g(m1)-ganglioside and Disease-Models--Animal

g(m1)-ganglioside has been researched along with Disease-Models--Animal* in 115 studies

Reviews

11 review(s) available for g(m1)-ganglioside and Disease-Models--Animal

ArticleYear
The Key Role of GM1 Ganglioside in Parkinson's Disease.
    Biomolecules, 2022, 01-21, Volume: 12, Issue:2

    We have endeavored in this review to summarize our findings, which point to a systemic deficiency of ganglioside GM1 in Parkinson's disease (PD) tissues. These include neuronal tissues well known to be involved in PD, such as substantia nigra of the brain and those of the peripheral nervous system, such as the colon and heart. Moreover, we included skin and fibroblasts in the study as well as peripheral blood mononuclear cells; these are tissues not directly involved in neuronal signaling. We show similar findings for ganglioside GD1a, which is the metabolic precursor to GM1. We discuss the likely causes of these GM1 deficiencies and the resultant biochemical mechanisms underlying loss of neuronal viability and normal functioning. Strong support for this hypothesis is provided by a mouse PD model involving partial GM1 deficiency based on mono-allelic disruption of the B4galnt1 gene. We point out that progressive loss of GM1/GD1a occurs in the periphery as well as the brain, thus obviating the need to speculate PD symptom transfer between these tissues. Finally, we discuss how these findings point to a potential disease-altering therapy for PD:GM1 replacement, as is strongly implicated in animal studies and clinical trials.

    Topics: Animals; Disease Models, Animal; G(M1) Ganglioside; Leukocytes, Mononuclear; Mice; Parkinson Disease; Substantia Nigra

2022
Axonal Guillain-Barré syndrome: concepts and controversies.
    The Lancet. Neurology, 2013, Volume: 12, Issue:12

    Acute motor axonal neuropathy (AMAN) is a pure motor axonal subtype of Guillain-Barré syndrome (GBS) that was identified in the late 1990s. In Asia and Central and South America, it is the major subtype of GBS, seen in 30-65% of patients. AMAN progresses more rapidly and has an earlier peak than demyelinating GBS; tendon reflexes are relatively preserved or even exaggerated, and autonomic dysfunction is rare. One of the main causes is molecular mimicry of human gangliosides by Campylobacter jejuni lipo-oligosaccharides. In addition to axonal degeneration, electrophysiology shows rapidly reversible nerve conduction blockade or slowing, presumably due to pathological changes at the nodes or paranodes. Autoantibodies that bind to GM1 or GD1a gangliosides at the nodes of Ranvier activate complement and disrupt sodium-channel clusters and axoglial junctions, which leads to nerve conduction failure and muscle weakness. Improved understanding of the disease mechanism and pathophysiology might lead to new treatment options and improve the outlook for patients with AMAN.

    Topics: Animals; Autoantibodies; Autoantigens; Axons; Campylobacter Infections; Campylobacter jejuni; Complement Activation; Diagnosis, Differential; Disease Models, Animal; Electrodiagnosis; G(M1) Ganglioside; Global Health; Guillain-Barre Syndrome; Humans; Immunoglobulins, Intravenous; Molecular Mimicry; Motor Neurons; Neural Conduction; Plasma Exchange; Randomized Controlled Trials as Topic; Ranvier's Nodes; Reflex, Abnormal; Sodium Channels

2013
Guillain-Barré syndrome--a classical autoimmune disease triggered by infection or vaccination.
    Clinical reviews in allergy & immunology, 2012, Volume: 42, Issue:2

    Guillain-Barré syndrome (GBS) is a rare autoimmune disorder, the incidence of which is estimated to be 0.6-4/100,000 person/year worldwide. Often, GBS occurs a few days or weeks after the patient has had symptoms of a respiratory or gastrointestinal microbial infection. The disorder is sub-acute developing over the course of hours or days up to 3 to 4 weeks. About a third of all cases of Guillain-Barré syndrome are preceded by Campylobacter jejuni infection. C. jejuni strains isolated from GBS patients have a lipooligosaccharide (LOS) with a GM1-like structure. Molecular mimicry between LOS and the peripheral nerves as a cause of GBS was demonstrated in animal models of human GBS. Following the "swine flu" virus vaccine program in the USA in 1976, an increase in incidence of GBS was observed and the calculated relative risk was 6.2. Later studies have found that influenza vaccines contained structures that can induce anti-GM1 (ganglioside) antibodies after inoculation into mice. More recent information has suggested that the occurrence of GBS after currently used influenza and other vaccines is rare. GBS involves genetic and environmental factors, may be triggered by infections or vaccinations, and predisposition can be predicted by analyzing some of these factors.

