rich plasma, and we used washed platelets to prepare cell extracts for amyloid precursor protein (APP) and alpha-synuclein assays. We used brain parenchyma sonography to estimate the substantia nigra echogenic area in a subset of 59 patients.. Serum concentrations of tHcy were increased in PD patients (median 14.8 micromol/L). tHcy (beta coefficient = -0.276) and serum creatinine (beta = -0.422) were significant predictors of the ratio of SAM/SAH in plasma (P < 0.01). The plasma SAM/SAH ratio was a significant determinant for DemTect scores (beta = 0.612, P = 0.004). Significant negative correlations were found between concentrations of SAH in plasma and platelet APP and between SAM and platelet alpha-synuclein. A larger echogenic area of the substantia nigra was related to higher serum concentrations of MMA (P = 0.016).. Markers of neurodegeneration (APP, alpha-synuclein) are related to markers of methylation (SAM, SAH) in patients with PD. Better cognitive function was related to higher methylation potential (SAM/SAH ratio).

    Topics: Aged; alpha-Synuclein; Amyloid beta-Peptides; Amyloid beta-Protein Precursor; Biomarkers; Blood Platelets; Cognition Disorders; Female; Folic Acid; Homocysteine; Humans; Male; Methylation; Middle Aged; Nerve Degeneration; Parkinson Disease; Peptide Fragments; S-Adenosylhomocysteine; S-Adenosylmethionine; Substantia Nigra; Ultrasonography; Vitamin B 6

2009
Relative preservation of thalamic centromedian nucleus in parkinsonian patients with dystonia.
    Movement disorders : official journal of the Movement Disorder Society, 2009, Oct-30, Volume: 24, Issue:14

    To determine whether variable thalamic degeneration in Parkinson's disease (PD) contributes to less drug responsive clinical features. Formalin-fixed thalami from longitudinally followed patients with PD and early dystonia (N = 6), early falls (N = 5) or no dystonia or falls (N = 6) and age-matched controls without neuropathology (N = 10) were serially sectioned, stained, and analyzed. Neurons in the centromedian parafascicular (CM-Pf) nucleus were quantified using the optical disector method and analysis of variance with post hoc testing used to determine variability in neurodegeneration between groups. Patients with PD were confirmed to have significant neurodegeneration in the CM-Pf complex, with no difference in the degree of neurodegeneration between patients with PD with early falls compared with patients with no history of falls or dystonia. In contrast, patients with PD with early dystonia had significantly less neurodegeneration of the CM but not the Pf than patients without this feature. Preservation of the CM in patients with PD with early dystonia would result in a relative increase in CM activity through the direct basal ganglia pathway and increased primary motor cortex activity. Overall this data provides evidence for pathway-specific neurodegeneration as an underlying feature of the clinical variability observed in patients with PD.

    Topics: Aged; alpha-Synuclein; Antiparkinson Agents; Atrophy; Cell Count; Disease Progression; Dystonia; Female; Humans; Imaging, Three-Dimensional; Intralaminar Thalamic Nuclei; Lewy Bodies; Male; Middle Aged; Nerve Degeneration; Parkinson Disease

2009
Beclin 1 gene transfer activates autophagy and ameliorates the neurodegenerative pathology in alpha-synuclein models of Parkinson's and Lewy body diseases.
    The Journal of neuroscience : the official journal of the Society for Neuroscience, 2009, Oct-28, Volume: 29, Issue:43

    Accumulation of the synaptic protein alpha-synuclein (alpha-syn) is a hallmark of Parkinson's disease (PD) and Lewy body disease (LBD), a heterogeneous group of disorders with dementia and parkinsonism, where Alzheimer's disease and PD interact. Accumulation of alpha-syn in these patients might be associated with alterations in the autophagy pathway. Therefore, we postulate that delivery of beclin 1, a regulator of the autophagy pathway, might constitute a strategy toward developing a therapy for LBD/PD. Overexpression of alpha-syn from lentivirus transduction in a neuronal cell line resulted in lysosomal accumulation and alterations in autophagy. Coexpression of beclin 1 activated autophagy, reduced accumulation of alpha-syn, and ameliorated associated neuritic alterations. The effects of beclin 1 overexpression on LC3 and alpha-syn accumulation were partially blocked by 3-MA and completely blocked by bafilomycin A1. In contrast, rapamycin enhanced the effects of beclin 1. To evaluate the potential effects of activating autophagy in vivo, a lentivirus expressing beclin 1 was delivered to the brain of a alpha-syn transgenic mouse. Neuropathological analysis demonstrated that beclin 1 injections ameliorated the synaptic and dendritic pathology in the tg mice and reduced the accumulation of alpha-syn in the limbic system without any significant deleterious effects. This was accompanied by enhanced lysosomal activation and reduced alterations in the autophagy pathway. Thus, beclin 1 plays an important role in the intracellular degradation of alpha-syn either directly or indirectly through the autophagy pathway and may present a novel therapeutic target for LBD/PD.

    Topics: alpha-Synuclein; Animals; Apoptosis Regulatory Proteins; Autophagy; Beclin-1; Brain; Cell Line, Tumor; Disease Models, Animal; Gene Transfer Techniques; Humans; Lewy Body Disease; Mice; Mice, Transgenic; Nerve Degeneration; Neurons; Parkinson Disease; Rats; Signal Transduction

2009
Chaperone proteostasis in Parkinson's disease: stabilization of the Hsp70/alpha-synuclein complex by Hip.
    The EMBO journal, 2009, Dec-02, Volume: 28, Issue:23

    The ATP-dependent protein chaperone heat-shock protein 70 (Hsp70) displays broad anti-aggregation functions and has a critical function in preventing protein misfolding pathologies. According to in vitro and in vivo models of Parkinson's disease (PD), loss of Hsp70 activity is associated with neurodegeneration and the formation of amyloid deposits of alpha-synuclein (alphaSyn), which constitute the intraneuronal inclusions in PD patients known as Lewy bodies. Here, we show that Hsp70 depletion can be a direct result of the presence of aggregation-prone polypeptides. We show a nucleotide-dependent interaction between Hsp70 and alphaSyn, which leads to the aggregation of Hsp70, in the presence of ADP along with alphaSyn. Such a co-aggregation phenomenon can be prevented in vitro by the co-chaperone Hip (ST13), and the hypothesis that it might do so also in vivo is supported by studies of a Caenorhabditis elegans model of alphaSyn aggregation. Our findings indicate that a decreased expression of Hip could facilitate depletion of Hsp70 by amyloidogenic polypeptides, impairing chaperone proteostasis and stimulating neurodegeneration.

    Topics: Adenosine Triphosphate; alpha-Synuclein; Amyloid; Animals; Animals, Genetically Modified; Caenorhabditis elegans; Carrier Proteins; Cell Line, Tumor; Homeostasis; HSP70 Heat-Shock Proteins; Humans; Molecular Chaperones; Multiprotein Complexes; Nerve Degeneration; Parkinson Disease; Peptides; Protein Folding; Protein Stability; Rats; Tumor Suppressor Proteins

2009
Degeneration in different parkinsonian syndromes relates to astrocyte type and astrocyte protein expression.
    Journal of neuropathology and experimental neurology, 2009, Volume: 68, Issue:10

    The reactive changes in different types of astrocytes were analyzed in parkinsonian syndromes in order to identify common reactions and their relationship to disease severity. Immunohistochemistry was used on formalin-fixed, paraffin-embedded sections from the putamen, pons, and substantia nigra from 13 Parkinson disease (PD), 29 multiple-system atrophy (MSA), 34 progressive supranuclear palsy (PSP), 10 corticobasal degeneration(CBD), and 13 control cases. Classic reactive astrocytes were observed in MSA, PSP, and CBD, but not PD cases; the extent of reactivity correlated with indices of neurodegeneration and disease stage. Approximately 40% to 45% of subcortical astrocytes in PD and PSP accumulated alpha-synuclein and phospho-tau, respectively; subcortical astrocytes in MSA and CBD cases did not accumulate these proteins. Protoplasmic astrocytes were identified from fibrous astrocytes by their expression of parkin coregulated gene and apolipoprotein D, and accumulated abnormal proteins in PD, PSP, and CBD, but not MSA. The increased reactivity of parkin coregulated gene-immunoreactive protoplasmic astrocytes correlated with parkin expression in PSP and CBD. Nonreactive protoplasmic astrocytes were observed in PD and MSA cases; in PD, they accumulated alpha-synuclein, suggesting that the attenuated response might be due to an increase in the level of alpha-synuclein. These heterogeneous astroglial responses in PD, MSA, PSP, and CBD indicate distinct underlying pathogenic mechanisms in each disorder.

    Topics: alpha-Synuclein; Apolipoproteins D; Astrocytes; Glycoproteins; Humans; Immunohistochemistry; Membrane Transport Proteins; Multiple System Atrophy; Nerve Degeneration; Parkinson Disease; Parkinsonian Disorders; Phosphorylation; Pons; Putamen; Severity of Illness Index; Substantia Nigra; Supranuclear Palsy, Progressive; tau Proteins

2009
alpha-synuclein and LRRK2: partners in crime.
    Neuron, 2009, Dec-24, Volume: 64, Issue:6

    In this issue of Neuron, Lin et al. report that LRRK2 modulates age-related neurodegeneration caused by overexpression of alpha-synuclein in the forebrain of transgenic mice. Overexpression of LRRK2 accelerates the progression of alpha-synuclein-mediated neuropathological changes, whereas deletion of LRRK2 alleviates these alterations. The results reveal an interesting interaction between alpha-synuclein and LRRK2, two gene products linked to dominantly inherited Parkinson's disease.

    Topics: alpha-Synuclein; Animals; Brain; Disease Models, Animal; Endoplasmic Reticulum; Gene Expression Regulation; Genetic Predisposition to Disease; Golgi Apparatus; Humans; Leucine-Rich Repeat Serine-Threonine Protein Kinase-2; Mice; Mice, Transgenic; Microtubules; Nerve Degeneration; Parkinson Disease; Protein Serine-Threonine Kinases; Protein Transport

2009
Leucine-rich repeat kinase 2 regulates the progression of neuropathology induced by Parkinson's-disease-related mutant alpha-synuclein.
    Neuron, 2009, Dec-24, Volume: 64, Issue:6

    Mutations in alpha-synuclein and Leucine-rich repeat kinase 2 (LRRK2) are linked to autosomal dominant forms of Parkinson's disease (PD). However, little is known about any potential pathophysiological interplay between these two PD-related genes. Here we show in transgenic mice that although overexpression of LRRK2 alone did not cause neurodegeneration, the presence of excess LRRK2 greatly accelerated the progression of neuropathological abnormalities developed in PD-related A53T alpha-synuclein transgenic mice. Moreover, we found that LRRK2 promoted the abnormal aggregation and somatic accumulation of alpha-synuclein in A53T mice, which likely resulted from the impairment of microtubule dynamics, Golgi organization, and the ubiquitin-proteasome pathway. Conversely, genetic ablation of LRRK2 preserved the Golgi structure and suppressed the aggregation and somatic accumulation of alpha-synuclein, and thereby delayed the progression of neuropathology in A53T mice. These findings demonstrate that overexpression of LRRK2 enhances alpha-synuclein-mediated cytotoxicity and suggest inhibition of LRRK2 expression as a potential therapeutic option for ameliorating alpha-synuclein-induced neurodegeneration.

    Topics: alpha-Synuclein; Animals; Brain; Disease Progression; Gene Expression Regulation; Genetic Predisposition to Disease; Golgi Apparatus; Inclusion Bodies; Leucine-Rich Repeat Serine-Threonine Protein Kinase-2; Mice; Mice, Knockout; Mice, Transgenic; Microtubules; Mutation; Nerve Degeneration; Neurons; Parkinson Disease; Protein Serine-Threonine Kinases

2009
Neurodegeneration in Parkinson's disease: genetics enlightens physiopathology.
    Journal of neural transmission. Supplementum, 2009, Issue:73

    Parkinson's disease (PD) is a severe neurodegenerative disorder of complex etiology and enigmatic physiopathology. In the past decade, the identification of genes involved in rare familial Parkinsonian syndromes has brought hope that understanding the functions of their products will provide insight into the molecular mechanisms responsible for neurodegeneration. The knowledge accumulated thus far has delineated two putative, potentially interconnected, disease-causing pathways: alpha-synuclein accumulation may be central to Parkinsonism due to alpha-synuclein gene defects, but possibly also to sporadic PD and other genetic forms presenting with Lewy bodies; altered mitochondrial physiology may be pivotal to Parkinsonian syndromes caused by parkin, PINK1, and possibly DJ-1 gene mutations. Adding new pieces to this fragmentary picture to determine to what extent sporadic PD and Parkinsonism due to distinct genetic causes share common pathogenic mechanisms remains a major challenge toward the development of future therapeutic strategies for these disabling disorders.

    Topics: alpha-Synuclein; Dopamine; Humans; Mitochondrial Diseases; Mutation; Nerve Degeneration; Neurons; Oxidative Stress; Parkinson Disease; Protein Kinases; Ubiquitin-Protein Ligases

2009
Fas-associated factor 1 and Parkinson's disease.
    Neurobiology of disease, 2008, Volume: 31, Issue:3

    Fas-associated factor 1 or FAF1 is a Fas-binding protein implicated in apoptosis. FAF1 is the product of a gene at PARK 10 locus on chromosome 1p32, a locus associated with late-onset PD [Hicks, A.A., Petursson, H., Jonsson, T., Stefansson, H., Johannsdottir, H.S., Sainz, J., Frigge, M.L.et al., 2002. A susceptibility gene for late-onset idiopathic Parkinson's disease. Ann Neurol. 52, 549-555.]. In the present study we investigated the role of FAF1 in cell death and in Parkinson's disease (PD) pathogenesis. FAF1 levels were significantly increased in frontal cortex of PD as well as in PD cases with Alzheimer's disease (AD) pathology compared to control cases. Changes in FAF1 expression were specific to PD-related alpha-synuclein pathology and nigral cell loss. In addition, PD-related insults including, mitochondrial complex I inhibition, oxidative stress, and increased alpha-synuclein expression specifically increased endogenous FAF1 expression in vitro. Increased FAF1 levels induced cell death and significantly potentiated toxic effects of PD-related stressors including, oxidative stress, mitochondrial complex I inhibition and proteasomal inhibition. These studies, together with previous genetic linkage studies, highlight the potential significance of FAF1 in pathogenesis of idiopathic PD.

    Topics: Adaptor Proteins, Signal Transducing; Aged; Aged, 80 and over; alpha-Synuclein; Apoptosis Regulatory Proteins; Brain; Cell Death; Cell Line; Chromosomes, Human, Pair 1; Electron Transport Complex I; Energy Metabolism; Frontal Lobe; Gene Expression Regulation; Genetic Predisposition to Disease; Humans; Middle Aged; Mitochondria; Nerve Degeneration; Neurons; Oxidative Stress; Parkinson Disease; Substantia Nigra

2008
Direct quantification of CSF alpha-synuclein by ELISA and first cross-sectional study in patients with neurodegeneration.
    Experimental neurology, 2008, Volume: 213, Issue:2

    Because accumulation of alpha-synuclein (alphaS) in the brain is a hallmark of Parkinson disease (PD) and related disorders, we examined its occurrence in human cerebrospinal fluid (CSF). Following affinity enrichment and trypsin digestion of CSF collected from a neurologically healthy donor, we identified several alphaS-derived peptides by mass spectrometry. The concentration of alphaS amounted to <0.001% of the CSF proteome. We then built, validated and optimized a sandwich-type, enzyme-linked immunoadsorbent assay (ELISA) to measure total alphaS levels in unconcentrated CSF. In a cross-sectional study of 100 living donors, we examined cell-free CSF samples from subjects clinically diagnosed with advanced PD, dementia with Lewy bodies (DLB), Alzheimer disease (AD), and a group of non-neurodegenerative disease controls (NCO). In these four groups the CSF alphaS concentrations ranged from 0.8 to 16.2 pg/microl. Mean CSF alphaS values were lower in donors with a primary synucleinopathy (PD, DLB: n=57) than in the other two groups (AD, NCO: n=35; p=0.025). By contrast, living Creutzfeldt-Jakob disease patients showed markedly elevated CSF alphaS levels (n=8; mean, 300 pg/microl; p<0.001). Our results unequivocally confirm the presence of alphaS in adult human CSF. In a first feasibility study employing a novel ELISA, we found relatively low CSF alphaS concentrations in subjects with parkinsonism linked to synucleinopathy, PD and DLB. In definite prion disease cases, we recorded a marked rise in total CSF alphaS resulting from rapid cell death. Our results will likely aid future biomarker explorations in neurodegenerative conditions and facilitate target validation studies.

    Topics: Adult; Aged; Aged, 80 and over; alpha-Synuclein; Amino Acid Sequence; Animals; Biomarkers; Cells, Cultured; Cross-Sectional Studies; Enzyme-Linked Immunosorbent Assay; Female; Humans; Male; Mice; Middle Aged; Molecular Sequence Data; Nerve Degeneration; Rats

2008
Neurofibrillary tau pathology modulated by genetic variation of alpha-synuclein.
    Annals of neurology, 2008, Volume: 64, Issue:3

    We analyzed whether genetic variation of alpha-synuclein modulates the extent of neuropathological changes in a population-based autopsied sample of 272 elderly Finns. None of the 11 markers was associated with the extent of neocortical beta-amyloid pathology. The intron 4 marker rs2572324 was associated with the extent of neurofibrillary pathology (p = 0.0006, permuted p = 0.004; Braak stages IV-VI vs 0-II). The same variant also showed a trend for association with neocortical Lewy-related pathology. These results suggest for the first time that variation of alpha-synuclein modulates neurofibrillary tau pathology and support the recent observations of an interaction of alpha-synuclein and tau in neurodegeneration.

    Topics: Aged, 80 and over; alpha-Synuclein; Brain; Cohort Studies; DNA Mutational Analysis; Female; Genetic Markers; Genetic Predisposition to Disease; Genetic Testing; Genetic Variation; Humans; Lewy Bodies; Male; Neocortex; Nerve Degeneration; Neurodegenerative Diseases; Neurofibrillary Tangles; Neurons; tau Proteins

2008
Rifampicin reduces alpha-synuclein in a transgenic mouse model of multiple system atrophy.
    Neuroreport, 2008, Aug-27, Volume: 19, Issue:13

    Multiple system atrophy (MSA) is a progressive neurodegenerative disorder characterized by oligodendrocytic cytoplasmic inclusions containing abnormally aggregated alpha-synuclein. This aggregation has been linked to the neurodegeneration observed in MSA. Current MSA treatments are aimed at controlling symptoms rather than tackling the underlying cause of neurodegeneration. This study investigates the ability of the antibiotic rifampicin to reduce alpha-synuclein aggregation and the associated neurodegeneration in a transgenic mouse model of MSA. We report a reduction in monomeric and oligomeric alpha-synuclein and a reduction in phosphorylated alpha-synuclein (S129) upon rifampicin treatment. This reduction in alpha-synuclein aggregation was accompanied by reduced neurodegeneration. On the basis of its anti-aggregenic properties, we conclude that rifampicin may have therapeutic potential for MSA.

    Topics: alpha-Synuclein; Animals; Antibiotics, Antitubercular; beta-Synuclein; Blotting, Western; Disease Models, Animal; Humans; Immunohistochemistry; Inclusion Bodies; Injections, Intraperitoneal; Mice; Mice, Transgenic; Microscopy, Confocal; Multiple System Atrophy; Nerve Degeneration; Oligodendroglia; Rifampin; Synucleins

2008
LRRK2 and parkin immunoreactivity in multiple system atrophy inclusions.
    Acta neuropathologica, 2008, Volume: 116, Issue:6

    Certain genetic defects in LRRK2 and parkin are pathogenic for Parkinson's disease (PD) and both proteins deposit in the characteristic Lewy bodies. LRRK2 is thought to be involved in the early initiation of Lewy bodies. The involvement of LRRK2 and parkin in the similar cellular deposition of fibrillar alpha-synuclein in glial cytoplasmic inclusions (GCI) in multiple system atrophy (MSA) has not yet been assessed. To determine whether LRRK2 and parkin may be similarly associated with the abnormal deposition of alpha-synuclein in MSA GCI, paraffin-embedded sections from the basal ganglia of 12 patients with MSA, 4 with PD and 4 controls were immunostained for LRRK2, parkin, alpha-synuclein and oligodendroglial proteins using triple labelling procedures. The severity of neuronal loss was graded and the proportion of abnormally enlarged oligodendroglia containing different combinations of proteins assessed in 80-100 cells per case. Parkin immunoreactivity was observed in only a small proportion of GCI. In contrast, LRRK2 was found in most of the enlarged oligodendroglia in MSA and colocalised with the majority of alpha-synuclein-immunopositive GCI. Degrading myelin sheaths containing LRRK2-immunoreactivity were also observed, showing an association with one of the earliest oligodendroglial abnormalities observed in MSA. The proportion of LRRK2-immunopositive GCI was negatively associated with an increase in neuronal loss and alpha-synuclein-immunopositive dystrophic axons. Our results indicate that an increase in LRRK2 expression occurs early in association with myelin degradation and GCI formation, and that a reduction in LRRK2 expression in oligodendroglia is associated with increased neuronal loss in MSA.

    Topics: Aged; alpha-Synuclein; Axons; Basal Ganglia; Female; Fluorescent Antibody Technique; Humans; Immunohistochemistry; Inclusion Bodies; Internal Capsule; Leucine-Rich Repeat Serine-Threonine Protein Kinase-2; Male; Middle Aged; Multiple System Atrophy; Myelin Sheath; Nerve Degeneration; Parkinson Disease; Protein Serine-Threonine Kinases; Putamen; Ubiquitin; Ubiquitin-Protein Ligases

2008
Targeted overexpression of human alpha-synuclein triggers microglial activation and an adaptive immune response in a mouse model of Parkinson disease.
    Journal of neuropathology and experimental neurology, 2008, Volume: 67, Issue:12

    Microglial activation and adaptive immunity have been implicated in the neurodegenerative processes in Parkinson disease. It has been proposed that these responses may be triggered by modified forms of alpha-synuclein (alpha-SYN), particularly nitrated species, which are released as a consequence of dopaminergic neurodegeneration. To examine the relationship between alpha-SYN, microglial activation, and adaptive immunity, we used a mouse model of Parkinson disease in which human alpha-SYN is overexpressed by a recombinant adeno-associated virus vector, serotype 2 (AAV2-SYN); this overexpression leads to slow degeneration of dopaminergic neurons. Microglial activation and components of the adaptive immune response were assessed using immunohistochemistry; quantitative polymerase chain reaction was used to examine cytokine expression. Four weeks after injection, there was a marked increase in CD68-positive microglia and greater infiltration of B and T lymphocytes in the substantia nigra pars compacta of the AAV2-SYN group than in controls. At 12 weeks, CD68 staining declined, but B- and T-cell infiltration persisted. Expression of proinflammatory cytokines was enhanced, whereas markers of alternative activation (i.e. arginase I and interleukins 4 and 13) were not altered. Increased immunoreactivity for mouse immunoglobulin was detected at all time points in the AAV2-SYN animals. These data show that overexpression of alpha-SYN alone, in the absence of overt neurodegeneration, is sufficient to trigger neuroinflammation with both microglial activation and stimulation of adaptive immunity.