    Topics: Adverse Drug Reaction Reporting Systems; Animals; Autoantibodies; Centers for Disease Control and Prevention, U.S.; Disease Models, Animal; G(M1) Ganglioside; Guillain-Barre Syndrome; Humans; Infections; Influenza Vaccines; Lipopolysaccharides; Mice; Molecular Mimicry; Peripheral Nerves; United States; United States Food and Drug Administration; Vaccination

2012
The translation of the pathological findings described in humans to experimental models of acute motor axonal neuropathy.
    Journal of the peripheral nervous system : JPNS, 2012, Volume: 17 Suppl 3

    Our mechanistic understanding of the Guillain-Barre' syndromes (GBS) was greatly enhanced by the human pathological studies of Jack Griffin and colleagues conducted in the mid-1990s. Subsequently, many of the pathological findings in human GBS were confirmed and extended in animal models. This brief account of GBS pathogenesis focuses on the studies that provided the mechanistic evidence for the role of anti-ganglioside antibodies and complement in injuring the motor axon in Campylobacter-associated GBS, thereby supporting the earlier seminal pathological observations.

    Topics: Animals; Autoantibodies; Autoantigens; Axons; Disease Models, Animal; G(M1) Ganglioside; Guillain-Barre Syndrome; Humans; Motor Neurons

2012
Ganglioside mimicry as a cause of Guillain-Barré syndrome.
    CNS & neurological disorders drug targets, 2006, Volume: 5, Issue:4

    Guillain-Barré syndrome (GBS), characterized by acute progressive limb weakness and areflexia, is the prototype of postinfectious autoimmune diseases. Campylobacter jejuni is the most frequently identified agent of infection in GBS patients, often preceding acute motor axonal neuropathy (AMAN), a variant of GBS. Anti-GM1, anti-GM1b, anti-GD1a, and anti-GalNAc-GD1a IgG antibodies are associated with AMAN. Carbohydrate mimicry [Galbeta1-3GalNAcbeta1-4(NeuAcalpha2-3)Galbeta1-] was seen between the lipo-oligosaccharide of C. jejuni isolated from an AMAN patient and human GM1 ganglioside. Sensitization with the lipo-oligosaccharide of C. jejuni induces AMAN in rabbits as does sensitization with GM1 ganglioside. Paralyzed rabbits have pathological changes in their peripheral nerves identical to changes seen in human GBS. C. jejuni infection may induce anti-ganglioside antibodies by molecular mimicry, eliciting AMAN. This is the first verification of the causative mechanism of molecular mimicry in an autoimmune disease. To express ganglioside mimics, C. jejuni requires specific gene combinations that function in sialic acid biosynthesis or transfer. The knockout mutants of these landmark genes of GBS show reduced reactivity with GBS patients' sera, and fail to induce an anti-ganglioside antibody response in mice. These genes are crucial for the induction of neuropathogenic cross-reactive antibodies. An approach for evaluating intravenous immune globulin, a treatment for GBS, based on our animal model of AMAN is also discussed in this review, and recent advances made in this field are described.

    Topics: Animals; Antigens, Bacterial; Autoantibodies; Campylobacter Infections; Disease Models, Animal; G(M1) Ganglioside; Gangliosides; Guillain-Barre Syndrome; Humans; Molecular Mimicry; Oligosaccharides

2006
[Axonal Guillain-Barré syndrome and physiological role of gangliosides in saltatory conduction of myelinated nerve fibers].
    Tanpakushitsu kakusan koso. Protein, nucleic acid, enzyme, 2004, Volume: 49, Issue:15 Suppl

    Topics: Animals; Autoantibodies; Autoimmunity; Disease Models, Animal; Epitopes; G(M1) Ganglioside; Gangliosides; Guillain-Barre Syndrome; Humans; Immunoglobulin G; Nerve Fibers, Myelinated; Neural Conduction; Ranvier's Nodes

2004
Spinal cord injury in the rat.
    Progress in neurobiology, 1998, Volume: 56, Issue:3

    Only limited therapeutic measures are currently available for the treatment of spinal cord injury. This review describes the pathologic mechanisms of trauma-induced spinal cord injury in rats, which will contribute to new understanding of the pathologic process leading to spinal cord injury and to further development of new therapeutic strategies. Spinal cord injury induced by trauma is a consequence of an initial physical insult and a subsequent progressive injury process that involves various pathochemical events leading to tissue destruction; the latter process should therefore be a target of pharmacological treatment. Recently, activated neutrophils have been shown to be implicated in the latter process of the spinal cord injury in rats. Activated neutrophils damage the endothelial cells by releasing inflammatory mediators such as neutrophil elastase and oxygen free radicals. Adhesion of activated neutrophils to the endothelial cell could also play a role in endothelial cell injury. This endothelial cell injury could in turn induce microcirculatory disturbances leading to spinal cord ischemia. We have found that some therapeutic agents that inhibit neutrophil activation alleviate the motor disturbances observed in the rat model of spinal cord injury. Methylprednisolone (MPS) and GM1 ganglioside, which are the only two pharmacological agents currently clinically available for treatment of acute spinal cord injury, do not inhibit neutrophil activation in this rat model. Taken together, these observations raise a possibility that other pharmacological agents that inhibit neutrophil activation used in conjunction with MPS or GM1 ganglioside may have a synergistic effect in the treatment of traumatic spinal cord injury in humans.

    Topics: Animals; Disease Models, Animal; G(M1) Ganglioside; Glucocorticoids; Leukocytes; Methylprednisolone; Rats; Spinal Cord Injuries

1998
Does effect of a neuroprotective agent on volume of experimental animal cerebral infarct predict effect of the agent on clinical outcome in human stroke?
    Annals of the New York Academy of Sciences, 1997, Oct-15, Volume: 825

    Topics: Animals; Cerebral Infarction; Cerebrovascular Disorders; Clinical Trials as Topic; Disease Models, Animal; Fibrinolytic Agents; G(M1) Ganglioside; Glucocorticoids; Hemodilution; Humans; Neuroprotective Agents; Nimodipine; Tissue Plasminogen Activator

1997
[Recent progress in development of psychotropic drugs (3)--Antiparkinsonian agents applied in the treatment of Parkinson's disease or are under investigation for patients or model animals].
    Nihon shinkei seishin yakurigaku zasshi = Japanese journal of psychopharmacology, 1996, Volume: 16, Issue:4

    Tremor, akinesia, rigidity and postual instability are key signs of Parkinson's disease. The most important one is akinesia, which includes decreased spontaneous locomotor activity, slowness of movement, awkwardness and freezing. On the other hand, an electrical focal lesion in the brain, neurotoxin to dopaminergic neurons such as 6-hydroxydopamine (6-OHDA) or I-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP), cholinomimetic tremorogenic agents such as oxotremorine or tremorine, monoamine depleting agents such as reserpine or tetrabenazine, or dopamine receptor antagonists such as haloperidol are applied to render animal parkinsonism. The estimation of locomotor activity can be done accurately in animal models. Tremor can be studied using the animals treated by cerebral focal lesion, neurotoxins or cholinomimetics. Skillfulness is hard to estimate in animals, however, it can be done in primates. Freezing appeared in patients with levodopa treatment over a long period. This is a specific motor sign in Parkinson's disease, and cannot be observed in animals. Supplementing dopamine by levodopa administration, retarding the metabolism of levodopa or dopamine by dopa decarboxylase inhibitor (DCI), monoamine oxidase inhibitor type B (MAO-B) inhibitor or catechol-O-methyltransferase (COMT) inhibitor, dopamine receptor agonists, anticholinergic agents, dopamine release enhancer/ uptake inhibitor, N-methyl-D-aspartate (NMDA) receptor antagonists, adenosine receptor antagonists, neurotrophic factors, GM1-ganglioside and nicotinic receptor agonists have been applied in the treatment of Parkinson's disease or are under investigation for patients. Agents to facilitate nerve growth or to inhibit the degeneration of nerves will be developed in the future.