    Topics: alpha-Synuclein; Animals; Antigens, CD; Antigens, Differentiation, Myelomonocytic; Brain; Chemotaxis, Leukocyte; Cytokines; Disease Models, Animal; Dopamine; Genetic Vectors; Humans; Immune System Phenomena; Immunoglobulins; Lymphocyte Activation; Male; Mice; Mice, Inbred C57BL; Microglia; Nerve Degeneration; Parkinson Disease; Substantia Nigra; Transfection

2008
Pathological lesions in colonic biopsies during Parkinson's disease.
    Gut, 2008, Volume: 57, Issue:12

    Topics: Aged; alpha-Synuclein; Biopsy; Case-Control Studies; Colon; Constipation; Enteric Nervous System; Humans; Immunohistochemistry; Male; Nerve Degeneration; Parkinson Disease; Pilot Projects; Submucous Plexus

2008
Application of a C. elegans dopamine neuron degeneration assay for the validation of potential Parkinson's disease genes.
    Journal of visualized experiments : JoVE, 2008, Jul-18, Issue:17

    Improvements to the diagnosis and treatment of Parkinson's disease (PD) are dependent upon knowledge about susceptibility factors that render populations at risk. In the process of attempting to identify novel genetic factors associated with PD, scientists have generated many lists of candidate genes, polymorphisms, and proteins that represent important advances, but these leads remain mechanistically undefined. Our work is aimed toward significantly narrowing such lists by exploiting the advantages of a simple animal model system. While humans have billions of neurons, the microscopic roundworm Caenorhabditis elegans has precisely 302, of which only eight produce dopamine (DA) in hemaphrodites. Expression of a human gene encoding the PD-associated protein, alpha-synuclein, in C. elegans DA neurons results in dosage and age-dependent neurodegeneration. Worms expressing human alpha-synuclein in DA neurons are isogenic and express both GFP and human alpha-synuclein under the DA transporter promoter (Pdat-1). The presence of GFP serves as a readily visualized marker for following DA neurodegeneration in these animals. We initially demonstrated that alpha-synuclein-induced DA neurodegeneration could be rescued in these animals by torsinA, a protein with molecular chaperone activity. Further, candidate PD-related genes identified in our lab via large-scale RNAi screening efforts using an alpha-synuclein misfolding assay were then over-expressed in C. elegans DA neurons. We determined that five of seven genes tested represented significant candidate modulators of PD as they rescued alpha-synuclein-induced DA neurodegeneration. Additionally, the Lindquist Lab (this issue of JoVE) has performed yeast screens whereby alpha-synuclein-dependent toxicity is used as a readout for genes that can enhance or suppress cytotoxicity. We subsequently examined the yeast candidate genes in our C. elegans alpha-synuclein-induced neurodegeneration assay and successfully validated many of these targets. Our methodology involves generation of a C. elegans DA neuron-specific expression vector using recombinational cloning of candidate gene cDNAs under control of the Pdat-1 promoter. These plasmids are then microinjected in wild-type (N2) worms, along with a selectable marker for successful transformation. Multiple stable transgenic lines producing the candidate protein in DA neurons are obtained and then independently crossed into the alpha-synuclein degenerative strain and assesse

    Topics: alpha-Synuclein; Animals; Animals, Genetically Modified; Caenorhabditis elegans; Disease Models, Animal; Dopamine; Genetic Vectors; Humans; Nerve Degeneration; Neurons; Parkinson Disease

2008
Axonal alpha-synuclein aggregates herald centripetal degeneration of cardiac sympathetic nerve in Parkinson's disease.
    Brain : a journal of neurology, 2008, Volume: 131, Issue:Pt 3

    Degeneration of the cardiac sympathetic nerve occurs in both Parkinson's disease (PD) and dementia with Lewy bodies and begins early in the disease progression of PD, accounting for reduced cardiac uptake of meta-iodobenzylguanidine even in the early stages of Lewy body disease (LBD). We previously demonstrated that degeneration of the distal axons of the cardiac sympathetic nerve precedes loss of their mother neurons in the paravertebral sympathetic ganglia, suggesting distal dominant degeneration of the cardiac sympathetic nerve in PD. Because alpha-synuclein is one of the key molecules in the pathogenesis of this disease, we further investigated how alpha-synuclein aggregates are involved in this distal-dominant degeneration. Both cardiac tissues and paravertebral sympathetic ganglia were obtained for comparison from 20 patients with incidental Lewy body disease (ILBD), 10 with PD, 20 with multiple system atrophy (MSA) and 10 control subjects. Immunohistochemical analysis was performed using antibodies against tyrosine hydroxylase (TH) as a marker for sympathetic nerves, phosphorylated neurofilament as a marker for axons and phosphorylated alpha-synuclein for pathological deposits. We found that (i) alpha-synuclein aggregates in the epicardial nerve fascicles, namely the distal axons of the cardiac sympathetic nerve, were much more abundant in ILBD with preserved TH-ir axons than in this disease with decreased TH-ir axons and PD; (ii) alpha-synuclein aggregates in the epicardial nerve fascicles were closely related to the disappearance of TH-ir axons; (iii) in ILBD with preserved TH-ir axons, alpha-synuclein aggregates were consistently more abundant in the epicardial nerve fascicles than in the paravertebral sympathetic ganglia; (iv) this distal-dominant accumulation of alpha-synuclein aggregates was reversed in ILBD with decreased TH-ir axons and PD, which both showed fewer of these axons but more abundant alpha-synuclein aggregates in the paravertebral sympathetic ganglia and (v) MSA was completely different from ILBD and PD based on the preservation of TH-ir axons and the scarcity of alpha-synuclein aggregates in either the cardiac tissues or the paravertebral sympathetic ganglia. These findings indicate that accumulation of alpha-synuclein aggregates in the distal axons of the cardiac sympathetic nervous system precedes that of neuronal somata or neurites in the paravertebral sympathetic ganglia and that heralds centripetal degeneration of the car

    Topics: Aged; Aged, 80 and over; alpha-Synuclein; Axons; Female; Ganglia, Sympathetic; Heart; Humans; Lewy Body Disease; Male; Middle Aged; Multiple System Atrophy; Nerve Degeneration; Parkinson Disease; Pericardium; Sympathetic Nervous System; Tyrosine 3-Monooxygenase

2008
Is alpha-synuclein the culprit of the Parkinsonian neurodegeneration?
    Experimental neurology, 2008, Volume: 209, Issue:1

    Topics: alpha-Synuclein; Animals; Disease Models, Animal; Humans; Nerve Degeneration; Parkinson Disease

2008
Induction of the phase II detoxification pathway suppresses neuron loss in Drosophila models of Parkinson's disease.
    The Journal of neuroscience : the official journal of the Society for Neuroscience, 2008, Jan-09, Volume: 28, Issue:2

    Alpha-synuclein aggregates are a common feature of sporadic Parkinson's disease (PD), and mutations that increase alpha-synuclein abundance confer rare heritable forms of PD. Although these findings suggest that alpha-synuclein plays a central role in the pathogenesis of this disorder, little is known of the mechanism by which alpha-synuclein promotes neuron loss or the factors that regulate alpha-synuclein toxicity. To address these matters, we tested candidate modifiers of alpha-synuclein toxicity using a Drosophila model of PD. In the current work, we focused on phase II detoxification enzymes involved in glutathione metabolism. We find that the neuronal death accompanying alpha-synuclein expression in Drosophila is enhanced by loss-of-function mutations in genes that promote glutathione synthesis and glutathione conjugation. This neuronal loss can be overcome by genetic or pharmacological interventions that increase glutathione synthesis or glutathione conjugation activity. Moreover, these same pharmacological agents suppress neuron loss in Drosophila parkin mutants, a loss-of-function model of PD. Our results suggest that oxidative stress is a feature of alpha-synuclein toxicity and that induction of the phase II detoxification pathway represents a potential preventative therapy for PD.

    Topics: Age Factors; Allyl Compounds; alpha-Synuclein; Animals; Animals, Genetically Modified; Cell Death; Disease Models, Animal; Disulfides; Dose-Response Relationship, Drug; Drosophila; Drosophila Proteins; Glutathione; Isothiocyanates; Metabolic Detoxication, Phase II; Metabolic Networks and Pathways; Mutation; Nerve Degeneration; Nerve Tissue Proteins; Neurons; Parkinson Disease; Sulfoxides; Thiocyanates; Tyrosine 3-Monooxygenase

2008
A molecular pathway of neurodegeneration linking alpha-synuclein to ApoE and Abeta peptides.
    Nature neuroscience, 2008, Volume: 11, Issue:3

    Pathogenic aggregates of alpha-synuclein are thought to contribute to the development of Parkinson's disease. Inclusion bodies containing alpha-synuclein are present in Parkinson's disease and other neurodegenerative diseases, including Alzheimer's disease. Moreover, alpha-synuclein mutations are found in cases of familial Parkinson's disease, and transgenic overexpression of alpha-synuclein causes neurodegeneration in mice. The molecular mechanisms involved, however, remain incompletely understood. Here we show that, in transgenic mice, alpha-synuclein induced neurodegeneration involves activation of the ubiquitin/proteasome system, a massive increase in apolipoprotein E (ApoE) levels and accumulation of insoluble mouse Abeta. ApoE was not protective, but was injurious, as deletion of ApoE delayed the neurodegeneration caused by alpha-synuclein and suppressed the accumulation of Abeta. Our data reveal a molecular link between central pathogenic mechanisms implicated in Parkinson's disease and Alzheimer's disease and suggest that intracellular alpha-synuclein is pathogenic, at least in part, by activation of extracellular signaling pathways involving ApoE.

    Topics: alpha-Synuclein; Alzheimer Disease; Amyloid beta-Peptides; Animals; Apolipoproteins E; Brain; Cell Death; Humans; Lewy Body Disease; Mice; Mice, Knockout; Mice, Transgenic; Nerve Degeneration; Neurodegenerative Diseases; Parkinson Disease; Peptides; Proteasome Endopeptidase Complex; Signal Transduction; Ubiquitin

2008
Alpha-synuclein aggregation alters tyrosine hydroxylase phosphorylation and immunoreactivity: lessons from viral transduction of knockout mice.
    Neuroscience letters, 2008, Apr-11, Volume: 435, Issue:1

    Tyrosine hydroxylase (TH), the rate limiting enzyme in catecholamine synthesis, is frequently used as a marker of dopaminergic neuronal loss in animal models of Parkinson's disease (PD). We have been exploring the normal function of the PD-related protein alpha-synuclein (alpha-Syn) with regard to dopamine synthesis. TH is activated by the phosphorylation of key seryl residues in the TH regulatory domain. Using in vitro models, our laboratory discovered that alpha-Syn inhibits TH by acting to reduce TH phosphorylation, which then reduces dopamine synthesis [X.-M. Peng, R. Tehranian, P. Dietrich, L. Stefanis, R.G. Perez, Alpha-synuclein activation of protein phosphatase 2A reduces tyrosine hydroxylase phosphorylation in dopaminergic cells, J. Cell. Sci. 118 (2005) 3523-3530; R.G. Perez, J.C. Waymire, E. Lin, J.J. Liu, F. Guo, M.J. Zigmond, A role for alpha-synuclein in the regulation of dopamine biosynthesis, J. Neurosci. 22 (2002) 3090-3099]. We recently began exploring the impact of alpha-Syn on TH in vivo, by transducing dopaminergic neurons in alpha-Syn knockout mouse (ASKO) olfactory bulb using wild type human alpha-Syn lentivirus. At 3.5-21 days after viral delivery, alpha-Syn expression was transduced primarily in periglomerular dopaminergic neurons. Cells with modest levels of alpha-Syn consistently co-labeled for Total-TH. However, cells bearing aggregated alpha-Syn, as revealed by proteinase K or Thioflavin-S treatment had significantly reduced Total-TH immunoreactivity, but high phosphoserine-TH labeling. On immunoblots, we noted that Total-TH immunoreactivity was equivalent in all conditions, although tissues with alpha-Syn aggregates again had higher phosphoserine-TH levels. This suggests that aggregated alpha-Syn is no longer able to inhibit TH. Although the reason(s) underlying reduced Total-TH immunoreactivity on tissue sections await(s) confirmation, the dopaminergic phenotype was easily verified using phosphorylation-state-specific TH antibodies. These findings have implications not only for normal alpha-Syn function in TH regulation, but also for measuring cell loss that is associated with synucleinopathy.

    Topics: alpha-Synuclein; Animals; Cells, Cultured; Disease Models, Animal; Dopamine; Down-Regulation; Genetic Vectors; Humans; Immunohistochemistry; Inclusion Bodies; Lentivirus; Male; Mice; Mice, Knockout; Nerve Degeneration; Neurons; Olfactory Bulb; Parkinson Disease; Phosphorylation; Substantia Nigra; Transduction, Genetic; Transfection; Tyrosine 3-Monooxygenase

2008
FK506 binding protein 12 differentially accelerates fibril formation of wild type alpha-synuclein and its clinical mutants A30P or A53T.
    Journal of neurochemistry, 2008, Volume: 106, Issue:1

    Aggregation of alpha-synuclein (alpha-SYN) plays a key role in Parkinson's disease. We have previously shown that aggregation of alpha-SYN in vitro is accelerated by addition of FK506 binding proteins (FKBP) and that this effect can be counteracted by FK506, a specific inhibitor of these enzymes. In this paper, we investigated in detail the effect of FKBP12 on early aggregation and on fibril formation of wild-type, A53T and A30P alpha-SYN. FKBP12 has a much smaller effect on the fibril formation of these two clinical mutants alpha-SYN. Using an inactive enzyme, we were able to discriminate between catalytic and non-catalytic effects that differentially influence the two processes. A model explaining non-linear concentration dependencies is proposed.

    Topics: alpha-Synuclein; Amino Acid Substitution; Brain; Catalytic Domain; Cell Line, Tumor; Humans; Immunosuppressive Agents; Mutation; Nerve Degeneration; Neurofibrillary Tangles; Neurons; Nonlinear Dynamics; Parkinson Disease; Tacrolimus; Tacrolimus Binding Protein 1A; Time Factors

2008
RNA interference mediated silencing of alpha-synuclein in MN9D cells and its effects on cell viability.
    Neuroscience bulletin, 2008, Volume: 24, Issue:2

    To silence the expression of alpha-synuclein in MN9D dopaminergic cells using vector mediated RNA interference (RNAi) and examined its effects on cell proliferation and viability.. We identified two 19-nucleotide stretches within the coding region of the alpha-synuclein gene and designed three sets of oligonucleotides to generate double-stranded (ds) oligos. The ds oligos were inserted into the pENTR/H1/TO vector and transfected into MN9D dopaminergic cells. alpha-Synuclein expression was detected by RT-PCR, real-time PCR, immunocytochemistry staining and Western blot. In addition, we measured cell proliferation using growth curves and cell viability by 3-(4, 5)-dimethylthiahiazo (-z-y1)-3, 5-di- phenytetrazoliumromide (MTT).. The mRNA and protein levels of alpha-synuclein gene were significantly down-regulated in pSH2/alpha-SYN-transfected cells compared with control MN9D and pSH/CON-transfected MN9D cells, while pSH1/alpha-SYN-transfected cells showed no significant difference. Silencing alpha-synuclein expression does not affect cell proliferation but may decrease cell viability.. Our results demonstrated pSH2/alpha-SYN is an effective small interfering RNA (siRNA) sequence and potent silencing of mouse alpha-synuclein expression in MN9D cells by vector-based RNAi, which provides the tools for studying the normal function of alpha-synuclein and examining its role in Parkinson's disease (PD) pathogenesis. alpha-Synuclein may be important for the viability of MN9D cells, and loss of alpha-synuclein may induce cell injury directly or indirectly.

    Topics: alpha-Synuclein; Animals; Cell Line; Cell Proliferation; Cell Survival; Down-Regulation; Gene Silencing; Genetic Vectors; Hybridomas; Mice; Mice, Inbred C57BL; Nerve Degeneration; Neurons; Oligonucleotides; Parkinson Disease; Plasmids; RNA Interference; RNA, Double-Stranded; RNA, Small Interfering; Transfection

2008
Proteasomal abnormalities in cortical Lewy body disease and the impact of proteasomal inhibition within cortical and cholinergic systems.
    Journal of neural transmission (Vienna, Austria : 1996), 2008, Volume: 115, Issue:6

    Dementia with Lewy bodies (DLB) accounts for 15-20% of the millions of people worldwide with dementia. In the current work we investigate the association between proteasome dysfunction and the development of cortical Lewy body pathology. Analysis of post-mortem cortical tissue indicated levels of the alpha-subunit of the 20S proteasome were significantly reduced in DLB cortex, but not Alzheimer's, in comparison to control and this reduction correlated with both the severity and duration of dementia. Application of proteasome inhibitors to rodent cortical primary neurones in vitro and by direct injection onto rodent cholinergic forebrain neurons in vivo gave rise to dose dependent neuronal death and in rodent cortex -- marked cholinergic deficits accompanied by the accumulation of inclusions that stained positive for alpha-synuclein and ubiquitin. These findings suggest that proteasomal abnormalities are present within cortical Lewy body disease and the experimental inhibition of proteasomal function mirrors the neuropathological changes seen within the disorder.

    Topics: Acetylcholine; Acetylcysteine; Aged; Aged, 80 and over; alpha-Synuclein; Animals; Basal Nucleus of Meynert; Cell Death; Cells, Cultured; Cerebral Cortex; Cholinergic Fibers; Disease Models, Animal; Dose-Response Relationship, Drug; Enzyme Inhibitors; Female; Humans; Lewy Bodies; Lewy Body Disease; Male; Nerve Degeneration; Nerve Tissue Proteins; Neural Pathways; Oligopeptides; Proteasome Endopeptidase Complex; Proteasome Inhibitors; Rats; Rats, Sprague-Dawley; Ubiquitin

2008
Favorable effects of a prolonged treatment with melatonin on the level of oxidative damage and neurodegeneration in senescence-accelerated mice.
    Journal of pineal research, 2008, Volume: 45, Issue:3

    Senescence-accelerated mice (SAMP8) and senescence-accelerated resistant mice (SAMR1) were studied at 5 and 10 months of age, respectively. In the animals, neurodegenerative processes and how they were influenced by melatonin were examined. Melatonin (10 mg/kg) or vehicle (ethanol at 0.066%) treatments were administrated from the age of 1 to 9 months in the drinking water. Differences in the neurodegenerative markers examined were found between the two strains with a more damaged protein, phosphorylated Tau at Ser392, increased neurofibrillary tangles (NT) and higher alpha-synuclein expression in SAMP8 versus SAMR1 mice overall, when the mice were 10 months of age. Changes in density of receptors and oxidative stress-related signaling with age were found in the brains of SAM strains at 10 months as shown by a marked decrease in the level of MT-1 melatonin receptor and retinoic acid receptor-related orphan receptor (ROR)-alpha1. This diminution was earlier and more pronounced in SAMP8 mice. Likewise, the levels of nuclear factor-kappa B (NF-kB) transcriptional factor were higher in SAMP8 mice compared with SAMR1 mice regardless of age confirming the direct role of oxidative stress in the aging process. Treatment with melatonin in SAMP8 and SAMR1 mice reduced the neurodegenerative changes with an increase of ROR-alpha1 levels without an apparent influence in the levels of MT-1 receptor. However, different melatonin effects on NF-kB signaling were observed suggesting that NF-kB could trigger inflammatory processes in a different way, being SAM strain-dependent and associated with age-related oxidative stress levels. The effectiveness of melatonin in improving age-related neural impairments is corroborated.

    Topics: Aging; Aging, Premature; alpha-Synuclein; Animals; Antioxidants; Brain; Lewy Bodies; Melatonin; Mice; Nerve Degeneration; Neurofibrillary Tangles; NF-kappa B p50 Subunit; Nuclear Receptor Subfamily 1, Group F, Member 1; Oxidative Stress; Protein Carbonylation; Receptor, Melatonin, MT1; Receptors, Cytoplasmic and Nuclear; Signal Transduction; tau Proteins; Trans-Activators

2008
Alpha-synuclein aggregation and cell death triggered by energy deprivation and dopamine overload are counteracted by D2/D3 receptor activation.
    Journal of neurochemistry, 2008, Volume: 106, Issue:2

    Progressive degeneration and intraneuronal Lewy bodies made of filamentous alpha-synuclein (alpha-syn) in dopaminergic cells of the nigrostriatal system are characteristics of Parkinson's disease (PD). Glucose uptake is reduced in some of the brain regions affected by PD neurodegenerative changes. Defects in mitochondrial activity in the substantia nigra have been observed in the brain of patients affected by PD and substantia nigra lesions can induce the onset of a secondary parkinsonism. Thus, energy starvation and consequently metabolic impairment to dopaminergic neurons may be related to the onset of PD. On this line, we evaluated the effect of nutrient starvation, reproduced 'in vitro' by glucose deprivation (GD), in primary mesecephalic neuronal cultures and dopaminergic-differentiated SH-SY5Y cells, to evaluate if decreased glucose support to dopaminergic cells can lead to mitochondrial damage, neurodegeneration and alpha-syn misfolding. Furthermore, we investigated the effect of dopamine (DA) treatment in the presence of a DA-uptake inhibitor or of the D(2)/D(3) receptor (D(2)R/D(3)R) agonist quinpirole on GD-treated cells, to evaluate the efficacy of these therapeutic compounds. We found that GD induced the formation of fibrillary aggregated alpha-syn inclusions containing the DA transporter in dopaminergic cells. These alterations were accompanied by dopaminergic cell death and were exacerbated by DA overload. Conversely, the block of DA uptake and D(2)R/D(3)R agonist treatment exerted neuroprotective effects. These data indicate that glucose starvation is likely involved in the induction of PD-related pathological changes in dopaminergic neurons. These changes may be counteracted by the block of DA uptake and by dopaminergic agonist treatment.

    Topics: alpha-Synuclein; Analysis of Variance; Animals; Benzothiazoles; Cell Death; Cells, Cultured; Dopamine; Dopamine Agents; Dopamine Plasma Membrane Transport Proteins; Embryo, Mammalian; Formazans; Gene Expression Regulation; Glucose; Mesencephalon; Mice; Mitochondria; Nerve Degeneration; Nerve Tissue Proteins; Neurons; Protein Transport; Receptors, Dopamine D2; Tetrazolium Salts; Thiazoles

2008
Olfactory pathogenesis of idiopathic Parkinson disease revisited.
    Movement disorders : official journal of the Movement Disorder Society, 2008, Jun-15, Volume: 23, Issue:8

    Idiopathic Parkinson disease (PD) is traditionally considered a movement disorder with hallmark lesions located in the substantia nigra pars compacta (SNpc). However, recent histopathological studies of some PD cases suggest the possibility of a multisystem disorder which progresses in a predictable sequence as described in Braak's staging criteria. The disease process starts in the dorsal motor nucleus of the vagus (dmX) and anterior olfactory nucleus and bulb, and from there, spreads through the brainstem nuclei to ultimately reach the SNpc, which then presents as symptomatic PD. In this article, we would like to revisit the olfactory pathogenesis of PD based on Braak's staging system and review anatomical pathways supporting such a possibility. We also suggest some biomarkers for early stages of PD. Additionally, we present and discuss the possibility that a prion-like process underlies the neurodegenerative changes in PD.