    Topics: Animals; Antiparkinson Agents; Cholinergic Antagonists; Disease Models, Animal; Dopamine Agents; G(M1) Ganglioside; Nerve Growth Factors; Nerve Tissue Proteins; Nicotinic Agonists; Parkinson Disease; Purinergic P1 Receptor Antagonists; Receptors, Dopamine; Receptors, N-Methyl-D-Aspartate

1996
[Anti-ganglioside antibodies and motor neuropathy in relation to nerve conduction block].
    No to shinkei = Brain and nerve, 1992, Volume: 44, Issue:8

    Topics: Adult; Animals; Autoantibodies; Disease Models, Animal; G(M1) Ganglioside; Humans; Male; Middle Aged; Motor Neuron Disease; Neural Conduction; Rats

1992
Neurotransmitter specific alterations in dementing disorders: insights from animal models.
    Journal of psychiatric research, 1984, Volume: 18, Issue:4

    Recent years have witnessed considerable change in the conceptualization of the pathophysiology of the cognitive impairments in dementing disorders, as a result of synaptic neurochemical analyses. Profound reductions in the forebrain cholinergic projections occur in Alzheimer's disease. In GM1 gangliosidosis, variable alterations in neurotransmitter related processes that are located in synaptic membranes have been described. Exploitation of animal models of human disorders resulting in dementia may further clarify the dynamic alterations in the biochemical processes required for effective neurotransmission in cortex.

    Topics: Alzheimer Disease; Animals; Brain; Cats; Cerebral Cortex; Cholinergic Fibers; Dementia; Disease Models, Animal; Dopamine; G(M1) Ganglioside; gamma-Aminobutyric Acid; Gangliosidoses; Glutamates; Glutamic Acid; Humans; Neurotransmitter Agents; Norepinephrine; Rats; Serotonin; Synaptic Transmission

1984

Trials

1 trial(s) available for g(m1)-ganglioside and Disease-Models--Animal

ArticleYear
Effects of ganglioside GM1 and erythropoietin on spinal cord injury in mice: Functional and immunohistochemical assessments.
    Clinics (Sao Paulo, Brazil), 2022, Volume: 77

    To evaluate the functional and immunohistochemical effects of ganglioside GM1 and erythropoietin following experimental spinal cord injury.. Thirty-two male BALB/c mice were subjected to experimental spinal cord injury using the NYU Impactor device and were randomly divided into the following groups: GM1 group, receiving standard ganglioside GM1 (30 mg/kg); erythropoietin group, receiving erythropoietin (1000 IU/kg); combination group, receiving both drugs; and control group, receiving saline (0.9%). Animals were evaluated according to the Basso Mouse Scale (BMS) and Hindlimb Mouse Function Score (MFS). After euthanasia, the immunohistochemistry of the medullary tissue of mice was analyzed. All animals received intraperitoneal treatment.. The GM1 group had higher BMS and MFS scores at the end of the experiment when compared to all other groups. The combination group had higher BMS and MFS scores than the erythropoietin and control groups. The erythropoietin group had higher BMS and MFS scores than the control group. Immunohistochemical tissue analysis showed a significant difference among groups. There was a significant increase in myelinated axons and in the myelinated axon length in the erythropoietin group when compared to the other intervention groups (p < 0.01).. Erythropoietin and GM1 have therapeutic effects on axonal regeneration in mice subjected to experimental spinal cord injury, and administration of GM1 alone had the highest scores on the BMS and MFS scales.

    Topics: Animals; Disease Models, Animal; Epoetin Alfa; Erythropoietin; G(M1) Ganglioside; Injections, Intraperitoneal; Male; Mice; Spinal Cord; Spinal Cord Injuries

2022

Other Studies

103 other study(ies) available for g(m1)-ganglioside and Disease-Models--Animal

ArticleYear
GM1 Reduced the Symptoms of Autism Spectrum Disorder by Suppressing α-Syn Through Activating Autophagy.
    Journal of molecular neuroscience : MN, 2023, Volume: 73, Issue:4-5

    Autism spectrum disorder (ASD) is a neurodevelopmental disorder that cannot be cured. The ASD rat model was developed in this study to demonstrate the role and mechanism of ganglioside GM1 (GM1). Rats were given valproic acid (VPA) to create the ASD rat model. The rats' behaviors were assessed using the Y-maze test, open-field test, three-chamber social interaction test, and Morris water maze test. Relative levels of glutathione (GSH), malondialdehyde (MDA), catalase (CAT), reactive oxygen species (ROS), and superoxide dismutase (SOD) were quantitated using relative kits. Nissl, TUNEL, immunofluorescent, and immunohistochemistry staining techniques were used. GM1 treatment improved the ASD model rats' behavior disorders, including locomotor activity and exploratory behavior, social interaction, learning and memory capacity, and repetitive behavior. Following GM1 injection, striatal neurons grew and apoptosis decreased. GM1 reduced the excessively elevated α-Syn in ASD by encouraging autophagy. The behavior disorder of ASD model rats was exacerbated by autophagy inhibition, which also increased α-Syn levels. By increasing autophagy, GM1 reduced α-Syn levels and, ultimately, improved behavioral abnormalities in ASD model rats.

    Topics: Animals; Autism Spectrum Disorder; Autophagy; Disease Models, Animal; Female; G(M1) Ganglioside; Humans; Maze Learning; Prenatal Exposure Delayed Effects; Rats; Social Behavior; Valproic Acid

2023
Evaluation of the effect of intrathecal GM1 in 24, 48, and 72 hours after acute spinal cord injury in rats.
    Clinics (Sao Paulo, Brazil), 2023, Volume: 78

    The aim of this study was to evaluate the best timing and feasibility of intrathecal application of sodium monosialoganglioside (GM1) after spinal cord contusion in Wistar rats as an experimental model.. Forty Wistar rats were submitted to contusion spinal cord injury after laminectomy. The animals were randomized and divided into four groups: Group 1 - Intrathecal application of GM1 24 hours after contusion; Group 2 - Intrathecal application of GM1 48 hours after contusion; Group 3 - intrathecal application of GM1 72 hours after contusion; Group 4 - Sham, with laminectomy and intrathecal application of 0.5 mL of 0.9% saline solution, without contusion. The recovery of locomotor function was evaluated at seven different moments by the Basso, Beattie, and Bresnahan (BBB) test. They were also assessed by the horizontal ladder, with sensory-motor behavioral assessment criteria, pre-and postoperatively.. This experimental study showed better functional scores in the group submitted to the application of GM1, with statistically significant results, showing a mean increase when evaluated on known motor tests like the horizontal ladder and BBB, at all times of evaluation (p < 0.05), especially in group 2 (48 hours after spinal cord injury). Also, fewer mistakes and slips over the horizontal ladder were observed, and many points were achieved at the BBB scale analysis.. The study demonstrated that the intrathecal application of GM1 after spinal cord contusion in Wistar rats is feasible. The application 48 hours after the injury presented the best functional results.

    Topics: Animals; Contusions; Disease Models, Animal; G(M1) Ganglioside; Rats; Rats, Wistar; Recovery of Function; Spinal Cord; Spinal Cord Injuries

2023
Naïve Huntington's disease microglia mount a normal response to inflammatory stimuli but display a partially impaired development of innate immune tolerance that can be counteracted by ganglioside GM1.
    Journal of neuroinflammation, 2023, Nov-23, Volume: 20, Issue:1

    Chronic activation and dysfunction of microglia have been implicated in the pathogenesis and progression of many neurodegenerative disorders, including Huntington's disease (HD). HD is a genetic condition caused by a mutation that affects the folding and function of huntingtin (HTT). Signs of microglia activation have been observed in HD patients even before the onset of symptoms. It is unclear, however, whether pro-inflammatory microglia activation in HD results from cell-autonomous expression of mutant HTT, is the response of microglia to a diseased brain environment, or both. In this study, we used primary microglia isolated from HD knock-in (Q140) and wild-type (Q7) mice to investigate their response to inflammatory conditions in vitro in the absence of confounding effects arising from brain pathology. We show that naïve Q140 microglia do not undergo spontaneous pro-inflammatory activation and respond to inflammatory triggers, including stimulation of TLR4 and TLR2 and exposure to necrotic cells, with similar kinetics of pro-inflammatory gene expression as wild-type microglia. Upon termination of the inflammatory insult, the transcription of pro-inflammatory cytokines is tapered off in Q140 and wild-type microglia with similar kinetics. However, the ability of Q140 microglia to develop tolerance in response to repeated inflammatory stimulations is partially impaired in vitro and in vivo, potentially contributing to the establishment of chronic neuroinflammation in HD. We further show that ganglioside GM1, a glycosphingolipid with anti-inflammatory effects on wild-type microglia, not only decreases the production of pro-inflammatory cytokines and nitric oxide in activated Q140 microglia, but also dramatically dampen microglia response to re-stimulation with LPS in an experimental model of tolerance. These effects are independent from the expression of interleukin 1 receptor associated kinase 3 (Irak-3), a strong modulator of LPS signaling involved in the development of innate immune tolerance and previously shown to be upregulated by immune cell treatment with gangliosides. Altogether, our data suggest that external triggers are required for HD microglia activation, but a cell-autonomous dysfunction that affects the ability of HD microglia to acquire tolerance might contribute to the establishment of neuroinflammation in HD. Administration of GM1 might be beneficial to attenuate chronic microglia activation and neuroinflammation.