    Topics: alpha-Synuclein; Alzheimer Disease; Amyloid beta-Peptides; Brain Stem; Cerebral Cortex; Humans; Lewy Bodies; Limbic System; Nerve Degeneration; Neural Pathways; Neurons; Olfactory Bulb; Olfactory Nerve; Parkinson Disease; Prion Diseases; Substantia Nigra; Vagus Nerve; Vagus Nerve Diseases

2008
Cardiac sympathetic denervation correlates with clinical and pathologic stages of Parkinson's disease.
    Movement disorders : official journal of the Movement Disorder Society, 2008, Jun-15, Volume: 23, Issue:8

    Attention has been drawn to cardiac sympathetic denervation in Parkinson's disease (PD) based on clinical studies using [123I] metaiodobenzylguanidine scintigraphy; however, the histologic correlates and time course of cardiac sympathetic denervation are poorly understood. To address these issues, we used tyrosine hydroxylase (TH) immunohistochemistry to detect cardiac sympathetic nerve fibers in the epicardium of 4 normal controls, 11 cases with incidental Lewy bodies (iLBs), and 14 cases of PD. Cardiac sympathetic innervation was significantly less in PD than in normal controls and cases with iLBs (P < 0.05). There was also a decrease in TH-immunoreactive fibers in iLB cases compared to normal controls (P < 0.01). TH-immunoreactive fibers correlated with the PD stage (r = -0.75, P < 0.001), as well as with Hoehn & Yahr clinical stage (r = -0.61, P < 0.001), and disease duration (r = -0.63, P < 0.001). Immunohistochemistry for alpha-synuclein showed neurites in epicardium in PD and iLB cases, but not in normal controls. The density of alpha-synuclein neurites correlated with Braak PD stage (r = 0.38, P < 0.05), Hoehn & Yahr clinical stage (r = 0.44, P < 0.05), and disease duration (r = 0.42, P < 0.05). This study demonstrates that cardiac sympathetic degeneration and alpha-synuclein pathology is present in presymptomatic phase of PD, and that both increase with disease duration and severity.

    Topics: Aged; Aged, 80 and over; alpha-Synuclein; Brain; Disease Progression; Female; Heart; Humans; Hypotension, Orthostatic; Lewy Bodies; Male; Nerve Degeneration; Nerve Fibers; Neurites; Parkinson Disease; Pericardium; Spinal Cord; Sympathetic Nervous System; Tyrosine 3-Monooxygenase

2008
Non-autonomous cell death in Parkinson's disease.
    The Lancet. Neurology, 2008, Volume: 7, Issue:6

    Topics: alpha-Synuclein; Animals; Brain Tissue Transplantation; Cell Death; Encephalitis; Fetal Tissue Transplantation; Gliosis; Graft Survival; Humans; Lewy Bodies; Mice; Microglia; Nerve Degeneration; Neurons; Parkinson Disease; Substantia Nigra

2008
The somatosensory cortex in multiple system atrophy.
    Journal of the neurological sciences, 2008, Aug-15, Volume: 271, Issue:1-2

    In multiple system atrophy (MSA), it has been accepted that the motor-related cortical area may degenerate. However, there have been few investigations of the postcentral cortex of the somatosensory area. For this reason, we investigated the effects of MSA on both the precentral and the postcentral cortex and were able to demonstrate degenerative changes in each. Furthermore, our study showed that degeneration of the postcentral cortex preceded that of the precentral cortex. In addition, we showed that the Betz cells were not selectively lost, but merely depleted like other neurons of the deep cortical layers. Therefore, the effects of MSA are apparently related to selective loss of the small-sized myelinated fibers in the corticospinal tract.

    Topics: alpha-Synuclein; Antigens, CD; Antigens, Differentiation, Myelomonocytic; Female; Glial Fibrillary Acidic Protein; Humans; Male; Middle Aged; Multiple System Atrophy; Nerve Degeneration; Somatosensory Cortex; Statistics, Nonparametric

2008
Tau and alpha-synuclein brainstem pathology in Alzheimer disease: relation with extrapyramidal signs.
    Acta neuropathologica, 2007, Volume: 113, Issue:1

    Extrapyramidal symptoms (EPS) in Alzheimer disease (AD) often increase with disease severity. Their neuropathological substrate is a matter of discussion. We investigated tau and alpha-synuclein (AS) pathologies in brainstem in AD patients with and without EPS. Among 160 elderly subjects with autopsy-proven AD (110 female, 50 male, aged 61-102, mean 84.1 +/- 8.3 SD years), 151 (94.4%) being demented, 35 (21.9%) had clinically reported EPS (rigidity, bradykinesia, gait impairment). Neuropathological examination included standardized classification of AD according to current criteria, and semiquantitative assessment of neuronal loss in substantia nigra (SN), locus coeruleus (LC), and of tau and AS lesions in brainstem, and, in addition, of cerebrovascular lesions. The prevalence of EPS was only slightly more frequent in higher Braak stages. Tau pathology in brainstem significantly increased with increasing Braak stages, while AS lesions did not. EPS correlated best with SN cell loss (P < 0.001) and much less with AS pathology in several brain areas (P < 0.05), except in medulla oblongata (P < 0.001). Although both pathologies in substantia nigra correlated with neuron loss (P < 0.001), nigral tau lesions, present in 88.5% of EPS positive cases (without AS lesions in 55.6%), did not correlate with EPS. Additional cerebrovascular changes apparently did not influence the development of EPS symptoms in fully developed AD. With other recent data, these results suggest that neuronal loss in SN, partly related to tau lesions, is a major pathological substrate of EPS in AD, but some cases with and without EPS may show no or only minimal nigral changes. However, often associated with nigral tau lesions and higher Braak stages, EPS in elderly patients may be a surrogate marker for severe neuritic AD pathology.

    Topics: Aged; Aged, 80 and over; alpha-Synuclein; Alzheimer Disease; Basal Ganglia Diseases; Brain Stem; Female; Humans; Male; Middle Aged; Nerve Degeneration; Psychiatric Status Rating Scales; Retrospective Studies; Statistics, Nonparametric; tau Proteins

2007
Aging of the rat mesostriatal system: differences between the nigrostriatal and the mesolimbic compartments.
    Experimental neurology, 2007, Volume: 204, Issue:1

    The impairment of the mesostriatal dopaminergic system has been considered responsible for motor and affective disturbances associated with aging and a risk factor for Parkinson's disease. However, the basic mechanisms underlying this phenomenon are still unknown. Here we used biochemical, molecular and morphological techniques directed at detecting flaws in the dopamine synthesis route and signs of dopaminergic degeneration in the rat mesostriatal system during normal aging. We found two different age-related processes. One is characterized by a dopa decarboxylase decrease, and involves both the nigrostriatal and mesolimbic compartments, and is responsible for a moderate dopamine loss in the dorsal striatum, where other parameters of dopamine synthesis are not affected. The other is characterized by axonal degeneration with aggregation of phosphorylated forms of tyrosine hydroxylase (TH) and amyloid precursor protein in degenerate terminals, and alpha-synuclein in their original somata. This process is restricted to mesolimbic regions and is responsible for the decline of TH activity and l-dopa levels and the greater decrease in dopamine levels in this compartment. These findings suggest that both the nigrostriatal and the mesolimbic systems are vulnerable to aging, but in contrast to what occurs in Parkinson's disease, the mesolimbic system is more vulnerable to aging than the nigrostriatal one.

    Topics: Aging; alpha-Synuclein; Amyloid beta-Protein Precursor; Animals; Axons; Corpus Striatum; Dopa Decarboxylase; Dopamine; Levodopa; Limbic System; Male; Nerve Degeneration; Phosphorylation; Rats; Rats, Sprague-Dawley; Substantia Nigra; Tyrosine 3-Monooxygenase

2007
Multiple system atrophy in a patient with the spinocerebellar ataxia 3 gene mutation.
    Movement disorders : official journal of the Movement Disorder Society, 2007, Jan-15, Volume: 22, Issue:2

    The cerebellar variant of multiple system atrophy (MSA-C) has overlapping clinical features with the hereditary spinocerebellar ataxias (SCAs), but can usually be distinguished on a clinical basis. We describe a patient who developed a sporadic, late-onset, rapidly progressive neurodegenerative disorder consistent with MSA-C. Genetic testing, however, showed an abnormal expansion of one allele of the spinocerebellar ataxia 3 (SCA3) gene. The clinical impression of MSA-C was confirmed by identification of numerous alpha-synuclein-containing glial cytoplasmic inclusions on autopsy. These findings suggest that abnormal expansion of the SCA3 gene may be a risk factor for the development of MSA-C.

    Topics: Alleles; alpha-Synuclein; Ataxin-3; Autopsy; Cerebellum; DNA Repeat Expansion; Fatal Outcome; Female; Humans; Inclusion Bodies; Middle Aged; Multiple System Atrophy; Myelin Sheath; Nerve Degeneration; Nerve Tissue Proteins; Neuroglia; Nuclear Proteins; Point Mutation; Putamen; Repressor Proteins; Severity of Illness Index; Spinocerebellar Ataxias

2007
Insights into REM sleep behavior disorder pathophysiology in brainstem-predominant Lewy body disease.
    Sleep medicine, 2007, Volume: 8, Issue:1

    Rapid eye movement (REM) sleep behavior disorder (RBD) is a parasomnia reflecting changes in the brain, but which specific neuronal networks are involved in human RBD pathogenesis has not yet been determined. To date, only one case of idiopathic RBD has undergone autopsy, in which "incidental Lewy body disease" was found. Due to the severe neuronal loss and gliosis in the substantia nigra (SN) and locus ceruleus (LC) in this case, degeneration of brainstem monoaminergic neurons was postulated as the underlying substrate for RBD. Additional cases of idiopathic RBD with neuropathologic examination may help clarify which key brainstem structures are involved.. Case report with neuropathologic analysis.. A man with polysomnographically proven RBD (onset age 57 years), but no other neurologic signs or symptoms, underwent neuropathologic examination upon his death at age 72. Histopathologic analysis showed Lewy body disease, but no significant neuronal loss or gliosis was present in the SN or LC.. This case represents another example of Lewy body disease associated with RBD. The minimal degenerative changes in the SN and LC call into question the role of these nuclei in RBD, at least in our case. We suggest additional cases of idiopathic RBD undergo neuropathologic analyses to better delineate the neurologic substrate of this intriguing parasomnia.

    Topics: Aged; alpha-Synuclein; Brain Stem; Dyspnea; Electromyography; Gliosis; Humans; Lewy Body Disease; Male; Middle Aged; Nerve Degeneration; Neurofibrillary Tangles; Polysomnography; Severity of Illness Index; Sleep, REM

2007
Wild-type and mutant alpha-synuclein induce a multi-component gene expression profile consistent with shared pathophysiology in different transgenic mouse models of PD.
    Experimental neurology, 2007, Volume: 204, Issue:1

    The pathophysiological processes that cause Parkinson's disease (PD) affect dopamine neurons residing in the substantia nigra with devastating consequences for normal movement. One important gene involved in both familial and sporadic PD is alpha-synuclein. We have generated three strains of alpha-synuclein transgenic mice to study the pathologic consequences of the targeted expression of mutant or wild-type human alpha-synuclein in a model system. We have analyzed gene expression patterns in these mice using high throughput microarrays in anatomical regions implicated in disease (substantia nigra and brainstem). Our study reveals gene dosage-dependent dysregulation of several genes important for the dopaminergic phenotype in mice over-expressing wild-type human alpha-synuclein in the substantia nigra at time points preceding neuronal cell death. Analysis of mutant alpha-synuclein mice at a time point when pathology is advanced reveals several new candidate genes that may play a role in neuronal demise and/or protein accumulation.

    Topics: alpha-Synuclein; Animals; Brain Stem; Cell Death; Disease Models, Animal; Dopamine; Gene Dosage; Gene Expression; Gene Expression Profiling; Gene Expression Regulation; Humans; Male; Mice; Mice, Transgenic; Mutation; Nerve Degeneration; Neurons; Oligonucleotide Array Sequence Analysis; Parkinson Disease; Reproducibility of Results; Substantia Nigra

2007
Presynaptic alpha-synuclein aggregates, not Lewy bodies, cause neurodegeneration in dementia with Lewy bodies.
    The Journal of neuroscience : the official journal of the Society for Neuroscience, 2007, Feb-07, Volume: 27, Issue:6

    Lewy bodies, the pathological hallmark of dementia with Lewy bodies (DLB), are large juxtanuclear inclusions of aggregated alpha-synuclein. However, the small number of cortical Lewy bodies relative to the total neuron count does not correlate with the extent of cognitive impairment. In contrast to dopaminergic neurons in Parkinson's disease, nerve cell loss is usually less prevalent in the cortex of DLB, suggesting a different mechanism of neurodegeneration. Because antibodies used for immunodetection per se do not generally differentiate the aggregated from the physiological and monomeric isoform of alpha-synuclein, we developed the paraffin-embedded tissue (PET) blot and the protein aggregate filtration (PAF) assay for the sensitive and selective detection of alpha-synuclein aggregates in tissue slides and brain homogenates, respectively. In contrast to common immunohistochemistry, the PET blot detected an enormous number of small alpha-synuclein aggregates, which, in contrast to the few Lewy bodies, may explain the cognitive impairment in DLB. Using the PAF assay, we demonstrate that the absolute majority of alpha-synuclein aggregates are located at presynaptic terminals, suggesting a severe pathological impact on synaptic function. Indeed, parallel to the massive presynaptic accumulation of alpha-synuclein aggregates, we observed significant synaptic pathology with almost complete loss of dendritic spines at the postsynaptic area. Our results provide strong evidence for a novel concept of neurodegeneration for DLB in which synaptic dysfunction is caused by presynaptic accumulation of alpha-synuclein aggregates. This concept may also be valid for Parkinson's disease.

    Topics: alpha-Synuclein; Centrifugation, Density Gradient; Dendrites; Filtration; Frontal Lobe; Humans; Inclusion Bodies; Lewy Bodies; Lewy Body Disease; Nerve Degeneration; Paraffin Embedding; Presynaptic Terminals; Synaptic Transmission; Synaptosomes

2007
Whole genome expression analyses of single- and double-knock-out mice implicate partially overlapping functions of alpha- and gamma-synuclein.
    Neurogenetics, 2007, Volume: 8, Issue:2

    alpha-Synuclein has been implicated in the pathogenesis of Parkinson's disease. The function of alpha-synuclein has not been deciphered yet; however, it might play a role in vesicle function, transport, or as a chaperone. alpha-Synuclein belongs to a family of three proteins, which includes beta- and gamma-synuclein. gamma-Synuclein shares 60% similarity with alpha-synuclein. Similar to alpha-synuclein, a physiological function for gamma-synuclein has not been defined yet, but it has been implicated in tumorgenesis and neurodegeneration. Interestingly, neither alpha- (SNCA(-/-)), gamma- (SNCG(-/-)), nor alpha/gamma- (SNCA_G(-/-)) deficient mice are present with any obvious phenotype. Using microarray analysis, we thus investigated whether deficiency of alpha- and gamma-synuclein leads to similar compensatory mechanisms at the RNA level and whether similar transcriptional signatures are altered in the brain. Sixty-five genes were differentially expressed in all mice. SNCA(-/-) mice and SNCG(-/-) mice shared 84 differentially expressed genes, SNCA(-/-) and SNCA_G(-/-) expressed 79 genes, and SNCG(-/-) and SNCA_G(-/-) expressed 148 genes. For many of the physiological pathways such as dopamine receptor signaling (down-regulated), cellular development, nervous system function, and cell death (up-regulated), we found groups of genes that were similarly altered in SNCA(-/-) and SNCG(-/-) mice. In one of the pathways altered in both models, we found Mapk1 as the core transcript. Other gene groups, however, such as TGF-beta signaling and apoptosis pathways genes were significantly up-regulated in the SNCA(-/-) mice but down-regulated in SNCG(-/-) mice. beta-synuclein expression was not significantly altered in any of the models.

    Topics: alpha-Synuclein; Animals; Brain; gamma-Synuclein; Genome; Mice; Mice, Inbred C57BL; Mice, Knockout; Nerve Degeneration; Oligonucleotide Array Sequence Analysis; Reverse Transcriptase Polymerase Chain Reaction; RNA; Transcription, Genetic

2007
Microglial activation mediates neurodegeneration related to oligodendroglial alpha-synucleinopathy: implications for multiple system atrophy.
    Movement disorders : official journal of the Movement Disorder Society, 2007, Nov-15, Volume: 22, Issue:15

    The role of microglial activation in multiple system atrophy (MSA) was investigated in a transgenic mouse model featuring oligodendroglial alpha-synuclein inclusions and loss of midbrain dopaminergic neurons by means of histopathology and morphometric analysis. Our findings demonstrate early progressive microglial activation in substantia nigra pars compacta (SNc) associated with increased expression of iNOS and correlating with dopaminergic neuronal loss. Suppression of microglial activation by early long-term minocycline treatment protected dopaminergic SNc neurons. The results suggest that oligodendroglial overexpression of alpha-synuclein may induce neuroinflammation related to nitrosive stress which is likely to contribute to neurodegeneration in MSA. Further, we detected increased toll-like receptor 4 immunoreactivity in both transgenic mice and MSA brains indicating a possible signaling pathway in MSA which needs to be further studied as a candidate target for neuroprotective interventions.

    Topics: Aged; alpha-Synuclein; Animals; Corpus Striatum; Disease Progression; Dopamine; Female; Humans; Male; Mice; Mice, Inbred C57BL; Mice, Transgenic; Microglia; Middle Aged; Minocycline; Multiple System Atrophy; Nerve Degeneration; Neuroprotective Agents; Nitric Oxide Synthase Type II; Oligodendroglia; Toll-Like Receptor 4

2007
Functional protein kinase arrays reveal inhibition of p-21-activated kinase 4 by alpha-synuclein oligomers.
    Journal of neurochemistry, 2007, Volume: 103, Issue:6

    There is increasing evidence that aggregation of alpha-synuclein contributes to the functional and structural deterioration in the CNS of Parkinson's disease patients and transgenic animal models. alpha-Synuclein binds to various cellular proteins and aggregated alpha-synuclein species may affect their physiological function. In the present study, we used protein arrays spotted with 178 active human kinases for a large-scale analysis of the effects of recombinant alpha-synuclein on kinase activities. Incubation with globular alpha-synuclein oligomers significantly inhibited autophosphorylation of p21-activated kinase (PAK4) compared to treatment with monomeric alpha-synuclein or beta-synuclein. A concentration-dependent inhibition was also observed in a solution-based kinase assay. To show in vivo relevance, we analyzed brainstem protein extracts from alpha-synuclein (A30P) transgenic mice where accumulation of alpha-synuclein oligomers has been demonstrated. By immunoblotting using a phospho-specific antibody, we detected a significant decline in phosphorylation of LIM kinase 1, a physiological substrate for PAK4. Suppression of PAK activity by amyloid-beta oligomers has been reported in Alzheimer's disease. Thus, PAKs may represent a target for various neurotoxic protein oligomers, and signaling deficits may contribute to the behavioral defects in chronic neurodegenerative diseases.

    Topics: alpha-Synuclein; Animals; beta-Synuclein; Brain Stem; Dose-Response Relationship, Drug; Enzyme Activation; Enzyme Inhibitors; Female; Inclusion Bodies; Lim Kinases; Macromolecular Substances; Mice; Mice, Inbred C57BL; Mice, Transgenic; Nerve Degeneration; Neurodegenerative Diseases; Neurons; p21-Activated Kinases; Phosphorylation; Polymers; Protein Array Analysis

2007
Disruption of dopamine homeostasis underlies selective neurodegeneration mediated by alpha-synuclein.
    The European journal of neuroscience, 2007, Volume: 26, Issue:11

    A key challenge in Parkinson's disease research is to understand mechanisms underlying selective degeneration of dopaminergic neurons mediated by genetic factors such as alpha-synuclein (alpha-Syn). The present study examined whether dopamine (DA)-dependent oxidative stress underlies alpha-Syn-mediated neurodegeneration using Drosophila primary neuronal cultures. Green fluorescent protein (GFP) was used to identify live dopaminergic neurons in primary cultures prepared on a marked photoetched coverslip, which allowed us to repeatedly access preidentified dopaminergic neurons at different time points in a non-invasive manner. This live tracking of GFP-marked dopaminergic neurons revealed age-dependent neurodegeneration mediated by a mutant human alpha-Syn (A30P). Degeneration was rescued when alpha-Syn neuronal cultures were incubated with 1 mm glutathione from Day 3 after culturing. Furthermore, depletion of cytoplasmic DA by 100 microm alpha-methyl-p-tyrosine completely rescued the early stage of alpha-Syn-mediated dopaminergic cell loss, demonstrating that DA plays a major role in oxidative stress-dependent neurodegeneration mediated by alpha-Syn. In contrast, overexpression of a Drosophila tyrosine hydroxylase gene (dTH1) alone caused DA neurodegeneration by enhanced DA synthesis in the cytoplasm. Age-dependent dopaminergic cell loss was comparable in alpha-Syn vs dTH1-overexpressed neuronal cultures, indicating that increased DA levels in the cytoplasm is a critical change downstream of mutant alpha-Syn function. Finally, overexpression of a Drosophila vesicular monoamine transporter rescued alpha-Syn-mediated neurodegeneration through enhanced sequestration of cytoplasmic DA into synaptic vesicles, further indicating that a main cause of selective neurodegeneration is alpha-Syn-induced disruption of DA homeostasis. All of these results demonstrate that elevated cytoplasmic DA is a main factor underlying the early stage of alpha-Syn-mediated neurodegeneration.

    Topics: Alanine; alpha-Methyltyrosine; alpha-Synuclein; Animals; Animals, Genetically Modified; Cells, Cultured; Dopamine; Drosophila; Embryo, Nonmammalian; Enzyme Inhibitors; Glutathione; Green Fluorescent Proteins; Homeostasis; Humans; Indoles; Mutation; Nerve Degeneration; Proline; Receptors, Dopamine D1; Time Factors; Tyrosine 3-Monooxygenase; Vesicular Monoamine Transport Proteins

2007
Parkinson's disease alpha-synuclein transgenic mice develop neuronal mitochondrial degeneration and cell death.
    The Journal of neuroscience : the official journal of the Society for Neuroscience, 2006, Jan-04, Volume: 26, Issue:1

    Alpha-synuclein (alpha-Syn) is enriched in nerve terminals. Two mutations in the alpha-Syn gene (Ala53--> Thr and Ala30--> Pro) occur in autosomal dominant familial Parkinson's disease. Mice overexpressing the human A53T mutant alpha-Syn develop a severe movement disorder, paralysis, and synucleinopathy, but the mechanisms are not understood. We examined whether transgenic mice expressing human wild-type or familial Parkinson's disease-linked A53T or A30P mutant alpha-syn develop neuronal degeneration and cell death. Mutant mice were examined at early- to mid-stage disease and at near end-stage disease. Age-matched nontransgenic littermates were controls. In A53T mice, neurons in brainstem and spinal cord exhibited large axonal swellings, somal chromatolytic changes, and nuclear condensation. Spheroid eosinophilic Lewy body-like inclusions were present in the cytoplasm of cortical neurons and spinal motor neurons. These inclusions contained human alpha-syn and nitrated synuclein. Motor neurons were depleted (approximately 75%) in A53T mice but were affected less in A30P mice. Axonal degeneration was present in many regions. Electron microscopy confirmed the cell and axonal degeneration and revealed cytoplasmic inclusions in dendrites and axons. Some inclusions were degenerating mitochondria and were positive for humanalpha-syn. Mitochondrial complex IV and V proteins were at control levels, but complex IV activity was reduced significantly in spinal cord. Subsets of neurons in neocortex, brainstem, and spinal cord ventral horn were positive for terminal deoxynucleotidyl transferase-mediated biotinylated UTP nick end labeling, cleaved caspase-3, and p53. Mitochondria in neurons had terminal deoxynucleotidyl transferase-mediated biotinylated UTP nick end labeling-positive matrices and p53 at the outer membrane. Thus, A53T mutant mice develop intraneuronal inclusions, mitochondrial DNA damage and degeneration, and apoptotic-like death of neocortical, brainstem, and motor neurons.

    Topics: Alanine; alpha-Synuclein; Animals; Brain Stem; Cell Death; DNA Damage; Humans; Mice; Mice, Transgenic; Mitochondria; Mutation; Neocortex; Nerve Degeneration; Parkinson Disease; Threonine

2006
Intersecting pathways to neurodegeneration in Parkinson's disease: effects of the pesticide rotenone on DJ-1, alpha-synuclein, and the ubiquitin-proteasome system.
    Neurobiology of disease, 2006, Volume: 22, Issue:2

    Sporadic Parkinson's disease (PD) is most likely caused by a combination of environmental exposures and genetic susceptibilities, although there are rare monogenic forms of the disease. Mitochondrial impairment at complex I, oxidative stress, alpha-synuclein aggregation, and dysfunctional protein degradation, have been implicated in PD pathogenesis, but how they are related to each other is unclear. To further evaluated PD pathogenesis here, we used in vivo and in vitro models of chronic low-grade complex I inhibition with the pesticide rotenone. Chronic rotenone exposure in vivo caused oxidative modification of DJ-1, accumulation of alpha-synuclein, and proteasomal impairment. Interestingly, the effects become more regionally restricted such that systemic complex I inhibition eventually results in highly selective degeneration of the nigrostriatal pathway. DJ-1 modifications, alpha-synuclein accumulation, and proteasomal dysfunction were also seen in vitro and these effects could be prevented with alpha-tocopherol. Thus, chronic exposure to a pesticide and mitochondrial toxin brings into play three systems, DJ-1, alpha-synuclein, and the ubiquitin-proteasome system, and implies that mitochondrial dysfunction and oxidative stress link environmental and genetic forms of the disease.

    Topics: alpha-Synuclein; Animals; Cell Line, Tumor; Disease Models, Animal; Electron Transport Complex I; Energy Metabolism; Humans; Insecticides; Male; Mitochondria; Nerve Degeneration; Neurons; Oncogene Proteins; Oxidative Stress; Parkinsonian Disorders; Peroxiredoxins; Proteasome Endopeptidase Complex; Protein Deglycase DJ-1; Rats; Rats, Inbred Lew; Rotenone; Signal Transduction; Substantia Nigra; Ubiquitin

2006
Alpha-synuclein facilitates the toxicity of oxidized catechol metabolites: implications for selective neurodegeneration in Parkinson's disease.
    FEBS letters, 2006, Apr-03, Volume: 580, Issue:8

    Free radicals, including dopamine (DA)-oxidized metabolites, have long been implicated in pathogenesis of Parkinson's disease (PD). However, the relationships between such oxidative stresses and alpha-synuclein (alpha-S), a major constituent of Lewy bodies, remain unknown. In this study, we established neuronal cells that constitutively express alpha-S and tetracycline-regulated tyrosinase. While tyrosinase overexpression induced apoptosis, co-expression of wild type or A53T mutant human alpha-S with tyrosinase further exacerbated cell death. In this process, the formation of alpha-S oligomers and the reduction in mitochondrial membrane potential were demonstrated. This cellular model may reconstitute the pathological metabolism of alpha-S in the synucleinopathy and provide a useful tool to explore possible pathomechanisms of nigral degeneration in PD.

    Topics: alpha-Synuclein; Apoptosis; Catechols; Cells, Cultured; Comet Assay; DNA Damage; Enzyme Activation; Gene Expression; Humans; Intracellular Membranes; Membrane Potentials; Mitochondrial Membranes; Mitogen-Activated Protein Kinases; Molecular Weight; Monophenol Monooxygenase; Multiprotein Complexes; Mutation; Nerve Degeneration; Oxidation-Reduction; Parkinson Disease

2006
Identification of rotenone-induced modifications in alpha-synuclein using affinity pull-down and tandem mass spectrometry.
    Analytical chemistry, 2006, Apr-01, Volume: 78, Issue:7

    Parkinson's disease is a movement disorder that results from a loss of dopaminergic neurons in the substantia nigra. The disease is characterized by mitochondrial dysfunction, oxidative stress, and the presence of "Lewy body" inclusions enriched with aggregated forms of alpha-synuclein, a presynaptic protein. Although alpha-synuclein is modified at various sites in Lewy bodies, it is unclear how sequence-specific posttranslational modifications modulate the aggregation of the protein in oxidatively stressed neurons. To begin to address this problem, we developed an affinity pull-down/mass spectrometry method to characterize the primary structure of histidine-tagged alpha-synuclein isolated from catecholaminergic neurons. Using this method, we mapped posttranslational modifications of alpha-synuclein from untreated neurons and neurons exposed to rotenone, an inhibitor of mitochondrial complex I. Various posttranslational modifications suggestive of oxidative damage or repair were identified in a region comprising a 20-residue stretch in the C-terminal part of the protein. The results indicate that alpha-synuclein is subject to discrete posttranslational modifications in neurons with impaired mitochondrial function. Our affinity pull-down/mass spectrometry method is a useful tool to examine how specific modifications of alpha-synuclein contribute to neurologic disorders such as Parkinson's disease.

    Topics: alpha-Synuclein; Amino Acid Sequence; Animals; Base Sequence; Cells, Cultured; Mitochondria; Molecular Sequence Data; Nerve Degeneration; Nervous System Diseases; Neurons; Oxidative Stress; Parkinson Disease; PC12 Cells; Protein Processing, Post-Translational; Rats; Rotenone; Tandem Mass Spectrometry

2006
Plasma membrane ion permeability induced by mutant alpha-synuclein contributes to the degeneration of neural cells.
    Journal of neurochemistry, 2006, Volume: 97, Issue:4

    Mutations in alpha-synuclein cause some cases of familial Parkinson's disease (PD), but the mechanism by which alpha-synuclein promotes degeneration of dopamine-producing neurons is unknown. We report that human neural cells expressing mutant alpha-synuclein (A30P and A53T) have higher plasma membrane ion permeability. The higher ion permeability caused by mutant alpha-synuclein would be because of relatively large pores through which most cations can pass non-selectively. Both the basal level of [Ca2+]i and the Ca2+ response to membrane depolarization are greater in cells expressing mutant alpha-synuclein. The membrane permeable Ca2+ chelator BAPTA-AM significantly protected the cells against oxidative stress, whereas neither L-type (nifedipine) nor N-type (omega-conotoxin-GVIA) Ca2+ channel blockers protected the cells. These findings suggest that the high membrane ion permeability caused by mutant alpha-synuclein may contribute to the degeneration of neurons in PD.

    Topics: alpha-Synuclein; Calcium; Calcium Channel Blockers; Calcium Channels; Calcium Signaling; Cell Death; Cell Line, Tumor; Cell Membrane; Cell Membrane Permeability; Chelating Agents; Dopamine; Humans; Mutation; Nerve Degeneration; Neurons; Parkinson Disease

2006
Transduced Tat-alpha-synuclein protects against oxidative stress in vitro and in vivo.
    Journal of biochemistry and molecular biology, 2006, May-31, Volume: 39, Issue:3

    Parkinson's disease (PD) is a common neurodegenerative disorder and is characterized by the progressive loss of dopaminergic neurons in the substantia nigra. Although many studies showed that the aggregation of alpha-synuclein might be involved in the pathogenesis of PD, its protective properties against oxidative stress remain to be elucidated. In this study, human wild type and mutant alpha-synuclein genes were fused with a gene fragment encoding the nine amino acid transactivator of transcription (Tat) protein transduction domain of HIV-1 in a bacterial expression vector to produce a genetic in-frame WT Tat-alpha-synuclein (wild type) and mutant Tat-alpha-synucleins (mutants; A30P and A53T), respectively, and we investigated the protective effects of wild type and mutant Tat-alpha-synucleins in vitro and in vivo. WT Tat-alpha-synuclein rapidly transduced into an astrocyte cells and protected the cells against paraquat induced cell death. However, mutant Tat-alpha-synucleins did not protect at all. In the mice models exposed to the herbicide paraquat, the WT Tat-alpha-synuclein completely protected against dopaminergic neuronal cell death, whereas mutants failed in protecting against oxidative stress. We found that these protective effects were characterized by increasing the expression level of heat shock protein 70 (HSP70) in the neuronal cells and this expression level was dependent on the concentration of transduced WT Tat-alpha-synuclein. These results suggest that transduced Tat-alpha-synuclein might protect cell death from oxidative stress by increasing the expression level of HSP70 in vitro and in vivo and this may be of potential therapeutic benefit in the pathogenesis of PD.

    Topics: alpha-Synuclein; Animals; Astrocytes; Cell Death; Cell Survival; Gene Products, tat; Genetic Vectors; HSP70 Heat-Shock Proteins; Humans; Male; Mice; Mice, Inbred C57BL; Mutation; Nerve Degeneration; Neurons; Oxidative Stress; Paraquat; Parkinsonian Disorders; Protein Transport; Recombinant Fusion Proteins; Transduction, Genetic

2006
Comparison of alpha-synuclein immunoreactivity and protein levels in ischemic hippocampal CA1 region between adult and aged gerbils and correlation with Cu,Zn-superoxide dismutase.
    Neuroscience research, 2006, Volume: 55, Issue:4

    In this study, we examined changes in the level and immunoreactivity of alpha-synuclein in the hippocampal CA1 region of adult (6 months old) and aged (24 months old) gerbils after 5 min of transient forebrain ischemia. The delayed neuronal death of CA1 pyramidal cells in adult gerbils was severer than that in aged gerbils 4 days after ischemia/reperfusion. Alpha-synuclein immunoreactivity in the CA1 region of adult and aged gerbils significantly changed after ischemia. In control animals, alpha-synuclein immunoreactivity and level in the aged-gerbil CA1 region were higher than those in the adult-gerbil CA1 region. In both adult and aged gerbils, alpha-synuclein immunoreactivity and level started to increase 3h after ischemia, and they were highest 1 day after ischemia. Thereafter, alpha-synuclein immunoreactivity and level decreased with time after ischemia. We also observed the effects of Cu,Zn-superoxide dismutase (SOD1) on ischemic damage using the Pep-1 transduction domain. Alpha-synuclein level in the CA1 region was lower in Pep-1-SOD1-treated adult and aged gerbils than in vehicle-treated adult and aged gerbils. We conclude that neuronal loss in the hippocampal CA1 region of adult gerbils was more prominent than that in aged gerbils 4 days after ischemia/reperfusion. The higher level of alpha-synuclein in the aged-gerbil CA1 region than that in the adult-gerbil CA1 region may be associated with the earlier induction of reactive oxygen species, and Pep-1-SOD1 potentially and reversibly inhibits the accumulation of alpha-synuclein in the CA1 region after transient ischemia.

    Topics: Aging; alpha-Synuclein; Animals; Brain Ischemia; Cell Death; Disease Models, Animal; Down-Regulation; Gerbillinae; Hippocampus; Immunohistochemistry; Male; Nerve Degeneration; Protein Structure, Tertiary; Pyramidal Cells; Reactive Oxygen Species; Reperfusion Injury; Superoxide Dismutase; Time Factors

2006
Alpha-synuclein blocks ER-Golgi traffic and Rab1 rescues neuron loss in Parkinson's models.
    Science (New York, N.Y.), 2006, Jul-21, Volume: 313, Issue:5785

    Alpha-synuclein (alphaSyn) misfolding is associated with several devastating neurodegenerative disorders, including Parkinson's disease (PD). In yeast cells and in neurons alphaSyn accumulation is cytotoxic, but little is known about its normal function or pathobiology. The earliest defect following alphaSyn expression in yeast was a block in endoplasmic reticulum (ER)-to-Golgi vesicular trafficking. In a genomewide screen, the largest class of toxicity modifiers were proteins functioning at this same step, including the Rab guanosine triphosphatase Ypt1p, which associated with cytoplasmic alphaSyn inclusions. Elevated expression of Rab1, the mammalian YPT1 homolog, protected against alphaSyn-induced dopaminergic neuron loss in animal models of PD. Thus, synucleinopathies may result from disruptions in basic cellular functions that interface with the unique biology of particular neurons to make them especially vulnerable.

    Topics: alpha-Synuclein; Animals; Animals, Genetically Modified; Caenorhabditis elegans; Cell Survival; Cells, Cultured; Disease Models, Animal; Dopamine; Drosophila; Endoplasmic Reticulum; Gene Expression; Gene Library; Golgi Apparatus; Humans; Mice; Nerve Degeneration; Neurons; Parkinsonian Disorders; Proteasome Endopeptidase Complex; Protein Folding; Protein Transport; Proteins; rab GTP-Binding Proteins; rab1 GTP-Binding Proteins; Rats; Recombinant Fusion Proteins; Saccharomyces cerevisiae; Saccharomyces cerevisiae Proteins

2006
Ventral tegmental area dopamine neurons are resistant to human mutant alpha-synuclein overexpression.
    Neurobiology of disease, 2006, Volume: 23, Issue:3

    Parkinson's disease (PD) is characterized by the formation of intracytoplasmic inclusions, which contain alpha-synuclein (alpha-syn) protein. While most profound neurodegeneration is seen in the dopamine (DA) synthesizing neurons located in the ventral midbrain, it is unclear why some DA cell groups are more susceptible than others. In the midbrain, the degeneration of the substantia nigra (SN) DA neurons is severe, whereas the involvement of the ventral tegmental area (VTA) neurons is relatively spared. In the present study, we overexpressed human A53T alpha-syn in the VTA neurons and found that A53T toxicity did not affect their survival. There was, however, a mild functional impairment seen as altered open field locomotor activity. Overexpression of A53T in the SN, on the other hand, led to profound cell loss. These results suggest that the selective susceptibility of nigral DA neurons is at least in part associated with factor(s) involved in handling of alpha-syn that is not shared by the VTA neurons. Secondly, these results highlight the fact that impaired but surviving neurons can have a substantial impact on DA-dependent behavior and should therefore be considered as a critical part of animal models where novel therapeutic interventions are tested.

    Topics: alpha-Synuclein; Animals; Dependovirus; Disease Models, Animal; Dopamine; Female; Gene Expression; Genetic Predisposition to Disease; Genetic Vectors; Humans; Inclusion Bodies; Mutation; Nerve Degeneration; Neurons; Parkinson Disease; Rats; Rats, Sprague-Dawley; Substantia Nigra; Tyrosine 3-Monooxygenase; Up-Regulation; Ventral Tegmental Area

2006
Relationship among alpha-synuclein accumulation, dopamine synthesis, and neurodegeneration in Parkinson disease substantia nigra.
    Journal of neuropathology and experimental neurology, 2006, Volume: 65, Issue:8

    The histologic hallmark of Parkinson disease (PD) is loss of pigmented neurons in the substantia nigra (SN) and locus ceruleus (LC) with accumulation of alpha-synuclein (alphaS). It has been reported that tyrosine hydroxylase (TH)-negative pigmented neurons are present in these nuclei of patients with PD. However, the relationship between TH immunoreactivity and alphaS accumulation remains uncertain. We immunohistochemically examined the SN and LC from patients with PD (n = 10) and control subjects (n = 7). A correlation study indicated a close relationship among decreased TH immunoreactivity, alphaS accumulation, and neuronal loss. In addition, 10% of pigmented neurons in the SN and 54.9% of those in the LC contained abnormal alphaS aggregates. Moreover, 82.3% of pigmented neurons bearing alphaS aggregates in the SN and 39.2% of those in the LC lacked TH immunoreactivity, suggesting that pigmented neurons in the SN have a greater tendency to lack TH activity than those in the LC. Recent studies have shown that this decrease of TH activity leads to a decrease of cytotoxic substances and that decreased dopamine synthesis leads to a reduction of cytotoxic alphaS oligomers. Therefore, the decrease of TH immunoreactivity in pigmented neurons demonstrated here can be considered to represent a cytoprotective mechanism in PD.

    Topics: Aged; Aged, 80 and over; alpha-Synuclein; Cell Death; Cell Survival; Cytoprotection; Dopamine; Down-Regulation; Humans; Immunohistochemistry; Inclusion Bodies; Locus Coeruleus; Middle Aged; Nerve Degeneration; Neurons; Parkinson Disease; Substantia Nigra; Tyrosine 3-Monooxygenase

2006
Cytosolic catechols inhibit alpha-synuclein aggregation and facilitate the formation of intracellular soluble oligomeric intermediates.
    The Journal of neuroscience : the official journal of the Society for Neuroscience, 2006, Sep-27, Volume: 26, Issue:39

    Aberrant aggregation of alpha-synuclein (alpha-syn) to form fibrils and insoluble aggregates has been implicated in the pathogenic processes of many neurodegenerative diseases. Despite the dramatic effects of dopamine in inhibiting the formation of alpha-syn fibrils by stabilization of oligomeric intermediates in cell-free systems, no studies have examined the effects of intracellular dopamine on alpha-syn aggregation. To study this process and its association with neurodegeneration, intracellular catechol levels were increased to various levels by expressing different forms of tyrosine hydroxylase, in cells induced to form alpha-syn aggregates. The increase in the steady-state dopamine levels inhibited the formation of alpha-syn aggregates and induced the formation of innocuous oligomeric intermediates. Analysis of transgenic mice expressing the disease-associated A53T mutant alpha-syn revealed the presence of oligomeric alpha-syn in nondegenerating dopaminergic neurons that do contain insoluble alpha-syn. These data indicate that intraneuronal dopamine levels can be a major modulator of alpha-syn aggregation and inclusion formation, with important implications on the selective degeneration of these neurons in Parkinson's disease.

    Topics: 3,4-Dihydroxyphenylacetic Acid; alpha-Synuclein; Amino Acid Substitution; Animals; Catechols; Cell Differentiation; Cell Line, Tumor; Cerebral Cortex; Corpus Striatum; Cytosol; Dopamine; Humans; Levodopa; Mice; Mice, Transgenic; Mutation, Missense; Nerve Degeneration; Neuroblastoma; Oxidation-Reduction; Parkinson Disease; Parkinsonian Disorders; Protein Conformation; Recombinant Fusion Proteins; Solubility; Transfection; Tretinoin; Tyrosine 3-Monooxygenase

2006
Impairment of microtubule-dependent trafficking by overexpression of alpha-synuclein.
    The European journal of neuroscience, 2006, Volume: 24, Issue:11

    Abnormal accumulation of alpha-synuclein (alpha-syn) has been linked to several neurological disorders, including Parkinson's disease (PD). However, the underlying mechanism by which alpha-syn accumulation affects neuronal function and survival remains unknown. Here, we provide data suggesting a possible effect of aggregated alpha-syn on the microtubule (MT) network. Consistent with the MT dysfunction, we also observed other degenerative changes, such as neuritic degeneration, trafficking defects, and Golgi fragmentation, which are common pathological features shared by many human neurodegenerative diseases. Neuritic degeneration and Golgi fragmentation were confirmed in primary cultures of dorsal root ganglia (DRG) neurons overexpressing alpha-syn. This effect of alpha-syn seems to have some selectivity to the MT system, as actin microfilaments and MT-independent trafficking remain unaffected. Within the degenerating neurites, we found numerous spherical co-aggregates of alpha-syn and tubulins, from which actin was excluded. These studies suggest that the MT system is a potential target of alpha-syn, and impairment of this system might have impacts on neuronal structure and function.

    Topics: Actin Cytoskeleton; alpha-Synuclein; Animals; Biological Transport, Active; Cell Line, Tumor; Chlorocebus aethiops; COS Cells; Ganglia, Spinal; Golgi Apparatus; Humans; Inclusion Bodies; Microtubules; Nerve Degeneration; Nervous System; Neurites; Neurons; Neurons, Afferent; Parkinson Disease; Protein Transport; Rats; Tubulin; Up-Regulation; Viral Proteins

2006
Aggregated alpha-synuclein activates microglia: a process leading to disease progression in Parkinson's disease.
    FASEB journal : official publication of the Federation of American Societies for Experimental Biology, 2005, Volume: 19, Issue:6

    A growing body of evidence indicates that an inflammatory process in the substantia nigra, characterized by activation of resident microglia, likely either initiates or aggravates nigral neurodegeneration in Parkinson's disease (PD). To study the mechanisms by which nigral microglia are activated in PD, the potential role of alpha-synuclein (a major component of Lewy bodies that can cause neurodegeneration when aggregated) in microglial activation was investigated. The results demonstrated that in a primary mesencephalic neuron-glia culture system, extracellular aggregated human alpha-synuclein indeed activated microglia; microglial activation enhanced dopaminergic neurodegeneration induced by aggregated alpha-synuclein. Furthermore, microglial enhancement of alpha-synuclein-mediated neurotoxicity depended on phagocytosis of alpha-synuclein and activation of NADPH oxidase with production of reactive oxygen species. These results suggest that nigral neuronal damage, regardless of etiology, may release aggregated alpha-synuclein into substantia nigra, which activates microglia with production of proinflammatory mediators, thereby leading to persistent and progressive nigral neurodegeneration in PD. Finally, NADPH oxidase could be an ideal target for potential pharmaceutical intervention, given that it plays a critical role in alpha-synuclein-mediated microglial activation and associated neurotoxicity.

    Topics: alpha-Synuclein; Animals; Astrocytes; Brain; Cells, Cultured; Dinoprostone; Dopamine; Embryo, Mammalian; Enzyme Activation; gamma-Aminobutyric Acid; Humans; Immunohistochemistry; Mesencephalon; Mice; Mice, Knockout; Microglia; NADPH Oxidases; Nerve Degeneration; Neuroglia; Neurons; Nitric Oxide; Nitrites; Oxidative Stress; Parkinson Disease; Phagocytosis; Polymers; Rats; Rats, Inbred F344; Reactive Oxygen Species; Recombinant Proteins; Substantia Nigra; Tritium; Tumor Necrosis Factor-alpha; Tyrosine 3-Monooxygenase

2005
Nitration and increased alpha-synuclein expression associated with dopaminergic neurodegeneration in equine pituitary pars intermedia dysfunction.
    Journal of neuroendocrinology, 2005, Volume: 17, Issue:2

    Equine pituitary pars intermedia dysfunction (PPID) is a spontaneously occurring progressive disease affecting aged horses and ponies. The pathogenesis of PPID is poorly understood, but the available evidence supports a loss of dopaminergic inhibition of the melanotropes of the pars intermedia. Horses with PPID have increased plasma concentrations of pars intermedia pro-opiomelanocortin-derived peptides that decrease in response to dopamine or dopamine agonist administration. Dopamine and dopamine metabolite concentrations are decreased in the pars intermedia of affected horses compared to age-matched control horses. Horses with disease that are treated with the dopamine agonist pergolide show improvement in clinical signs and normalisation of diagnostic test results. In the present study, immunohistochemical evaluation of pituitary and hypothalamic tissue demonstrated reduced tyrosine hydroxylase immunoreactivity in affected horses compared to age-matched and young controls, supporting the role of dopaminergic neurodegeneration in PPID. In addition, immunohistochemical evaluation revealed an increase in the oxidative stress marker, 3-nitrotyrosine and in nerve terminal protein, alpha-synuclein that colocalised in the pars intermedia of horses with disease. These findings suggest a role for nitration of overexpressed alpha-synuclein in the pathogenesis of neurodegeneration in PPID.