    Topics: Animals; Cytokines; Disease Models, Animal; G(M1) Ganglioside; Humans; Huntington Disease; Lipopolysaccharides; Mice; Microglia; Neuroinflammatory Diseases

2023
Peripheral Delivery of Ganglioside GM1 Exacerbates the Pathogenesis of Alzheimer's Disease in a Mouse Model.
    Neuroscience bulletin, 2022, Volume: 38, Issue:1

    Topics: Alzheimer Disease; Amyloid beta-Peptides; Animals; Disease Models, Animal; G(M1) Ganglioside; Mice; Peptide Fragments

2022
AAV9-coGLB1 Improves Lysosomal Storage and Rescues Central Nervous System Inflammation in a Mutant Mouse Model of GM1 Gangliosidosis.
    Current gene therapy, 2022, Volume: 22, Issue:4

    GM1 gangliosidosis (GM1) is an autosomal recessive disorder characterized by the deficiency of beta-galactosidase (β-gal), a ubiquitous lysosomal enzyme that catalyzes the hydrolysis of GM1 ganglioside.. The study aims to explore the application of the AAV9-coGLB1 for effective treatment in a GM1 gangliosidosis mutant mouse model.. We designed a novel adeno-associated virus 9 (AAV9) vector expressing β-gal (AAV9- coGLB1) to treat GM1 gangliosidosis. The vector, injected via the caudal vein at 4 weeks of age, drove the widespread and sustained expression of β-gal for up to 32 weeks in the Glb1. The increased levels of β-gal reduced the pathological damage occurring in GM1 mice. Histological analyses showed that myelin deficits and neuron-specific pathology were reduced in the cerebral cortex region of AAV9-coGLB1-treated mice. Immunohistochemical staining showed that the accumulation of GM1 ganglioside was also reduced after gene therapy. The reduction of the storage in these regions was accompanied by a decrease in activated microglia. In addition, AAV9 treatment reversed the blockade of autophagic flux in GM1 mice.. These results show that AAV9-coGLB1 reduces the pathological signs of GM1 gangliosidosis in a mouse model.

    Topics: Animals; Central Nervous System; Dependovirus; Disease Models, Animal; G(M1) Ganglioside; Gangliosidosis, GM1; Inflammation; Lysosomes; Mice

2022
GM1 ganglioside modifies microglial and neuroinflammatory responses to α-synuclein in the rat AAV-A53T α-synuclein model of Parkinson's disease.
    Molecular and cellular neurosciences, 2022, Volume: 120

    Among the pathological events associated with the dopaminergic neurodegeneration characteristic of Parkinson's disease (PD) are the accumulation of toxic forms of α-synuclein and microglial activation associated with neuroinflammation. Although numerous other processes may participate in the pathogenesis of PD, the two factors mentioned above may play critical roles in the initiation and progression of dopamine neuron degeneration in PD. In this study, we employed a slowly progressing model of PD using adeno-associated virus-mediated expression of human A53T α-synuclein into the substantia nigra on one side of the brain and examined the microglial response in the striatum on the injected side compared to the non-injected (control) side. We further examined the extent to which administration of the neuroprotective ganglioside GM1 influenced α-synuclein-induced glial responses. Changes in a number of microglial morphological measures (i.e., process length, number of endpoints, fractal dimension, lacunarity, density, and cell perimeter) were indicative of the presence of activated microglial and an inflammatory response on the injected side of the brain, compared to the control side. In GM1-treated animals, no significant differences in microglial morphology were observed between the injected and control striata. Follow-up studies showed that mRNA expression for several inflammation-related genes was increased on the A53T α-synuclein injected side vs. the non-injected side in saline-treated animals and that such changes were not observed in GM1-treated animals. These data show that inhibition of microglial activation and potentially damaging neuroinflammation by GM1 ganglioside administration may be among the many factors that contribute to the neuroprotective effects of GM1 in this model and possibly in human PD.