    Topics: alpha-Synuclein; Animals; Blotting, Western; Chronic Disease; Dopamine; Horse Diseases; Horses; Immunohistochemistry; Nerve Degeneration; Nerve Tissue Proteins; Nitrogen; Oxidative Stress; Pituitary ACTH Hypersecretion; Pituitary Gland; Synucleins; Tyrosine; Tyrosine 3-Monooxygenase

2005
Torsin-mediated protection from cellular stress in the dopaminergic neurons of Caenorhabditis elegans.
    The Journal of neuroscience : the official journal of the Society for Neuroscience, 2005, Apr-13, Volume: 25, Issue:15

    Parkinson's disease (PD) is linked genetically to proteins that function in the management of cellular stress resulting from protein misfolding and oxidative damage. Overexpression or mutation of alpha-synuclein results in the formation of Lewy bodies and neurodegeneration of dopaminergic (DA) neurons. Human torsinA, mutations in which cause another movement disorder termed early-onset torsion dystonia, is highly expressed in DA neurons and is also a component of Lewy bodies. Previous work has established torsins as having molecular chaperone activity. Thus, we examined the ability of torsinA to manage cellular stress within DA neurons of the nematode Caenorhabditis elegans. Worm DA neurons undergo a reproducible pattern of neurodegeneration after treatment with 6-hydroxydopamine (6-OHDA), a neurotoxin commonly used to model PD. Overexpression of torsins in C. elegans DA neurons results in dramatic suppression of neurodegeneration after 6-OHDA treatment. In contrast, expression of either dystonia-associated mutant torsinA or combined overexpression of wild-type and mutant torsinA yielded greatly diminished neuroprotection against 6-OHDA. We further demonstrated that torsins seem to protect DA neurons from 6-OHDA through downregulating protein levels of the dopamine transporter (DAT-1) in vivo. Additionally, we determined that torsins protect robustly against DA neurodegeneration caused by overexpression of alpha-synuclein. Using mutant nematodes lacking DAT-1 function, we also showed that torsin neuroprotection from alpha-synuclein-induced degeneration occurs in a manner independent of this transporter. Together, these data have mechanistic implications for movement disorders, because our results demonstrate that torsin proteins have the capacity to manage sources of cellular stress within DA neurons.

    Topics: Adrenergic Agents; alpha-Synuclein; Analysis of Variance; Animals; Animals, Genetically Modified; Blotting, Western; Caenorhabditis elegans; Cationic Amino Acid Transporter 2; Cell Count; Disease Models, Animal; Dopamine; Dopamine Plasma Membrane Transport Proteins; Drug Interactions; Embryo, Mammalian; Embryo, Nonmammalian; Fluorescent Antibody Technique; Gene Expression Regulation; Green Fluorescent Proteins; Humans; Molecular Chaperones; Mutagenesis; Nerve Degeneration; Neurons; Oxidopamine; Time Factors

2005
Alpha-synuclein phosphorylation controls neurotoxicity and inclusion formation in a Drosophila model of Parkinson disease.
    Nature neuroscience, 2005, Volume: 8, Issue:5

    Alpha-synuclein is phosphorylated at serine 129 (Ser129) in intracellular protein aggregates called Lewy bodies. These inclusion bodies are the characteristic pathologic lesions of Parkinson disease. Here we define the role of phosphorylation of Ser129 in alpha-synuclein toxicity and inclusion formation using a Drosophila model of Parkinson disease. Mutation of Ser129 to alanine to prevent phosphorylation completely suppresses dopaminergic neuronal loss produced by expression of human alpha-synuclein. In contrast, altering Ser129 to the negatively charged residue aspartate, to mimic phosphorylation, significantly enhances alpha-synuclein toxicity. The G protein-coupled receptor kinase 2 (Gprk2) phosphorylates Ser129 in vivo and enhances alpha-synuclein toxicity. Blocking phosphorylation at Ser129 substantially increases aggregate formation. Thus Ser129 phosphorylation status is crucial in mediating alpha-synuclein neurotoxicity and inclusion formation. Because increased number of inclusion bodies correlates with reduced toxicity, inclusion bodies may protect neurons from alpha-synuclein toxicity.

    Topics: alpha-Synuclein; Amino Acid Substitution; Animals; Animals, Genetically Modified; Aspartic Acid; beta-Adrenergic Receptor Kinases; Brain; Central Nervous System; Cyclic AMP-Dependent Protein Kinases; Disease Models, Animal; Dopamine; Drosophila; Drosophila Proteins; G-Protein-Coupled Receptor Kinase 2; Inclusion Bodies; Nerve Degeneration; Nerve Tissue Proteins; Neurons; Parkinson Disease; Phosphorylation; Point Mutation; Retina; Serine; Synucleins

2005
The pathology of the substantia nigra in Alzheimer disease with extrapyramidal signs.
    Neurology, 2005, Apr-26, Volume: 64, Issue:8

    Extrapyramidal signs (EPS) are common in Alzheimer disease (AD) and increase in prevalence as AD advances. The neuropathologic substrate responsible for EPS in AD remains to be fully characterized.. Subjects had a clinical diagnosis of AD confirmed by neuropathologic examination. EPS during life were documented by clinical methods assessing bradykinesia, cogwheel rigidity, rest tremor, and parkinsonian gait. Subjects with EPS and previous neuroleptic exposure were excluded. Twenty-eight subjects were in the EPS group and 104 subjects were without EPS. Neuron loss, alpha-synuclein (ASYN)-labeled pathology, and tau-labeled pathology in the substantia nigra were measured using semiquantitative techniques such that higher scores represented increased pathologic burden.. Presence of nigral ASYN-labeled pathology was more common (50 vs 28.9%; p < 0.05) in the EPS group than in those without EPS. There was more nigral neuron loss in the EPS group (1.50 vs 1.11 in no-EPS group; p < 0.05). Tau-labeled burden was not different by group comparisons; however, EPS onset at later stages of dementia severity was associated with increased tau-labeled pathology (Kendall tau-B = 0.48, p < 0.01) and this association remained after controlling for dementia severity at death. Additionally, moderate to severe tau burden was more common in the subgroup with "pure AD" (definite AD without other neuropathology) with EPS (81.8%) than cases without EPS (49.0%; p < 0.05). Four subjects with EPS (14.3%) had little to no significant nigral pathologic changes.. Clinically detected extrapyramidal signs (EPS) in Alzheimer disease (AD) are associated with substantia nigra pathology including alpha-synuclein aggregation, hyperphosphorylated tau accumulation, and neuron loss that may account for the increasing prevalence of EPS as AD progresses. In some cases, limited nigral pathology suggests extranigral factors in the clinical symptoms of EPS.

    Topics: Aged; Aged, 80 and over; alpha-Synuclein; Alzheimer Disease; Autopsy; Female; Humans; Lewy Bodies; Longitudinal Studies; Male; Middle Aged; Nerve Degeneration; Neurofibrillary Tangles; Neurons; Neuropsychological Tests; Parkinsonian Disorders; Substantia Nigra; tau Proteins

2005
Rotenone induces aggregation of gamma-tubulin protein and subsequent disorganization of the centrosome: relevance to formation of inclusion bodies and neurodegeneration.
    Neuroscience, 2005, Volume: 133, Issue:1

    Neurodegenerative disorders are characterized by progressive loss of specific neurons in the central nervous system. Although they have different etiologies and clinical manifestations, most of them share similar histopathologic characteristics such as the presence of inclusion bodies in both neurons and glial cells, which represent intracellular aggregation of misfolded or aberrant proteins. In Parkinson's disease, formation of inclusion bodies has been associated with the aggresome-related process and consequently with the centrosome. However, the significance of the centrosome in the neurodegenerative process remains obscure. In the present study, the morphological and functional changes in the centrosome induced by rotenone, a common insecticide used to produce experimental Parkinsonism, were examined both in vitro and in vivo. Aggregation of gamma-tubulin protein, which is a component of the centrosome matrix and recently identified in Lewy bodies of Parkinson's disease, was observed in primary cultures of mesencephalic cells treated with rotenone. Rotenone-treated neurons and astrocytes showed enlarged and multiple centrosomes. These centrosomes also displayed multiple aggregates of alpha-synuclein protein. Neurons with disorganized centrosomes exhibited neurite retraction and microtubule destabilization, and astrocytes showed disturbances of mitotic spindles. The Golgi apparatus, which is closely related to the centrosome, was dispersed in both rotenone-treated neuronal cells and the substantia nigra of rotenone-treated rats. Our findings suggested that recruitment of abnormal proteins in the centrosome contributed to the formation of inclusion bodies, and that rotenone markedly affected the structure and function of the centrosome with consequent induction of cytoskeleton disturbances, disassembly of the Golgi apparatus and collapse of neuronal cells.

    Topics: alpha-Synuclein; Animals; Astrocytes; Blotting, Western; Cell Death; Cells, Cultured; Centrosome; Dopamine; Female; Golgi Apparatus; Hypokinesia; Immunohistochemistry; Inclusion Bodies; Male; Mesencephalon; Nerve Degeneration; Nerve Tissue Proteins; Neurites; Pregnancy; Rats; Rats, Inbred Lew; Rats, Sprague-Dawley; Rotenone; Spindle Apparatus; Synucleins; Tubulin; Tyrosine 3-Monooxygenase; Uncoupling Agents

2005
Cleavage of alpha-synuclein by calpain: potential role in degradation of fibrillized and nitrated species of alpha-synuclein.
    Biochemistry, 2005, May-31, Volume: 44, Issue:21

    Alpha-synuclein (alpha-syn) is a major protein component of the neuropathological hallmarks of Parkinson's disease and related neurodegenerative disorders termed synucleinopathies. Neither the mechanism of alpha-syn fibrillization nor the degradative process for alpha-syn has been elucidated. Previously, we showed that wild-type, mutated, and fibrillar alpha-syn proteins are substrates of calpain I in vitro. In this study, we demonstrate that calpain-mediated cleavage near and within the middle region of soluble alpha-syn with/without tyrosine nitration and oxidation generates fragments that are unable to self-fibrillize. More importantly, these fragments prevent full-length alpha-syn from fibrillizing. Calpain-mediated cleavage of alpha-syn fibrils composed of wild-type or nitrated alpha-syn generate C-terminally truncated fragments that retain their fibrillar structure and induce soluble full-length alpha-syn to co-assemble. Therefore, calpain-cleaved soluble alpha-syn inhibits fibrillization, whereas calpain-cleaved fibrillar alpha-syn promotes further co-assembly. These results provide insight into possible disease mechanisms underlying synucleinopathies since the formation of alpha-syn fibrils could be causally linked to the onset/progression of these disorders.

    Topics: alpha-Synuclein; Calpain; Chymotrypsin; Humans; Hydrolysis; Microscopy, Immunoelectron; Nerve Degeneration; Nerve Tissue Proteins; Nitrates; Parkinson Disease; Peptide Fragments; Peroxynitrous Acid; Recombinant Proteins; Solubility; Synucleins; Tyrosine

2005
DJ-1 is present in a large molecular complex in human brain tissue and interacts with alpha-synuclein.
    Journal of neurochemistry, 2005, Volume: 93, Issue:6

    DJ-1 is a ubiquitously expressed protein involved in various cellular processes including cell proliferation, RNA-binding, and oxidative stress. Mutations that result in loss of DJ-1 function lead to early onset parkinsonism in humans, and DJ-1 protein is present in pathological lesions of several tauopathies and synucleinopathies. In order to further investigate the role of DJ-1 in human neurodegenerative disease, we have generated novel polyclonal and monoclonal antibodies to human DJ-1 protein. We have characterized these antibodies and confirmed the pathological co-localization of DJ-1 with other neurodegenerative disease-associated proteins, as well as the decrease in DJ-1 solubility in disease tissue. In addition, we report the presence of DJ-1 in a large molecular complex (> 2000 kDa), and provide evidence for an interaction between endogenous DJ-1 and alpha-synuclein in normal and diseased tissue. These findings provide new avenues towards the study of DJ-1 function and how loss of its activity may lead to parkinsonism. Furthermore, our results provide further evidence for the interplay between neurodegenerative disease-associated proteins.

    Topics: alpha-Synuclein; Animals; Antibodies; Antibody Specificity; Brain; Drosophila; Humans; Inclusion Bodies; Intracellular Signaling Peptides and Proteins; Macromolecular Substances; Mice; Molecular Sequence Data; Molecular Weight; Nerve Degeneration; Nerve Tissue Proteins; Oncogene Proteins; Parkinson Disease; Protein Deglycase DJ-1; Sequence Homology, Amino Acid; Solubility; Synucleins; tau Proteins

2005
Alpha-synuclein expression in the substantia nigra of MPTP-lesioned non-human primates.
    Neurobiology of disease, 2005, Volume: 20, Issue:3

    Changes in the expression of alpha-synuclein are likely to underlie its normal function as well as its role in pathological processes. The relationship between toxic injury and alpha-synuclein expression was assessed in the substantia nigra of squirrel monkeys treated with a single injection of MPTP and sacrificed 1 week or 1 month later. At 1 week, when stereological cell counting revealed only a small decrease (-10%) in the number of dopaminergic neurons, alpha-synuclein mRNA and protein were markedly enhanced. Increased alpha-synuclein immunoreactivity was evident at the level of neuronal fibers whereas nigral cell bodies were devoid of detectable protein. At 1 month post-MPTP, neuronal loss rose to 40%. Both alpha-synuclein mRNA and protein remained elevated but, noticeably, a robust alpha-synuclein immunoreactivity characterized a significant number of cell bodies. Neuromelanin granules are hallmarks of dopaminergic neurons in primates. Therefore, the number of alpha-synuclein-positive cells that also contained neuromelanin was counted throughout the substantia nigra. At 1 month, the vast majority of alpha-synuclein-immunoreactive neurons contained neuromelanin, and approximately 80% of the dopaminergic cell bodies that survived MPTP toxicity stained positive for alpha-synuclein. The results indicate that a single toxic insult is capable of inducing a sustained alpha-synuclein up-regulation in the primate brain. They support a direct relationship between neuronal injury and enhanced alpha-synuclein expression, and suggest that protein elevation within cell bodies may be a late feature of neurons that have endured a toxic stress.

    Topics: alpha-Synuclein; Animals; Cell Count; Cell Death; Disease Models, Animal; Dopamine; Female; Melanins; Nerve Degeneration; Neurons; Oxidative Stress; Parkinsonian Disorders; RNA, Messenger; Saimiri; Substantia Nigra; Up-Regulation

2005
Inhibition of alpha-synuclein fibrillization by dopamine analogs via reaction with the amino groups of alpha-synuclein. Implication for dopaminergic neurodegeneration.
    The FEBS journal, 2005, Volume: 272, Issue:14

    Fibrillization of alpha-synuclein (alpha-Syn) is closely associated with the formation of Lewy bodies in neurons and dopamine (DA) is a potent inhibitor for the process, which is implicated in the causative pathogenesis of Parkinson's disease (PD). To elucidate any molecular mechanism that may have biological relevance, we tested the inhibitory abilities of DA and several analogs including chemically synthetic and natural polyphenols in vitro. The MS and NMR characterizations strongly demonstrate that DA and its analogs inhibit alpha-Syn fibrillization by a mechanism where the oxidation products (quinones) of DA analogs react with the amino groups of alpha-Syn chain, generating alpha-Syn-quinone adducts. It is likely that the amino groups of alpha-Syn undergo nucleophilic attack on the quinone moiety of DA analogs to form imino bonds. The covalently cross-linked alpha-Syn adducts by DA are primarily large molecular mass oligomers, while those by catechol and p-benzoquinone (or hydroquinone) are largely monomers or dimers. The DA quinoprotein retains the same cytotoxicity as the intact alpha-Syn, suggesting that the oligomeric intermediates are the major elements that are toxic to the neuronal cells. This finding implies that the reaction of alpha-Syn with DA is relevant to the selective dopaminergic loss in PD.

    Topics: alpha-Synuclein; Cross-Linking Reagents; Dopamine; Flavonoids; Molecular Structure; Nerve Degeneration; Nerve Tissue Proteins; Oxidation-Reduction; Phenols; Polyphenols; Protein Conformation; Quinones; Spectrometry, Mass, Electrospray Ionization; Synucleins

2005
A comparison of degeneration in motor thalamus and cortex between progressive supranuclear palsy and Parkinson's disease.
    Brain : a journal of neurology, 2005, Volume: 128, Issue:Pt 10

    Changes in motor cortical activation are associated with the major symptoms observed in both Parkinson's disease and progressive supranuclear palsy (PSP). While research has concentrated on basal ganglia abnormalities as central to these cortical changes, several studies in both disorders have shown pathology in the thalamus and motor cortices. In particular, we recently reported an 88% loss of corticocortical projection neurones in the pre-supplementary motor (pre-SMA) cortex in Parkinson's disease. Further analysis of the degree of neuronal loss and pathology in motor cortices and their thalamocortical relays in Parkinson's disease and PSP is warranted. Six cases with PSP, nine cases with Parkinson's disease and nine controls were selected from a prospectively studied brain donor cohort. alpha-Synuclein, ubiquitin and tau immunohistochemistry were used to identify pathological lesions. Unbiased stereological methods were used to analyse atrophy and neuronal loss in the motor thalamus [ventral anterior, ventrolateral anterior and ventrolateral posterior (VLp) nuclei] and motor cortices (primary motor, dorsolateral premotor and pre-SMA cortices). Analysis of variance and post hoc testing was used to determine differences between groups. In Parkinson's disease, the motor thalamus and motor cortices (apart from the pre-SMA) were preserved containing only rare alpha-synuclein-positive and ubiquitin-positive Lewy bodies. In contrast, patients with PSP had significant atrophy and neuronal loss in VLp (22 and 30%, respectively), pre-SMA (21 and 51%, respectively) and primary motor cortices (33 and 54%, respectively). In the primary motor cortex of PSP cases, neuronal loss was confined to inhibitory interneurones, whereas in the pre-SMA both interneurones (reduced by 26%) and corticocortical projection neurones (reduced by 82%) were affected. Tau-positive neurofibrillary and glial tangles were observed throughout the motor thalamus and motor cortices in PSP. These non-dopaminergic lesions in motor circuits are likely to contribute to the pathogenesis of both PSP and Parkinson's disease. The selective involvement of the VLp and primary motor cortex in PSP implicates these cerebellothalamocortical pathways as differentiating this disease, possibly contributing to the early falls.

    Topics: Aged; alpha-Synuclein; Factor Analysis, Statistical; Female; Humans; Immunohistochemistry; Interneurons; Lewy Bodies; Male; Motor Cortex; Motor Neurons; Nerve Degeneration; Neurons; Parkinson Disease; Prospective Studies; Supranuclear Palsy, Progressive; tau Proteins; Thalamus; Ubiquitin; Ventral Thalamic Nuclei

2005
Grading of neuropathology in multiple system atrophy: proposal for a novel scale.
    Movement disorders : official journal of the Movement Disorder Society, 2005, Volume: 20 Suppl 12

    Multiple system atrophy (MSA), a sporadic progressive synucleinopathy of advanced age, is separated into two clinic opathological subtypes: MSA-P (striatonigral degeneration [SND]) with predominant parkinsonian features and MSA-C (olivopontocerebellar atrophy [OPCA]) with predominant cerebellar ataxia. We propose a novel morphological grading system for both subtypes to compare lesion intensities and their possible clinical validity. Forty-two autopsy cases of MSA were separated into four grades (SND 0-III and OPCA 0-III) based on semiquantitative assessment of neuronal loss, astrogliosis, and presence of alpha-synuclein-positive glial cytoplasmic inclusions (GCI) in striatum, globus pallidus, substantia nigra, pontine basis, cerebellum, and inferior olives. Whereas a recent grading system restricted to SND reflected disease progression and dopa-responsiveness, there was considerable variation in the morphological combination between SND and OPCA, with only around half the cases with OPCA II (moderate) and III (severe) showing comparable grades of both types, whereas OPCA 0 and I (no or little degeneration) was combined with all grades of SND. Twenty-two cases showing OPCA 0 + SND II (n = 3), OPCA I + SND I-II (n = 11), and OPCA I + SND III (n = 8) were classified as pure or predominant SND, consistent with MSA-P. Twenty cases showing OPCA II + SND II/III (n = 7) and OPCA III + SND III (n = 13) were classified as predominant OPCA, consistent with MSA-C. In MSA-P, the mean age of onset was higher than it was in MSA-C (55.1 vs. 50.5 years), but the mean duration of illness was shorter in MSA-P (5.3 vs. 6.7 years). Presenting symptoms in MSA-P were mainly parkinsonism, whereas in MSA-C they were mainly gait disorders (14 vs. 1; P < 0.001). Among clinical key symptoms, parkinsonism was more frequent than were cerebellar signs in MSA-P; in MSA-C it was the reverse (P < 0.01), whereas other symptoms (autonomic/urinary failure) showed no differences. Parkinsonism was infrequent in MSA-C even when OPCA was associated with SND, suggesting a masking effect by cerebellar system involvement. High terminal Hoehn and Yahr stages were more frequent in MSA-P (P < 0.01), some with good-to-moderate initial levodopa (L-dopa) response. Although the proposed morphological grading of both MSA-P and -C correlates well with initial symptoms and clinical key features of both types, further prospective studies are required to validate the clinical utility of the proposed MSA gradi

    Topics: alpha-Synuclein; Cerebellar Ataxia; Corpus Striatum; Diagnosis, Differential; Disease Progression; Humans; Middle Aged; Multiple System Atrophy; Nerve Degeneration; Olivopontocerebellar Atrophies; Severity of Illness Index

2005
In vitro models of multiple system atrophy.
    Movement disorders : official journal of the Movement Disorder Society, 2005, Volume: 20 Suppl 12

    alpha-Synuclein represents the major constituent of oligodendroglial cytoplasmic inclusions, the hallmark lesion of multiple system atrophy (MSA), a progressive disorder that is associated with selective degenerative cell loss in basal ganglia, cerebellum, brainstem, and spinal cord. The role of abnormal alpha-synuclein aggregation in oligodendroglial cells is still obscure, in particular, whether alpha-synuclein might impair oligodendroglial and, secondarily, neuronal integrity of those cells in the diseased brain. In an attempt to answer some of these questions, we have developed an "in vitro model of MSA" by expressing the wild-type or C-terminally truncated form of alpha-synuclein in glial cell cultures. With this simplified system, we have demonstrated that alpha-synuclein significantly affects the survival of glia and its vulnerability to environmental stress, which might represent a major step in the pathogenesis of MSA.

    Topics: alpha-Synuclein; Basal Ganglia; Brain Stem; Cerebellum; Disease Progression; Environment; Humans; In Vitro Techniques; Microglia; Multiple System Atrophy; Nerve Degeneration; Oxidative Stress; Spinal Cord; Tumor Necrosis Factor-alpha

2005
Tau gene transfer, but not alpha-synuclein, induces both progressive dopamine neuron degeneration and rotational behavior in the rat.
    Neurobiology of disease, 2005, Volume: 20, Issue:1

    Using a viral vector for mutant (P301L) tau, we studied the effects of gene transfer to the rat substantia nigra in terms of structural and functional properties of dopaminergic neurons. The mutant tau vector caused progressive loss of pars compacta dopaminergic neurons over time, reduced striatal dopamine content, and amphetamine-stimulated rotational behavior consistent with a specific lesion effect. In addition, structural studies demonstrated neurofibrillary tangles and neuritic pathology. Wild-type tau had similar effects on neuronal loss and rotational behavior. In contrast, mutant alpha-synuclein vectors did not induce rotational behavior, although alpha-synuclein filaments formed in nigrostriatal axons. Dopamine neuron function is affected by tau gene transfer and appears to be more susceptible to tau- rather than alpha-synuclein-related damage in this model. Both tau and alpha-synuclein are important for substantia nigra neurodegeneration models in rats, further indicating their potential as therapeutic targets for human diseases involving loss of dopamine neurons.

    Topics: alpha-Synuclein; Animals; Behavior, Animal; Disease Models, Animal; Dopamine; Gene Transfer Techniques; Genetic Vectors; Male; Motor Activity; Movement Disorders; Mutation; Nerve Degeneration; Neurofibrillary Tangles; Rats; Rats, Sprague-Dawley; Rotation; Substantia Nigra; tau Proteins; Tauopathies

2005
Alpha-synuclein cooperates with CSPalpha in preventing neurodegeneration.
    Cell, 2005, Nov-04, Volume: 123, Issue:3

    Alpha-synuclein and cysteine-string protein-alpha (CSPalpha) are abundant synaptic vesicle proteins independently linked to neurodegeneration. Dominantly inherited mutations in alpha-synuclein cause Parkinson's disease, but the physiological role of alpha-synuclein remains unknown. Deletion of CSPalpha produces rapidly progressive neurodegeneration in mice, presumably because the cochaperone function of CSPalpha is essential for neuronal survival. Here, we report the surprising finding that transgenic expression of alpha-synuclein abolishes the lethality and neurodegeneration caused by deletion of CSPalpha. Conversely, ablation of endogenous synucleins exacerbates these phenotypes. Deletion of CSPalpha inhibits SNARE complex assembly; transgenic alpha-synuclein ameliorates this inhibition. In preventing neurodegeneration in CSPalpha-deficient mice, alpha-synuclein does not simply substitute for CSPalpha but acts by a downstream mechanism that requires phospholipid binding by alpha-synuclein. These observations reveal a powerful in vivo activity of alpha-synuclein in protecting nerve terminals against injury and suggest that this activity operates in conjunction with CSPalpha and SNARE proteins on the presynaptic membrane interface.

    Topics: alpha-Synuclein; Animals; beta-Synuclein; Brain; HSP40 Heat-Shock Proteins; Mice; Mice, Knockout; Nerve Degeneration; Phospholipids; SNARE Proteins; Spinal Cord; Synaptic Membranes

2005
Neurological and neurodegenerative alterations in a transgenic mouse model expressing human alpha-synuclein under oligodendrocyte promoter: implications for multiple system atrophy.
    The Journal of neuroscience : the official journal of the Society for Neuroscience, 2005, Nov-16, Volume: 25, Issue:46

    Multiple system atrophy (MSA) is a progressive, neurodegenerative disease characterized by parkinsonism, ataxia, autonomic dysfunction, and accumulation of alpha-synuclein (alpha-syn) in oligodendrocytes. To better understand the mechanisms of neurodegeneration and the role of alpha-syn accumulation in oligodendrocytes in the pathogenesis of MSA, we generated transgenic mouse lines expressing human (h) alpha-syn under the control of the murine myelin basic protein promoter. Transgenic mice expressing high levels of halpha-syn displayed severe neurological alterations and died prematurely at 6 months of age. Furthermore, mice developed progressive accumulation of halpha-syn-immunoreactive inclusions in oligodendrocytes along the axonal tracts in the brainstem, basal ganglia, cerebellum, corpus callosum, and neocortex. The inclusions also reacted with antibodies against phospho-serine (129) halpha-syn and ubiquitin, and halpha-syn was found in the detergent-insoluble fraction. In high-expresser lines, the white matter tracts displayed intense astrogliosis, myelin pallor, and decreased neurofilament immunostaining. Accumulation of halpha-syn in oligodendrocytes also leads to prominent neurodegenerative changes in the neocortex with decreased dendritic density and to loss of dopaminergic fibers in the basal ganglia. The oligodendrocytic inclusions were composed of fibrils and accompanied by mitochondrial alterations and disruption of the myelin lamina in the axons. Together, these studies support the contention that accumulation of alpha-syn in oligodendrocytes promotes neurodegeneration and recapitulates several of the key functional and neuropathological features of MSA.

    Topics: alpha-Synuclein; Animals; Disease Models, Animal; Humans; Male; Mice; Mice, Inbred C57BL; Mice, Inbred DBA; Mice, Transgenic; Middle Aged; Multiple System Atrophy; Nerve Degeneration; Oligodendroglia; Promoter Regions, Genetic

2005
alpha-Synuclein selectively increases manganese-induced viability loss in SK-N-MC neuroblastoma cells expressing the human dopamine transporter.
    Neuroscience letters, 2004, Jan-02, Volume: 354, Issue:1

    The established or potentially toxic agents implicated in the nigral cell death in Parkinson's disease, dopamine, 1-methyl-4-phenylpyridinium (MPP(+)), iron, and manganese, were examined as to their effects on the viability of cells overexpressing alpha-synuclein. SK-N-MC neuroblastoma cells stably expressing the human dopamine transporter were transfected with human alpha-synuclein and cell clones with and without alpha-synuclein immunoreactivity were obtained. Cells were exposed for 24-72 h to 1-10 microM dopamine, 0.1-3 microM MPP(+), 0.1-1 mM FeCl(2) or 30-300 microM MnCl(2) added to the culture medium. There was no difference between cells expressing alpha-synuclein and control cells after exposure to dopamine, MPP(+) or FeCl(2). However, MnCl(2) resulted in a significantly stronger decreased viability of cells overexpressing alpha-synuclein after 72 h. These findings suggest that manganese may co-operate with alpha-synuclein in triggering neuronal cell death such as seen in manganese parkinsonism. The relevance of our observations for the pathoetiology of Parkinson's disease proper remains to be determined.

    Topics: 1-Methyl-4-phenylpyridinium; alpha-Synuclein; Cell Death; Cell Line, Tumor; Cell Survival; Chlorides; Dopamine; Dopamine Plasma Membrane Transport Proteins; Ferrous Compounds; Gene Expression; Herbicides; Humans; Manganese Compounds; Membrane Glycoproteins; Membrane Transport Proteins; Nerve Degeneration; Nerve Tissue Proteins; Neuroblastoma; Synucleins; Transfection

2004
Extrapyramidal features in patients with motor neuron disease and dementia; a clinicopathological correlative study.
    Acta neuropathologica, 2004, Volume: 107, Issue:4

    Motor neuron disease (MND) may be complicated by frontotemporal dementia and/or an extrapyramidal movement disorder. Several studies have identified the pathological substrate for dementia in MND as being ubiquitin-immunoreactive inclusions and dystrophic neurites in the extramotor neocortex and hippocampus. Although degenerative changes have previously been noted in the basal ganglia and substantia nigra in MND, detailed pathological studies with clinical correlation are lacking. We examined postmortem material from eight patients with a history of MND and dementia, four of whom also had prominent extrapyramidal features. All cases showed the expected degenerative changes in the pyramidal motor system and ubiquitin-positive inclusions in the extramotor cortex. In addition, the cases with a history of extrapyramidal features had striking pathology in the basal ganglia and substantia nigra; neuronal loss and gliosis ranged from moderate to severe and immunohistochemistry demonstrated numerous neuronal inclusions and dystrophic neurites, which were reactive for ubiquitin, but not tau or alpha-synuclein. Similar pathology was either absent or much milder in cases without extrapyramidal features. This study illustrates the utility of ubiquitin immunohistochemistry in demonstrating the range of pathology in MND and provides a neuropathological correlate for the extrapyramidal features which may occur in MND with dementia.

    Topics: alpha-Synuclein; Basal Ganglia Diseases; Corpus Striatum; Dementia; Glial Fibrillary Acidic Protein; Hippocampus; Humans; Immunohistochemistry; Inclusion Bodies; Motor Neuron Disease; Nerve Degeneration; Nerve Tissue Proteins; Neuroglia; Neurons; Postmortem Changes; Substantia Nigra; Synucleins; tau Proteins; Ubiquitin

2004
The new mutation, E46K, of alpha-synuclein causes Parkinson and Lewy body dementia.
    Annals of neurology, 2004, Volume: 55, Issue:2

    Familial parkinsonism and dementia with cortical and subcortical Lewy bodies is uncommon, and no genetic defect has been reported in the previously described sibships. We present a Spanish family with autosomal dominant parkinsonism, dementia, and visual hallucinations of variable severity. The postmortem examination showed atrophy of the substantia nigra, lack of Alzheimer pathology, and numerous Lewy bodies which were immunoreactive to alpha-synuclein and ubiquitin in cortical and subcortical areas. Sequencing of the alpha-synuclein gene showed a novel, nonconservative E46K mutation in heterozygosis. The E46K mutation was present in all affected family members and in three young asymptomatic subjects, but it was absent in healthy and pathological controls. The novel mutation, that substitutes a dicarboxylic amino acid, glutamic acid, with a basic amino acid such as lysine in a much conserved area of the protein, is likely to produce severe disturbance of protein function. Our data show that, in addition to the previously described hereditary alpha-synucleinopathies, dementia with Lewy bodies is related to mutation of alpha-synuclein.

    Topics: Aged; alpha-Synuclein; Amino Acid Sequence; Base Sequence; Brain; DNA Mutational Analysis; DNA Primers; Female; Humans; Immunohistochemistry; Lewy Bodies; Lewy Body Disease; Male; Middle Aged; Molecular Sequence Data; Mutation; Nerve Degeneration; Nerve Tissue Proteins; Parkinson Disease; Pedigree; Polymerase Chain Reaction; Sequence Homology; Synucleins; Tomography, Emission-Computed, Single-Photon

2004
Comparison of kindreds with parkinsonism and alpha-synuclein genomic multiplications.
    Annals of neurology, 2004, Volume: 55, Issue:2

    Genomic triplication of the alpha-synuclein gene recently has been associated with familial Parkinson's disease in the Spellman-Muenter kindred. Here, we present an independent family, of Swedish-American descent, with hereditary early-onset parkinsonism with dementia due to alpha-synuclein triplication. Brain tissue available from affected individuals in both kindreds provided the opportunity to compare their clinical, pathological, and biochemical phenotypes. Of note, studies of brain mRNA and soluble protein levels demonstrate a doubling of alpha-synuclein expression, consistent with molecular genetic data. Pathologically, cornu ammonis 2/3 hippocampal neuronal loss appears to be a defining feature of this form of inherited parkinsonism. The profound implications of alpha-synuclein overexpression for idiopathic synucleinopathies are discussed.

    Topics: alpha-Synuclein; Brain; DNA Mutational Analysis; Electrophoresis, Polyacrylamide Gel; Gene Dosage; Gene Expression; Humans; Male; Middle Aged; Mutation; Nerve Degeneration; Nerve Tissue Proteins; Parkinsonian Disorders; Pedigree; Polymerase Chain Reaction; RNA, Messenger; Synucleins

2004
Enhanced substantia nigra mitochondrial pathology in human alpha-synuclein transgenic mice after treatment with MPTP.
    Experimental neurology, 2004, Volume: 186, Issue:2

    Recent studies have implicated alpha-synuclein (alpha-S) in the pathogenesis of Parkinson's disease (PD). The mechanisms underlying PD are not completely understood; however, mitochondrial complex I inhibition and oxidative injury may be involved. Because the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) is a potent complex I inhibitor that can cause oxidative injury and mimic many aspects of PD in treated animals, we sought to determine whether the overexpression of alpha-S in transgenic (tg) mice (alpha-S-tg) would enhance the substantia nigra (SN) pathology resulting from treatment with MPTP. For this purpose, alpha-S-tg mice were produced expressing high levels of wild-type (wt) human alpha-S under the control of the neuron-specific Thy-1 promoter. Alpha-S-tg mice and non-tg controls were treated with MPTP (15 mg/kg ip, twice a week for 2 weeks) or saline (Sal) and then examined 2 weeks after completion of treatment by transmission electron microscopy (EM). We found that alpha-S-tg mice treated with MPTP had extensive mitochondrial alterations, increases in mitochondrial size, filamentous neuritic aggregations, axonal degeneration, and formation of electron dense perinuclear cytoplasmic inclusions in the SN that did not occur in the hippocampus or neocortex, nor in MPTP-treated non-tg mice or Sal-treated alpha-S-tg mice. These findings support the potential involvement of alpha-S expression in the vulnerability of SN neurons to toxicity from mitochondrial complex I inhibitors and the subsequent development of neurodegenerative pathology.

    Topics: 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine; alpha-Synuclein; Animals; Dopamine Agents; Dose-Response Relationship, Drug; Humans; Lewy Bodies; Mice; Mice, Inbred C57BL; Mice, Transgenic; Microscopy, Electron; Mitochondria; Nerve Degeneration; Nerve Tissue Proteins; Reverse Transcriptase Polymerase Chain Reaction; Substantia Nigra; Synucleins; Thy-1 Antigens

2004
Lentiviral nigral delivery of GDNF does not prevent neurodegeneration in a genetic rat model of Parkinson's disease.
    Neurobiology of disease, 2004, Volume: 17, Issue:2

    Viral delivery of glial cell line-derived neurotrophic factor (GDNF) currently represents one of the most promising neuroprotective strategies for Parkinson's Disease (PD). However, the effect of this neurotrophic factor has never been tested in the newly available genetic models of PD based on the viral expression of mutated alpha-synuclein. In this study, we evaluated the ability of lentiviral vectors coding for GDNF (lenti-GDNF) to prevent nigral dopaminergic degeneration associated with the lentiviral mediated expression of the A30P mutant human alpha-synuclein (lenti-A30P). This virally based rat model develops a progressive and selective loss of dopamine neurons associated with the appearance of alpha-synuclein containing inclusions, thus recapitulating the major hallmarks of PD. Lenti-GDNF was injected in the substantia nigra 2 weeks before nigral administration of lenti-A30P. Although a robust expression of GDNF was observed in the whole nigrostriatal pathway due to retrograde and/or anterograde transport, lenti-GDNF did not prevent the alpha-synuclein-induced dopaminergic neurodegeneration in the lentiviral-based genetic rat model of PD. These results suggest that sustained GDNF treatment cannot modulate the cellular toxicity related to abnormal folded protein accumulation as mutated human alpha-synuclein.

    Topics: alpha-Synuclein; Animals; Female; Gene Transfer Techniques; Genetic Vectors; Glial Cell Line-Derived Neurotrophic Factor; Humans; Immunohistochemistry; Lentivirus; Nerve Degeneration; Nerve Growth Factors; Nerve Tissue Proteins; Neuroprotective Agents; Parkinson Disease; Rats; Rats, Wistar; Synucleins

2004
Rotenone potentiates dopamine neuron loss in animals exposed to lipopolysaccharide prenatally.
    Experimental neurology, 2004, Volume: 190, Issue:2

    We previously demonstrated that treating gravid female rats with the bacteriotoxin lipopolysaccharide (LPS) led to the birth of offspring with fewer than normal dopamine (DA) neurons. This DA neuron loss was long-lived and associated with permanent increases in the pro-inflammatory cytokine tumor necrosis factor alpha (TNFalpha). Because of this pro-inflammatory state, we hypothesized that these animals would be more susceptible to subsequent exposure of DA neurotoxins. We tested this hypothesis by treating female Sprague-Dawley rats exposed to LPS or saline prenatally with a subtoxic dose of the DA neurotoxin rotenone (1.25 mg/kg per day) or vehicle for 14 days when they were 16 months old. After another 14 days, the animals were sacrificed. Tyrosine hydroxylase-immunoreactive (THir) cell counts were used as an index of DA neuron survival. Animals exposed to LPS prenatally or rotenone postnatally exhibited a 22% and 3%, respectively, decrease in THir cell counts relative to controls. The combined effects of prenatal LPS and postnatal rotenone exposure produced a synergistic 39% THir cell loss relative to controls. This loss was associated with decreased striatal DA and increased striatal DA activity ([HVA]/[DA]) and TNFalpha. Animals exposed to LPS prenatally exhibited a marked increase in the number of reactive microglia that was further increased by rotenone exposure. Prenatal LPS exposure also led to increased levels of oxidized proteins and the formation of alpha-Synuclein and eosin positive inclusions resembling Lewy bodies. These results suggest that exposure to low doses of an environmental neurotoxin like rotenone can produce synergistic DA neuron losses in animals with a preexisting pro-inflammatory state. This supports the notion that Parkinson's disease (PD) may be caused by multiple factors and the result of "multiple hits" from environmental toxins.

    Topics: alpha-Synuclein; Animals; Brain; Dopamine; Drug Synergism; Enzyme-Linked Immunosorbent Assay; Female; Immunohistochemistry; Insecticides; Lewy Bodies; Lipopolysaccharides; Microglia; Nerve Degeneration; Nerve Tissue Proteins; Neurons; Pregnancy; Prenatal Exposure Delayed Effects; Rats; Rats, Sprague-Dawley; Rotenone; Synucleins; Tyrosine 3-Monooxygenase

2004
Limbic neuropathology in idiopathic Parkinson's disease with concomitant dementia.
    Folia neuropathologica, 2004, Volume: 42, Issue:3

    To study pathological background of dementia in idiopathic Parkinson's disease (PD), 41 autopsy brains (31 cases with and 10 cases without dementia) were investigated. The severity of degenerative changes was evaluated in selected limbic regions (trans- and entorhinal cortex, hippocampus, and amygdala). The densities of Lewy bodies (LBs), Lewy neurites (LNs), neurofibrillary tangles (NFTs), and amyloid neuritic plaques (NPs) were determined on immunohistochemically stained sections using antibodies against alpha-synuclein, tau-protein, and amyloid-beta. Precisely defined modern criteria for selecting study cohort (Newcastle, CERAD and Braak et al.) ensured homogeneity of the study sample and reliability of the results. Comparisons between the cases of Parkinson's disease with dementia (PDD) and those without (PD-only) revealed that the former were characterised by significantly higher densities of LBs and LNs in transentorhinal and entorhinal cortices as well as in the CA2-3 region of the hippocampus and cortical complex of amygdala. In the PDD sub-set we found statistically significant correlation of LBs with LNs counts in CA2-3 region of hippocampus as well as of LBs counts in transentorhinal cortex with LNs counts in CA2-3 hippocampal region. The relationship was also observed between LBs counts in CA2-3 region of the hippocampus and LNs counts in cortical complex of amygdala. Our studies suggest that dementia in PD may be associated with the presence of degenerative changes of PD-type in leading limbic structures, without co-existent Alzheimer's disease (AD). They also imply that LBs and LNs may appear to be morphological hallmarks of the pathological process associated with dementia in PD. LBs and LNs distribution pattern and correlations of LBs with LNs counts in limbic regions observed in our study suggest the cumulative patomechanism of changes dependent on transsynaptic alpha-syn pathology and indicate the spread of the pathological process via axonal transport. The coexistence of the small number of changes of AD-type may exacerbate cognitive deficits in PDD.

    Topics: Aged; Aged, 80 and over; alpha-Synuclein; Amyloid beta-Peptides; Dementia; Female; Humans; Immunohistochemistry; Lewy Bodies; Limbic System; Male; Middle Aged; Nerve Degeneration; Nerve Tissue Proteins; Neurofibrillary Tangles; Parkinson Disease; Plaque, Amyloid; Synucleins; tau Proteins

2004
The mouse MPTP model: gene expression changes in dopaminergic neurons.
    The European journal of neuroscience, 2003, Volume: 17, Issue:1

    Parkinson's disease (PD) is a common neurodegenerative disorder, characterized by the progressive loss of dopaminergic neurons in the substantia nigra. Although valuable animal models have been developed, our knowledge of the aetiology and pathogenic factors implicated in PD is still insufficient to develop causal therapeutic strategies aimed at halting its progression. The neurotoxicity induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) is one of the most valuable models for analysing pathological aspects of PD. In this paper we studied the gene expression patterns underlying the pathogenesis of MPTP-induced neurodegeneration. We treated young and old C57BL/6 mice with different schedules of MPTP to induce degenerative processes that vary in intensity and time-course. During the first week after intoxication we used nonradioactive in situ-hybridization to investigate the expression patterns of genes associated with (i) dopamine metabolism and signalling; (ii) familial forms of PD; (iii) protein folding and (iv) energy metabolism. MPTP injections induced different severities of neuronal injury depending on the age of the animals and the schedule of administration as well as a significant degeneration in the striatum. In situ hybridization showed that MPTP intoxication initiated a number of gene expression changes that (i) were restricted to the neurons of the substantia nigra pars compacta; (ii) were correlated in intensity and number of changes with the age of the animals and the severity of histopathological disturbances; (iii) displayed in each a significant down-regulation by the end of one week after the last MPTP injection, but (iv) varied within one MPTP regimen in expression levels during the observation period. The subacute injection of MPTP into one-year-old mice induced the most severe changes in gene expression. All genes investigated were affected. However, alpha-synuclein was the only gene that was exclusively up-regulated in MPTP-treated animals displaying cell death.

    Topics: alpha-Synuclein; Animals; Corpus Striatum; Disease Models, Animal; Dopamine; Dopamine Plasma Membrane Transport Proteins; Drug Administration Schedule; Gene Expression; Heat-Shock Proteins; Immunohistochemistry; In Situ Hybridization; In Situ Nick-End Labeling; Leukocyte Common Antigens; Ligases; Male; Membrane Glycoproteins; Membrane Transport Proteins; Mice; Mice, Inbred C57BL; MPTP Poisoning; NADH Dehydrogenase; Nerve Degeneration; Nerve Tissue Proteins; Neurons; Parkinson Disease, Secondary; Substantia Nigra; Synucleins; Time Factors; Tyrosine 3-Monooxygenase; Ubiquitin-Protein Ligases

2003
Immunoexpression of 14-3-3 proteins in glial cytoplasmic inclusions of multiple system atrophy.
    Acta neuropathologica, 2003, Volume: 106, Issue:1

    Glial cytoplasmic inclusions (GCIs) are the histological hallmark of multiple system atrophy (MSA). In six postmortem brains of patients with MSA, 14-3-3-protein immunoreactivity was identified in GCIs predominately in the white matter tissue of the basal forebrain and cerebellum. Using double immunohistochemistry, co-localization of 14-3-3-protein and alpha-synuclein immunoreactivities in the GCIs was confirmed. The immunolabeling rate of GCIs with 14-3-3 proteins varied regionally from approximately 40% to 90%. Semiquantitative analysis yielded a significant negative correlation between degree of tissue degeneration and density of 14-3-3-protein-immunoreactive GCIs. The 14-3-3 proteins are active cofactors involved in cellular regulation through binding to phosphorylated motifs in target proteins and alpha-synuclein is a known target of 14-3-3. Our study suggests that 14-3-3 proteins are closely associated with alpha-synuclein in GCIs and 14-3-3 proteins may be candidate cofactors of alpha-synuclein in GCI formation.

    Topics: 14-3-3 Proteins; Aged; Aged, 80 and over; alpha-Synuclein; Brain; Humans; Immunohistochemistry; Inclusion Bodies; Middle Aged; Multiple System Atrophy; Nerve Degeneration; Nerve Tissue Proteins; Neuroglia; Synucleins; Tyrosine 3-Monooxygenase

2003
Accumulation of phosphorylated alpha-synuclein in aging human brain.
    Journal of neuropathology and experimental neurology, 2003, Volume: 62, Issue:6

    Alpha-synuclein in Lewy bodies (LBs) is phosphorylated at Ser129. We raised monoclonal and polyclonal antibodies to this phosphorylation site (psyn) and examined 157 serial autopsy brains from a geriatric hospital. Anti-psyn immunoreactivity was observed in 40 of these cases (25.5%). Immunohistochemistry revealed 4 novel types of pathology: diffuse neuronal cytoplasmic staining (pre-LB); neuropil thread-like structures (Lewy threads); dot-like structures similar to argyrophilic grains (Lewy dots); and axons in the white matter (Lewy axons). This novel pathology was abundantly present around LBs and also involved the limbic subcortical white matter, the cerebral cortical molecular layer, and the spongiform changes of the medial temporal lobe associated with cases of dementia with LBs (DLB). The phosphorylated alpha-synuclein was limited to the temporal lobe in cases of Parkinson disease, spread from the temporal lobe to the frontal lobe in cases of DLB transitional form and further spread to the parietal and occipital lobes in DLB neocortical form. Our findings suggest that LB-related pathology initially involves the neuronal perikarya, dendrites, and axons, causes impairment of axonal transport and synaptic transmission, and later leads to the formation of LBs, a hallmark of functional disturbance long before neuronal cell death.

    Topics: Aged; Aged, 80 and over; Aging; alpha-Synuclein; Antibodies, Monoclonal; Axons; Blotting, Western; Brain; Dementia; Female; Fluorescent Dyes; Humans; Immunohistochemistry; Lewy Bodies; Male; Microtubule-Associated Proteins; Middle Aged; Nerve Degeneration; Nerve Tissue Proteins; Neurofilament Proteins; Neurons; Phosphorylation; Synucleins

2003
Gene expression changes presage neurodegeneration in a Drosophila model of Parkinson's disease.
    Human molecular genetics, 2003, Oct-01, Volume: 12, Issue:19

    Transgenic Drosophila expressing human alpha-synuclein faithfully replicate essential features of human Parkinson's disease, including age-dependent loss of dopaminergic neurons, Lewy-body-like inclusions and locomotor impairment. To define the transcriptional program encoding molecular machinery involved in alpha-synuclein pathology, we characterized expression of the entire Drosophila genome at pre-symptomatic, early and advanced disease stages. Fifty-one signature transcripts, including lipid, energy and membrane transport mRNAs, were tightly associated with alpha-synuclein expression. Most importantly, at the pre-symptomatic stage, when the potential for neuroprotection is greatest, expression changes revealed specific pathology. In age-matched tau transgenic Drosophila, the transcription of alpha-synuclein associated genes was normal, suggesting highly distinct pathways of neurodegeneration. Temporal profiling of progressive gene expression changes in neurodegenerative disease models provides unbiased starting points for defining disease mechanisms and for identifying potential targets for neuroprotective drugs at pre-clinical stages.

    Topics: alpha-Synuclein; Animals; Animals, Genetically Modified; Disease Models, Animal; Drosophila; Gene Expression; Gene Expression Profiling; Genome; Humans; Lewy Bodies; Mutation; Nerve Degeneration; Nerve Tissue Proteins; Parkinson Disease; Polymerase Chain Reaction; Synucleins; tau Proteins; Time Factors; Transcription, Genetic; Transgenes

2003
Degenerative axonal changes in the hippocampus and amygdala in Parkinson's disease.
    Folia neuropathologica, 2003, Volume: 41, Issue:4

    The morphological background of cognitive and emotional impairments in Parkinson's disease (PD) has not yet been fully explained. We evaluated the expression of synaptic proteins: alpha- and beta-synuclein, synaptophysin and synaptobrevin and ultrastructural changes of perikaryons and axons in limbic structures at post-mortem from cases of PD to estimate degenerative axonal pathology in the hippocampus and amygdala [corrected]. Limbic structures (enthorinal cortex, hippocampus, and amygdala) are essential for the cognitive processes and emotional behaviour. We found that presynaptic axon pathology is mostly connected with hippocampal CA2-3 and dentate hilar regions as well as with the cortical and medio-central complexes of amygdala. Heterogeneous immunoreactivity of alpha-synuclein and diversified ultrastructure of Lewy bodies (LBs) and Lewy neurites (LNs) indicate their consecutive developmental stages. We observed an excessive perineuroneal expression of synaptophysin in the dentate hilar region in all PD cases, except one. This suggests that the dysfunction of synapses in this region may result from axonal pathology. Our study indicates a relation between cognitive and behavioural symptomatology in PD and alpha-synuclein dependent axonal pathology in the hippocampus and amygdala.

    Topics: Aged; Aged, 80 and over; alpha-Synuclein; Amygdala; Axons; beta-Synuclein; Female; Hippocampus; Humans; Immunohistochemistry; Lewy Bodies; Male; Membrane Proteins; Microscopy, Electron; Middle Aged; Nerve Degeneration; Nerve Tissue Proteins; Neurofibrillary Tangles; Parkinson Disease; Plaque, Amyloid; R-SNARE Proteins; Synaptophysin; Synucleins

2003
alpha -Synucleinopathy and selective dopaminergic neuron loss in a rat lentiviral-based model of Parkinson's disease.
    Proceedings of the National Academy of Sciences of the United States of America, 2002, Aug-06, Volume: 99, Issue:16

    Parkinson's disease (PD) is characterized by the progressive loss of substantia nigra dopaminergic neurons and the presence of cytoplasmic inclusions named Lewy bodies. Two missense mutations of the alpha-synuclein (alpha-syn; A30P and A53T) have been described in several families with an autosomal dominant form of PD. alpha-Syn also constitutes one of the main components of Lewy bodies in sporadic cases of PD. To develop an animal model of PD, lentiviral vectors expressing different human or rat forms of alpha-syn were injected into the substantia nigra of rats. In contrast to transgenic mice models, a selective loss of nigral dopaminergic neurons associated with a dopaminergic denervation of the striatum was observed in animals expressing either wild-type or mutant forms of human alpha-syn. This neuronal degeneration correlates with the appearance of abundant alpha-syn-positive inclusions and extensive neuritic pathology detected with both alpha-syn and silver staining. Lentiviral-mediated expression of wild-type or mutated forms of human alpha-syn recapitulates the essential neuropathological features of PD. Rat alpha-syn similarly leads to protein aggregation but without cell loss, suggesting that inclusions are not the primary cause of cell degeneration in PD. Viral-mediated genetic models may contribute to elucidate the mechanism of alpha-syn-induced cell death and allow the screening of candidate therapeutic molecules.

    Topics: alpha-Synuclein; Animals; Animals, Genetically Modified; Biomarkers; Brain; Disease Models, Animal; Dopamine; Gene Expression; Genetic Vectors; HIV-1; Humans; Lentivirus; Lewy Bodies; Nerve Degeneration; Nerve Tissue Proteins; Neurons; Parkinson Disease; Rats; Substantia Nigra; Synucleins; Tumor Cells, Cultured; Tyrosine 3-Monooxygenase

2002
Lewy bodies in progressive supranuclear palsy.
    Acta neuropathologica, 2002, Volume: 104, Issue:3

    Lewy bodies (LBs), whose major component is alpha-synuclein, are a pathological hallmark of Parkinson's disease (PD) but have rarely been reported in progressive supranuclear palsy (PSP). Whether LBs in PSP represent the aging process or the coexistence of PD remains unclear. We found LBs in 5 of 16 patients with PSP. In 4 patients LBs were distributed widely throughout the brain stem and cerebrum in a pattern similar to that in PD. In the remaining patient one LB was found in the pontine reticular formation. Semiquantitative analysis showed that neuronal loss in the locus coeruleus and the dorsal vagal nucleus was more severe in patients with LBs than in patients without LBs. Double-labeling immunohistochemical studies showed co-localization of alpha-synuclein and tau in some neurons. Our study suggests that patients who have PSP with LBs constitute a subset of patients with PSP in whom Lewy body disease is also present.

    Topics: Aged; Aged, 80 and over; alpha-Synuclein; Anatomy, Regional; Humans; Immunohistochemistry; Lewy Bodies; Middle Aged; Nerve Degeneration; Nerve Tissue Proteins; Peptide Fragments; Supranuclear Palsy, Progressive; Synucleins; tau Proteins

2002
Chaperone suppression of alpha-synuclein toxicity in a Drosophila model for Parkinson's disease.
    Science (New York, N.Y.), 2002, Feb-01, Volume: 295, Issue:5556

    Parkinson's disease is a movement disorder characterized by degeneration of dopaminergic neurons in the substantia nigra pars compacta. Dopaminergic neuronal loss also occurs in Drosophila melanogaster upon directed expression of alpha-synuclein, a protein implicated in the pathogenesis of Parkinson's disease and a major component of proteinaceous Lewy bodies. We report that directed expression of the molecular chaperone Hsp70 prevented dopaminergic neuronal loss associated with alpha-synuclein in Drosophila and that interference with endogenous chaperone activity accelerated alpha-synuclein toxicity. Furthermore, Lewy bodies in human postmortem tissue immunostained for molecular chaperones, also suggesting that chaperones may play a role in Parkinson's disease progression.

    Topics: alpha-Synuclein; Animals; Animals, Genetically Modified; Brain Chemistry; Disease Models, Animal; Dopamine; Drosophila melanogaster; Drosophila Proteins; Female; Heat-Shock Proteins; HSC70 Heat-Shock Proteins; HSP40 Heat-Shock Proteins; HSP70 Heat-Shock Proteins; Humans; Inclusion Bodies; Lewy Bodies; Male; Nerve Degeneration; Nerve Tissue Proteins; Neurodegenerative Diseases; Neurons; Parkinson Disease; Parkinsonian Disorders; Synucleins; Transgenes

2002
Dopamine-dependent neurotoxicity of alpha-synuclein: a mechanism for selective neurodegeneration in Parkinson disease.
    Nature medicine, 2002, Volume: 8, Issue:6

    The mechanism by which dopaminergic neurons are selectively lost in Parkinson disease (PD) is unknown. Here we show that accumulation of alpha-synuclein in cultured human dopaminergic neurons results in apoptosis that requires endogenous dopamine production and is mediated by reactive oxygen species. In contrast, alpha-synuclein is not toxic in non-dopaminergic human cortical neurons, but rather exhibits neuroprotective activity. Dopamine-dependent neurotoxicity is mediated by 54 83-kD soluble protein complexes that contain alpha-synuclein and 14-3-3 protein, which are elevated selectively in the substantia nigra in PD. Thus, accumulation of soluble alpha-synuclein protein complexes can render endogenous dopamine toxic, suggesting a potential mechanism for the selectivity of neuronal loss in PD.

    Topics: 14-3-3 Proteins; alpha-Synuclein; Apoptosis; Cells, Cultured; Dopamine; Humans; Nerve Degeneration; Nerve Tissue Proteins; Neurons; Neuroprotective Agents; Neurotoxins; Parkinson Disease; Phosphoproteins; Substantia Nigra; Synucleins; Transfection; Tumor Cells, Cultured; Tyrosine 3-Monooxygenase

2002
Concurrence of alpha-synuclein and tau brain pathology in the Contursi kindred.
    Acta neuropathologica, 2002, Volume: 104, Issue:1

    Previous genetic analysis of the familial Parkinson's disease Contursi kindred led to the identification of an Ala53Thr pathogenic mutation in the alpha-synuclein gene. We have re-examined one of the original brains from this kindred using new immunohistochemical reagents, thioflavin S staining and immunoelectron microscopy. Surprisingly, we uncovered a dense burden of alpha-synuclein neuritic pathology and rare Lewy bodies. Immunoelectron microscopy demonstrated fibrillar alpha-synuclein-immunoreactive aggregates. Unexpected tau neuritic and less frequent perikaryal inclusions were also observed. Some inclusions were comprised of both proteins with almost complete spatial disparity. We suggest that it is important to recognize that the neurodegenerative process caused by the Ala53Thr mutation in alpha-synuclein is not identical to that seen in typical idiopathic Parkinson's disease brains.

    Topics: Adult; alpha-Synuclein; Archives; Benzothiazoles; Brain; Fluorescent Antibody Technique; Fluorescent Dyes; Humans; Male; Microscopy, Immunoelectron; Nerve Degeneration; Nerve Tissue Proteins; Neurons; Parkinson Disease; Synucleins; tau Proteins; Thiazoles

2002
Glial involvement in the degeneration process of Lewy body-bearing neurons and the degradation process of Lewy bodies in brains of dementia with Lewy bodies.
    Journal of the neurological sciences, 2001, Feb-15, Volume: 184, Issue:1

    Glial involvement in the degeneration process of Lewy body (LB)-bearing neurons and the degradation process of LBs in the cerebral cortex and amygdala in brains of dementia with Lewy bodies was investigated immunohistochemically. HLA-DR-positive microglia frequently extended their processes to degenerated neurons with alpha-synuclein-positive LBs, while some GFAP-positive astroglial processes attached to weakly alpha-synuclein-positive extracellular LBs. Some intracellular LBs were immunoreactive to anti-C4d antibody, and these LB-bearing neurons were involved by activated microglia. About half of the intracellular LBs were immunoreactive to anti-chromogranin-A (CGA) antibody, and most of CGA-positive LB-bearing neurons were surrounded by microglia. Although we could find no evident participation of TNF-alpha, a candidate cytokine that is up-regulated by microglia following CGA stimulation, in the degeneration process of LB-bearing neurons, some intracellular LBs were immunoreactive to the antibody to NF-kappaB, a transcriptional factor activated by cytokines. These findings suggest that microglia participate in the degeneration process of LB-bearing neurons via varying immunogenic elements including complement proteins, CGA and probably some cytokines, and that astroglia participate in the degradation process of LBs.

    Topics: alpha-Synuclein; Brain; Brain Chemistry; Chromogranin A; Chromogranins; Humans; Immunohistochemistry; Lewy Body Disease; Nerve Degeneration; Nerve Tissue Proteins; Neuroglia; Neurons; NF-kappa B; Synucleins; Tumor Necrosis Factor-alpha

2001
Alpha-synuclein protein is not scavenged in neuronal loss induced by kainic acid or focal ischemia.
    Brain research, 2001, Apr-13, Volume: 898, Issue:1

    Alpha-synuclein, a presynaptic protein, is markedly included in Lewy bodies (LB) in Parkinson's and LB diseases. In this study, neuronal loss and the activation of glial cells such as microglia and astrocytes were induced by neurodegenerative insults such as the injection of kainic acid and occlusion of the middle cerebral artery. In contrast, immunoreactivity for alpha-synuclein did not change even at 7 days after these insults. These results suggest that alpha-synuclein protein may be so scarcely scavenged by glial cells that it readily condenses in neurodegenerative regions.

    Topics: alpha-Synuclein; Animals; Brain; Brain Ischemia; Cell Death; Kainic Acid; Male; Nerve Degeneration; Nerve Tissue Proteins; Neurons; Rats; Rats, Wistar; Synucleins; Time Factors

2001
Co-localization of alpha-synuclein and phosphorylated tau in neuronal and glial cytoplasmic inclusions in a patient with multiple system atrophy of long duration.
    Acta neuropathologica, 2001, Volume: 101, Issue:3

    Neuronal and glial cytoplasmic inclusions (NCIs and GCIs), which contain alpha-synuclein as a major component, are characteristic cytopathological features of multiple system atrophy (MSA). We report MSA of 19 years' duration in a 73-year-old woman. Her initial symptom was parkinsonism, with dementia appearing about 8 years later. Postmortem examination showed marked atrophy of the frontal and temporal white matter and limbic system, in addition to the pathology typical of MSA. In the limbic system, severe neuronal loss and astrocytosis were observed, and the remaining neurons often had lightly eosinophilic, spherical cytoplasmic inclusions. Interestingly, a double-labeling immunofluorescence study revealed that the NCIs in the dentate gyrus and amygdaloid nucleus, and the GCIs in the frontal and temporal white matter often expressed both alpha-synuclein NACP-5 and phosphorylated tau AT8 epitopes. Double-immunolabeling electron microscopy of the NCIs in the dentate gyrus and the GCIs in the temporal white matter clearly revealed labeling of their constituent granule-associated filaments with NACP-5, and some of them were also labeled with AT8. These findings strongly suggested that some alpha-synuclein filaments were decorated with phosphorylated tau without formation of fibrils such as paired helical filaments. Immunoblotting of sarkosyl-insoluble tau indicated that the accumulated tau consisted mainly of four-repeat tau isoforms of 383 amino acids and 412 amino acids. We consider that the limbic system can be a major site of neurodegeneration in MSA of long duration. The mechanisms of such abnormal tau accumulation in the NCIs and GCIs are unknown.

    Topics: Aged; alpha-Synuclein; Brain; Cytoplasm; Female; Gliosis; Humans; Immunoblotting; Immunohistochemistry; Inclusion Bodies; Microscopy, Electron; Multiple System Atrophy; Nerve Degeneration; Nerve Tissue Proteins; Neuroglia; Neurons; Phosphorylation; Protein Isoforms; Protein Structure, Tertiary; Sarcosine; Solubility; Synucleins; tau Proteins

2001
A neuropathological study of the disturbance of the nigro-amygdaloid connections in brains from patients with dementia with Lewy bodies.
    Journal of the neurological sciences, 2001, Apr-01, Volume: 185, Issue:2

    We neuropathologically and immunohistochemically investigated characteristics of the central amygdaloid nucleus lesion and its relationship with the substantia nigra lesion in dementia with Lewy bodies (DLB) brains. Nine DLB, four Parkinson's disease (PD) and four Alzheimer-type dementia (ATD) cases were examined. The degree of neuronal loss in the substantia nigra was (+)-(+++) in DLB cases, (+++) in PD cases and (+) in ATD cases. All DLB cases showed spongy change and ubiquitin-positive spheroids in the central nucleus. The degree of spongy change was (+)-(+++) in DLB cases, (+) in PD cases and (-)-(+) in ATD cases, which was correlated with the degree of neuronal loss in the substantia nigra in DLB cases. The number of ubiquitin-positive spheroids was parallel to the degree of spongy change. The central nucleus receives dense dopaminergic fibers from the substantia nigra. Many ubiquitin-positive spheroids were also positive to alpha-synuclein and tyrosine-hydroxylase, suggesting that they derive from the degeneration of terminal or distal axons of Lewy body-bearing dopaminergic neurons in the substantia nigra. The disturbance of the dopaminergic connections from the substantia nigra to the central nucleus may be responsible for psychotic symptoms in DLB patients.

    Topics: Aged; Aged, 80 and over; alpha-Synuclein; Amygdala; Female; Humans; Immunohistochemistry; Lewy Body Disease; Locus Coeruleus; Male; Middle Aged; Nerve Degeneration; Nerve Tissue Proteins; Neural Pathways; Neurons; Substantia Nigra; Synucleins; Tyrosine 3-Monooxygenase; Ubiquitins

2001
Alpha-synuclein. Axonal transport, ligand interaction and neurodegeneration.
    Advances in experimental medicine and biology, 2001, Volume: 487

    Topics: alpha-Synuclein; Animals; Axonal Transport; Axons; Ligands; Microtubule-Associated Proteins; Nerve Degeneration; Nerve Tissue Proteins; Parkinson Disease; Phosphoproteins; Synucleins

2001
Glial cell death induced by overexpression of alpha-synuclein.
    Journal of neuroscience research, 2001, Sep-01, Volume: 65, Issue:5

    alpha-Synuclein is present in intracellular protein aggregates that are hallmarks of common neurodegenerative disorders including Parkinson disease, dementia with Lewy bodies, and multiple system atrophy. alpha-Synuclein is localized in neurons and presynaptic terminals. Under pathological conditions, however, it is also found in glia. The role of alpha-synuclein in glial cells and its relevance to the molecular pathology of neurodegenerative diseases is presently unclear. To investigate the consequence of alpha-synuclein overexpression in glia, we transfected U373 astrocytoma cells with vectors encoding wild-type human alpha-synuclein or C-terminally truncated synuclein fused to red fluorescent protein. alpha-synuclein immunocytochemistry of transfected astroglial cells revealed diffuse cytoplasmic labeling associated with discrete inclusions both within cell bodies and processes. Susceptibility to oxidative stress was increased in astroglial cells overexpressing alpha-synuclein, particularly in the presence of cytoplasmic inclusions. Furthermore, overexpression of alpha-synuclein induced apoptotic death of astroglial cells as shown by TUNEL staining. Our in vitro model is the first to replicate salient features of the glial pathology associated with alpha-synucleinopathies. It provides a simple testbed to further explore the cascade of events that leads to apoptotic glial cell death in some of these disorders; it may also be useful to assess the effects of therapeutic interventions including antioxidative and antiapoptotic strategies.

    Topics: alpha-Synuclein; Apoptosis; Astrocytes; Cell Size; Green Fluorescent Proteins; Humans; Immunohistochemistry; Indicators and Reagents; L-Lactate Dehydrogenase; Luminescent Proteins; Nerve Degeneration; Nerve Tissue Proteins; Neurodegenerative Diseases; Oxidative Stress; Peptide Fragments; Protein Structure, Tertiary; Recombinant Fusion Proteins; Red Fluorescent Protein; Synucleins; Transfection; Tumor Cells, Cultured

2001
Expression of alpha-synuclein in non-apoptotic, slowly degenerating facial motoneurones.
    Journal of neurocytology, 2001, Volume: 30, Issue:6

    The discovery that missense mutations in the alpha-synuclein gene represent a rare genetic cause of Parkinson's disease (PD) has had significant impact on the development of research into neurodegenerative disorders. It is becoming increasingly clear that alpha-synuclein plays a central role in the pathological process, which causes Lewy body formation and neurodegeneration in PD. Importantly, there is evidence to suggest that mutated alpha-synuclein is toxic to both nerve cells and glia. However, the regulation and function of wild-type alpha-synuclein are as yet ill defined. Using the facial nerve axotomy model, we have addressed the question whether the expression of alpha-synuclein in nerve cells may change in response to injury. We were particularly interested in testing the hypothesis that the severity of neuronal injury had an effect on alpha-synuclein metabolism. Facial nerve cut and crush, respectively, were performed in adult rats where normal facial motoneurones do not express alpha-synuclein. Following axotomy, a subset of facial motoneurones newly expressed high levels of alpha-synuclein immunoreactivity in their cell body and, occasionally, their nucleus. Significantly more nerve cells were labelled following facial nerve transection than following facial nerve crush. Confocal microscopy revealed a granular pattern of alpha-synuclein aggregation in degenerating nerve cells. Interestingly, the observed cell death phenotype was clearly non-apoptotic and developed over days or weeks rather than hours. Thus, axotomy of adult rat facial motoneurones triggers de novo expression of alpha-synuclein and this expression is associated with a non-apoptotic, slow form a neurodegeneration. In addition, the extent of alpha-synuclein expression is related to the severity of neuronal injury.

    Topics: alpha-Synuclein; Animals; Apoptosis; Axotomy; Facial Nerve; Motor Neurons; Nerve Crush; Nerve Degeneration; Nerve Tissue Proteins; Rats; Rats, Inbred Lew; Synucleins

2001
A Drosophila model of Parkinson's disease.
    Nature, 2000, Mar-23, Volume: 404, Issue:6776

    Parkinson's disease is a common neurodegenerative syndrome characterized by loss of dopaminergic neurons in the substantia nigra, formation of filamentous intraneuronal inclusions (Lewy bodies) and an extrapyramidal movement disorder. Mutations in the alpha-synuclein gene are linked to familial Parkinson's disease and alpha-synuclein accumulates in Lewy bodies and Lewy neurites. Here we express normal and mutant forms of alpha-synuclein in Drosophila and produce adult-onset loss of dopaminergic neurons, filamentous intraneuronal inclusions containing alpha-synuclein and locomotor dysfunction. Our Drosophila model thus recapitulates the essential features of the human disorder, and makes possible a powerful genetic approach to Parkinson's disease.

    Topics: alpha-Synuclein; Animals; Animals, Genetically Modified; Cloning, Molecular; Disease Models, Animal; Dopamine; Drosophila; Humans; Inclusion Bodies; Lewy Bodies; Locomotion; Mutation; Nerve Degeneration; Nerve Tissue Proteins; Neurons; Parkinson Disease; Retinal Degeneration; Serotonin; Synucleins; Tyrosine 3-Monooxygenase

2000
Alpha-synuclein and Parkinson's disease: selective neurodegenerative effect of alpha-synuclein fragment on dopaminergic neurons in vitro and in vivo.
    Annals of neurology, 2000, Volume: 47, Issue:5

    Missense mutations in the alpha-synuclein gene were associated with a familial Parkinson's disease, and alpha-synuclein is a major component of Lewy bodies, the intracellular inclusions that neuropathologically characterize Parkinson's disease. We investigated the neurotoxic activity of the nonamyloid component (NAC) of senile plaque, the fibrillogenic fragment (61-95) of alpha-synuclein, in vitro and in vivo. Rat primary mesencephalic neurons were exposed for 6 days to low concentrations of preaggregated NAC (0.5-10.0 microM). The number of dopaminergic neurons and dopamine content were both reduced with no effect on the general viability of the cells. At higher concentrations (25-100 microM), the neurotoxic effect of NAC was extended to all neurons. Preaggregated NAC was also toxic on a PC12 dopaminergic cell line differentiated with nerve growth factor. The intracellular localization of NAC has been identified by the exposure of neuronal cells to fluorescent peptide. In vivo application of aggregated NAC in the substantia nigra induced loss of dopaminergic neurons. Our data illustrate the selective neurotoxic effect of NAC for dopaminergic neurons and support the central role of alpha-synuclein in the pathogenesis of Parkinson's disease.

    Topics: alpha-Synuclein; Animals; Carrier Proteins; Cell Aggregation; Cells, Cultured; Disease Models, Animal; Dopamine; In Vitro Techniques; Male; Mutation, Missense; Nerve Degeneration; Nerve Growth Factor; Nerve Tissue Proteins; Neurons; Parkinson Disease; Peptide Fragments; Peptides; Point Mutation; Rats; Substantia Nigra

2000
Degeneration process of Lewy bodies in the brains of patients with dementia with Lewy bodies using alpha-synuclein-immunohistochemistry.
    Neuroscience letters, 2000, May-26, Volume: 286, Issue:1

    We investigated the degeneration process of Lewy bodies (LB) in the brains of dementia with Lewy bodies, using alpha-synuclein-immunohistochemistry. Intracellular LB, LB-related neurites and some extracellular LB were positively immunostained with anti-alpha-synuclein antibodies. Concentric LB-bearing neurons had no microglial involvement, while degenerated neurons with ill-defined LB displayed intense microglial involvement. The late stage of extracellular LB were immunoelectron-microscopically composed of loose aggregates of filamentous components with lost alpha-synuclein-immunoreactivity and penetrated astroglial processes. These findings suggest that microglias are involved during the stages from degenerated LB-bearing neurons to extracellular LB, while astroglias are involved during the stage of extracellular LB. Some intracellular LB were positive for anti-C3d and -C4d antibodies, suggesting that the classical complement pathway is activated in degenerated LB-bearing neurons, inducing microglial activation and neuronal death.

    Topics: Aged; alpha-Synuclein; Astrocytes; Brain; Cell Death; Disease Progression; Humans; Immunohistochemistry; Lewy Bodies; Lewy Body Disease; Microglia; Microscopy, Electron; Nerve Degeneration; Nerve Tissue Proteins; Neurites; Neurons; Synucleins

2000
Neuropathology in mice expressing human alpha-synuclein.
    The Journal of neuroscience : the official journal of the Society for Neuroscience, 2000, Aug-15, Volume: 20, Issue:16

    The presynaptic protein alpha-synuclein is a prime suspect for contributing to Lewy pathology and clinical aspects of diseases, including Parkinson's disease, dementia with Lewy bodies, and a Lewy body variant of Alzheimer's disease. alpha-Synuclein accumulates in Lewy bodies and Lewy neurites, and two missense mutations (A53T and A30P) in the alpha-synuclein gene are genetically linked to rare familial forms of Parkinson's disease. Under control of mouse Thy1 regulatory sequences, expression of A53T mutant human alpha-synuclein in the nervous system of transgenic mice generated animals with neuronal alpha-synucleinopathy, features strikingly similar to those observed in human brains with Lewy pathology, neuronal degeneration, and motor defects, despite a lack of transgene expression in dopaminergic neurons of the substantia nigra pars compacta. Neurons in brainstem and motor neurons appeared particularly vulnerable. Motor neuron pathology included axonal damage and denervation of neuromuscular junctions in several muscles examined, suggesting that alpha-synuclein interfered with a universal mechanism of synapse maintenance. Thy1 transgene expression of wild-type human alpha-synuclein resulted in similar pathological changes, thus supporting a central role for mutant and wild-type alpha-synuclein in familial and idiotypic forms of diseases with neuronal alpha-synucleinopathy and Lewy pathology. These mouse models provide a means to address fundamental aspects of alpha-synucleinopathy and test therapeutic strategies.

    Topics: alpha-Synuclein; Animals; Central Nervous System; Gene Expression Regulation; Humans; Lewy Bodies; Mice; Mice, Transgenic; Motor Activity; Motor Neurons; Movement Disorders; Mutation; Nerve Degeneration; Nerve Tissue Proteins; Neurodegenerative Diseases; Psychomotor Performance; Synucleins

2000
Alpha-synuclein-immunoreactive cortical Lewy bodies are associated with cognitive impairment in Parkinson's disease.
    Acta neuropathologica, 2000, Volume: 100, Issue:3

    Amygdala, hippocampus and six cortical gyri were examined for the Lewy body (LB) degeneration and Alzheimer's disease (AD) type changes in 45 patients with Parkinson's disease (PD). For detection of LBs, the brain areas were stained with an antibody against alpha-synuclein. The extent of neuropathological lesions was investigated in relation to cognitive dysfunction and apolipoprotein E (apoE) epsilon4 allele dosage. At least one cortical LB was found in 95% of cases (43/45). Furthermore, 40% of cases (18/45) had histological findings of definite AD (CERAD class C). Those PD cases with the apoE epsilon4 allele had a significantly greater number of cortical LBs than those without the apoE epsilon4 allele, but this was statistically significant only in precentral, angular and temporal gyri. The LB density correlated better with the number of plaques than with the density of tangles. The number of LBs in several cortical areas correlated significantly with the cognitive impairment. In stepwise linear regression analysis, the number of LBs in the cingulate gyrus and the amount of tangles in the temporal cortex remained statistically significant. When the CERAD class C was excluded, the correlation between cognitive decline and the number of LBs in cortical areas became even more pronounced. A stepwise linear regression analysis in these cases found the number of LBs in the frontal gyrus to be the statistically most significant predictor of cognitive impairment. This study shows, for the first time, that in PD, alpha-synuclein-positive cortical LBs are associated with cognitive impairment independent of AD-type pathology.

    Topics: Aged; Aged, 80 and over; alpha-Synuclein; Alzheimer Disease; Amygdala; Cerebral Cortex; Cognition Disorders; Female; Humans; Lewy Bodies; Male; Middle Aged; Nerve Degeneration; Nerve Tissue Proteins; Neurofibrillary Tangles; Neurons; Parkinson Disease; Plaque, Amyloid; Synucleins

2000
Induction of neuronal death by alpha-synuclein.
    The European journal of neuroscience, 2000, Volume: 12, Issue:8

    The molecular and cellular mechanisms underlying neuronal loss in neurodegenerative diseases are unclear. It is generally thought that aggregation of mutated, abnormally modified or abnormally folded proteins leads to the accumulation of extracellular, intracellular or intranuclear deposits that severely compromise cell physiology, leading to the death of the affected neurons. However, there is growing evidence that neuronal apoptosis in the absence of obvious pathological deposits could have a serious impact on the pathogenesis of neurodegenerative diseases. alpha-Synuclein has been implicated in aetiology and pathogenesis of certain neurodegenerative diseases, although the precise role of this protein in neurodegeneration is uncertain. The normal functions of alpha-synuclein and other members of the synuclein family in the development and function of the nervous system also remain elusive. Here we show that overexpression of wild-type and mutant forms of alpha-synuclein in cultured neurons, but not the closely related persyn (gamma-synuclein), causes apoptosis. These findings suggest that abnormalities of alpha-synuclein metabolism could lead to the neuronal loss occurring in certain forms of neurodegeneration before the formation of characteristic pathological lesions.

    Topics: alpha-Synuclein; Amino Acid Sequence; Animals; Apoptosis; Cell Survival; Cells, Cultured; gamma-Synuclein; Gene Expression; Humans; Mice; Molecular Sequence Data; Mutation; Neoplasm Proteins; Nerve Degeneration; Nerve Tissue Proteins; Neurons; Nodose Ganglion; Parkinson Disease; Synucleins

2000
Alpha-synuclein accumulation in a case of neurodegeneration with brain iron accumulation type 1 (NBIA-1, formerly Hallervorden-Spatz syndrome) with widespread cortical and brainstem-type Lewy bodies.
    Acta neuropathologica, 2000, Volume: 100, Issue:5

    We studied a 27-year-old woman who died after a 6-year history of progressive dementia, dystonia, ataxia, apraxia, spasticity, choreoathetosis, visual and auditory hallucinations, and optic atrophy. Magnetic resonance imaging showed decreased intensity in the globus pallidus, substantia nigra, and dentate nuclei in T2-weighted images, supporting the clinical diagnosis of neurodegeneration with brain iron accumulation type 1 (NBIA-1; formerly known as Hallervorden-Spatz syndrome). At autopsy the brain showed mild frontotemporal atrophy and discoloration of the globus pallidus and the substantia nigra pars reticularis. Histologically, features typical of NBIA-1 were found including widespread axonal spheroids and large deposits of iron pigment in the discolored regions. Additionally, excessive numbers of Lewy bodies (LBs) were found throughout all examined brain stem and cortical regions. LBs of both types, as well as Lewy neurites in this case of NBIA-1, were strongly labeled by antibodies against alpha-synuclein. These findings give further evidence that accumulation of alpha-synuclein is generally associated with LB formation, i.e., in Parkinson's disease, dementia with Lewy bodies and NBIA-1. The case presented here is particularly notable for its high number of LBs in all areas of the cerebral cortex.

    Topics: Adult; alpha-Synuclein; Brain; Brain Stem; Cerebral Cortex; Female; Humans; Immunohistochemistry; Iron; Lewy Bodies; Magnetic Resonance Imaging; Microscopy, Electron; Nerve Degeneration; Nerve Tissue Proteins; Pantothenate Kinase-Associated Neurodegeneration; Synucleins

2000
Activated microglial cells and complement factors are unrelated to cortical Lewy bodies.
    Acta neuropathologica, 2000, Volume: 100, Issue:6

    Inflammatory mechanisms have been demonstrated in Alzheimer's disease (AD) but their presence in other neurodegenerative disorders is not well documented. Complement factors and activated microglia have been reported in the substantia nigra of Parkinson's disease (PD). In the present study we investigated the cingulate gyrus of 25 autopsied patients with clinically and neuropathologically well-documented PD, with or without dementia, for the presence of (activated) microglial cells and their relation with Lewy body (LB)-bearing neurons. In addition, we studied the presence of complement factors in LBs. Of the 25 patient, 15 were clinically demented, fulfilling criteria for dementia with LBs (DLB); 7 also fulfilled CERAD morphological criteria for probable or definite Alzheimer type of dementia. Microglia clustering was seen around congophilic plaques with or without tau pathology. Microglial cells were not associated with LB-bearing neurons or noncongophilic plaques. The cortex of DLB patients without AD plaques did not show more microglial cells than the cortex of non-demented controls. The number of microglia was the lowest in young control patients who died immediately after trauma. Complement factor C3d was occasionally seen in diffusely ubiquinated neurons but late complement factors were not detected in these neurons. Double staining for complement and alpha-synuclein was negative, suggesting the absence of complement in LBs. In contrast, AD plaques in the same sections showed complement factors C3c, C3d, C1q and C5-9. In conclusion, we have found no evidence that inflammatory mechanism are involved in LB formation in cerebral cortex.

    Topics: Aged; Aged, 80 and over; alpha-Synuclein; Alzheimer Disease; Complement System Proteins; Encephalitis; Gyrus Cinguli; Humans; Lewy Bodies; Lewy Body Disease; Microglia; Middle Aged; Nerve Degeneration; Nerve Tissue Proteins; Neurons; Parkinson Disease; Ricin; Synucleins; tau Proteins; Ubiquitins

2000
Crosslinking of alpha-synuclein by advanced glycation endproducts--an early pathophysiological step in Lewy body formation?
    Journal of chemical neuroanatomy, 2000, Volume: 20, Issue:3-4

    An excess of reactive carbonyl compounds (carbonyl stress) and their reaction products, advanced glycation endproducts (AGEs), are thought to play a decisive role in the pathogenesis of neurodegenerative disorders and Parkinson's disease (PD) in particular. Accumulation of AGEs in various intracellular pathological hallmarks of PD, such as Lewy bodies, densely crosslinked intracellular protein deposits formed from neurofilament components and alpha-synuclein, have already been described in patients in advanced stages of the disease. There is, however, no indication of the involvement of AGE-induced crosslinking of alpha-synuclein in very early stages of the disease. In this study, we observed that AGEs and alpha-synuclein are similarly distributed in very early Lewy bodies in the human brain in cases with incidental Lewy body disease. These cases might be viewed as pre-Parkinson patients, i.e. patients who came for autopsy before the possible development of clinical signs of PD. AGEs are both markers of transition metal induced oxidative stress as well as, inducers of protein crosslinking and free radical formation by chemical and cellular processes. Thus, it is likely that AGE promoted formation of Lewy bodies reflects very early causative changes rather than late epiphenomenons of PD.

    Topics: Aged; Aged, 80 and over; alpha-Synuclein; Animals; Antibodies; Cross-Linking Reagents; Female; Glycation End Products, Advanced; Humans; Lewy Bodies; Male; Nerve Degeneration; Nerve Tissue Proteins; Oxidative Stress; Parkinson Disease; Rabbits; Solubility; Synucleins

2000
Increased expression of rat synuclein in the substantia nigra pars compacta identified by mRNA differential display in a model of developmental target injury.
    Journal of neurochemistry, 1999, Volume: 73, Issue:6

    Human alpha-synuclein was identified on the basis of proteolytic fragments derived from senile plaques of Alzheimer's disease, and it is the locus of mutations in some familial forms of Parkinson's disease. Its normal function and whether it may play a direct role in neural degeneration remain unknown. To explore cellular responses to neural degeneration in the dopamine neurons of the substantia nigra, we have developed a rodent model of apoptotic death induced by developmental injury to their target, the striatum. We find by mRNA differential display that synuclein is up-regulated in this model, and thus it provides an opportunity to examine directly whether synuclein plays a role in the death of these neurons or, alternatively, in compensatory responses. Up-regulation of mRNA is associated with an increase in the number of neuronal profiles immunostained for synuclein protein. At a cellular level, synuclein is almost exclusively expressed in normal neurons, rather than apoptotic profiles. Synuclein is up-regulated throughout normal postnatal development of substantia nigra neurons, but it is not further up-regulated during periods of natural cell death. We conclude that up-regulation of synuclein in the target injury model is unlikely to mediate apoptotic death and propose that it may be due to a compensatory response in neurons destined to survive.

    Topics: alpha-Synuclein; Amino Acid Sequence; Animals; Apoptosis; Base Sequence; Blotting, Northern; Blotting, Western; Cells, Cultured; Dopamine; Gene Expression Profiling; Gene Expression Regulation, Developmental; Genes; In Situ Hybridization; Molecular Sequence Data; Nerve Degeneration; Nerve Tissue Proteins; Neurons; Neurotoxins; Parkinson Disease; Quinolinic Acid; Rats; Reverse Transcriptase Polymerase Chain Reaction; RNA Splicing; RNA, Antisense; RNA, Messenger; Substantia Nigra; Subtraction Technique; Synucleins

1999
Accumulation of alpha-synuclein/NACP is a cytopathological feature common to Lewy body disease and multiple system atrophy.
    Acta neuropathologica, 1998, Volume: 96, Issue:5

    Recently, we have shown that the precursor of the non-Abeta component of Alzheimer's disease amyloid (NACP), also known as alpha-synuclein, is a major component of Lewy bodies (LBs) as well as neuronal and glial cytoplasmic inclusions in multiple system atrophy (MSA). To elucidate whether the accumulation of NACP is specific to LB disease and MSA, we further studied 83 autopsied cases with various neurological disorders, using anti-NACP antibodies. In LB disease, NACP immunoreactivity was present in all of the LBs and Lewy neurites in both the central and peripheral nervous systems, the pale bodies in the substantia nigra, and dystrophic neurites in the hippocampal CA2/3 region. Immunoelectron microscopy revealed that the reaction product was localized within filamentous structures and associated granular structures. In MSA, NACP immunoreactivity was found in the intracytoplasmic inclusions of both neuronal and oligodendroglial cells, neuronal intranuclear inclusions, and swollen neuronal processes. No NACP immunoreactivity was found in a variety of other neuronal or glial inclusions in other disorders, including Alzheimer's disease, Pick's disease, progressive supranuclear palsy, corticobasal degeneration, motor neuron disease and triplet-repeat diseases. These findings strongly suggest that the accumulation of NACP is a cytopathological feature common to LB disease and MSA.

    Topics: alpha-Synuclein; Cellular Senescence; Humans; Immunohistochemistry; Lewy Bodies; Microscopy, Immunoelectron; Multiple System Atrophy; Nerve Degeneration; Nerve Tissue Proteins; Nervous System Diseases; Parkinson Disease; Synucleins

1998
Novel alpha-synuclein-immunoreactive proteins in brain samples from the Contursi kindred, Parkinson's, and Alzheimer's disease.
    Experimental neurology, 1998, Volume: 154, Issue:2

    A specific mutation (A53T) in the encoding region for alpha-synuclein has been identified in a large multigenerational family with an autosomal dominant parkinsonism known as the Contursi kindred. In this study, we used a monoclonal antibody directed against alpha-synuclein in order to identify novel proteins in the brain of an affected member of this kindred who had come to autopsy. Homogenates from the frontal cortex and caudate nucleus were examined using Western blot techniques and compared to matched autopsy specimens from control subjects and patients with various forms of parkinsonism. Western blots, using a 15-min exposure time, revealed the expected 19-kDa band representing alpha-synuclein in all brain samples examined. However, a novel band in the 36-kDa range was also present in the Contursi brain which was not seen in cortex or caudate from control brains or in frontal cortex from 14 cases of typical Parkinson's disease. With a 24-h exposure time, this band was faintly seen in the caudate nucleus of three of the Parkinson's disease cases. Surprisingly, the 36-kDa band (as well as other high-molecular-weight bands) was also present in frontal cortex and caudate nucleus in 3 additional cases that met diagnostic criteria for both Parkinson's disease and Alzheimer's disease. A preliminary analysis of samples from the frontal cortex of 10 Alzheimer's disease cases revealed a 36-kDa band in only one instance. The identification of novel alpha-synuclein-immunoreactive bands in these various forms of parkinsonism may open new research avenues for exploring the relationship between abnormal protein deposition in the brain and one or more neurodegenerative disorders, including the Contursi form of familial parkinsonism.

    Topics: Adult; Aged; Aged, 80 and over; alpha-Synuclein; Alzheimer Disease; Antibody Specificity; Blotting, Western; Brain Chemistry; DNA Mutational Analysis; Family Health; Female; Frontal Lobe; Humans; Lewy Bodies; Male; Middle Aged; Mutation; Nerve Degeneration; Nerve Tissue Proteins; Parkinson Disease; Phosphoproteins; Substantia Nigra; Synucleins

1998
NACP, a presynaptic protein, immunoreactivity in Lewy bodies in Parkinson's disease.
    Neuroscience letters, 1997, Dec-12, Volume: 239, Issue:1

    NACP, originally identified as a precursor of the non-Abeta component of Alzheimer's disease amyloid (NAC), is now known to be identical to alpha-synuclein, a presynaptic protein in the human brain. Recently, a mutation in the alpha-synuclein gene in families with autosomal dominant Parkinson's disease (PD) was identified. We carried out immunohistochemical examinations of the brains of sporadic PD patients using anti-NACP and anti-ubiquitin antibodies. Consistent with previous studies, the anti-NACP antibody immunostained the neuropil in a punctate pattern throughout the brain. Moreover, much stronger NACP immunoreactivity was found in Lewy bodies and degenerating neurites in the brainstem. Serial sections immunolabeled with anti-ubiquitin or anti-NACP showed that all ubiquitin-immunoreactive LBs were also NACP-immunoreactive. These findings suggest that alteration of NACP metabolism is involved in the pathogenesis of PD, particularly in Lewy body formation, leading to neurodegeneration.

    Topics: Aged; Aged, 80 and over; alpha-Synuclein; Amyloid; Case-Control Studies; Humans; Immunohistochemistry; Lewy Bodies; Middle Aged; Nerve Degeneration; Nerve Tissue Proteins; Neurites; Parkinson Disease; Protein Precursors; Recombinant Proteins; Synucleins

1997
The core Alzheimer's peptide NAC forms amyloid fibrils which seed and are seeded by beta-amyloid: is NAC a common trigger or target in neurodegenerative disease?
    Chemistry & biology, 1995, Volume: 2, Issue:3

    NAC is a 35-amino-acid peptide which has been isolated from the insoluble core of Alzheimer's disease (AD) amyloid plaque. It is a fragment of alpha-synuclein (or NACP), a neuronal protein of unknown function. We noted a striking sequence similarity between NAC, the carboxyl terminus of the beta-amyloid protein, and a region of the scrapie prion protein (PrP) which has been implicated in amyloid formation.. NAC was prepared by chemical synthesis and was found to form amyloid fibrils via a nucleation-dependent polymerization mechanism. NAC amyloid fibrils effectively seed beta 1-40 amyloid formation. Amyloid fibrils comprising peptide models of the homologous beta and PrP sequences were also found to seed amyloid formation by NAC.. The in vitro model studies presented here suggest that seeding of NAC amyloid formation by the beta-amyloid protein, or seeding of amyloid fibrils of the beta-amyloid protein by NAC, may occur in vivo. Accumulation of ordered NAC aggregates in the synapse may be responsible for the neurodegeneration observed in AD and the prion disorders. Alternatively, neurodegeneration may be caused by the loss of alpha-synuclein (NACP) function.

    Topics: alpha-Synuclein; Amino Acid Sequence; Amyloid; Amyloid beta-Peptides; Chemical Phenomena; Chemistry, Physical; Crystallization; Humans; Kinetics; Microscopy, Electron; Molecular Sequence Data; Nerve Degeneration; Nerve Tissue Proteins; Solubility; Solutions; Spectroscopy, Fourier Transform Infrared; Synapses; Synucleins

1995