    Topics: alpha-Synuclein; Animals; Disease Models, Animal; Dopamine; G(M1) Ganglioside; Inflammation; Microglia; Parkinson Disease; Rats; Substantia Nigra

2022
Schwann cell nodal membrane disruption triggers bystander axonal degeneration in a Guillain-Barré syndrome mouse model.
    The Journal of clinical investigation, 2022, 07-15, Volume: 132, Issue:14

    In Guillain-Barré syndrome (GBS), both axonal and demyelinating variants can be mediated by complement-fixing anti-GM1 ganglioside autoantibodies that target peripheral nerve axonal and Schwann cell (SC) membranes, respectively. Critically, the extent of axonal degeneration in both variants dictates long-term outcome. The differing pathomechanisms underlying direct axonal injury and the secondary bystander axonal degeneration following SC injury are unresolved. To investigate this, we generated glycosyltransferase-disrupted transgenic mice that express GM1 ganglioside either exclusively in neurons [GalNAcT-/--Tg(neuronal)] or glia [GalNAcT-/--Tg(glial)], thereby allowing anti-GM1 antibodies to solely target GM1 in either axonal or SC membranes, respectively. Myelinated-axon integrity in distal motor nerves was studied in transgenic mice exposed to anti-GM1 antibody and complement in ex vivo and in vivo injury paradigms. Axonal targeting induced catastrophic acute axonal disruption, as expected. When mice with GM1 in SC membranes were targeted, acute disruption of perisynaptic glia and SC membranes at nodes of Ranvier (NoRs) occurred. Following glial injury, axonal disruption at NoRs also developed subacutely, progressing to secondary axonal degeneration. These models differentiate the distinctly different axonopathic pathways under axonal and glial membrane targeting conditions, and provide insights into primary and secondary axonal injury, currently a major unsolved area in GBS research.

    Topics: Animals; Autoantibodies; Disease Models, Animal; G(M1) Ganglioside; Gangliosides; Guillain-Barre Syndrome; Mice; Mice, Transgenic; Schwann Cells

2022
Intranasal infusion of GD3 and GM1 gangliosides downregulates alpha-synuclein and controls tyrosine hydroxylase gene in a PD model mouse.
    Molecular therapy : the journal of the American Society of Gene Therapy, 2021, 10-06, Volume: 29, Issue:10

    Parkinson's disease (PD) is characterized by Lewy bodies (composed predominantly of alpha-synuclein [aSyn]) and loss of pigmented midbrain dopaminergic neurons comprising the nigrostriatal pathway. Most PD patients show significant deficiency of gangliosides, including GM1, in the brain, and GM1 ganglioside appears to keep dopaminergic neurons functioning properly. Thus, supplementation of GM1 could potentially provide some rescuing effects. In this study, we demonstrate that intranasal infusion of GD3 and GM1 gangliosides reduces intracellular aSyn levels. GM1 also significantly enhances expression of tyrosine hydroxylase (TH) in the substantia nigra pars compacta of the A53T aSyn overexpressing mouse, following restored nuclear expression of nuclear receptor related 1 (Nurr1, also known as NR4A2), an essential transcription factor for differentiation, maturation, and maintenance of midbrain dopaminergic neurons. GM1 induces epigenetic activation of the TH gene, including augmentation of acetylated histones and recruitment of Nurr1 to the TH promoter region. Our data indicate that intranasal administration of gangliosides could reduce neurotoxic proteins and restore functional neurons via modulating chromatin status by nuclear gangliosides.

    Topics: Administration, Intranasal; alpha-Synuclein; Animals; Cell Line; Disease Models, Animal; Down-Regulation; Epigenesis, Genetic; G(M1) Ganglioside; Gangliosides; Gene Expression Regulation; Humans; Male; Mice; Parkinson Disease; Substantia Nigra; Tyrosine 3-Monooxygenase

2021
Mice deficient in GM1 manifest both motor and non-motor symptoms of Parkinson's disease; successful treatment with synthetic GM1 ganglioside.
    Experimental neurology, 2020, Volume: 329

    Parkinson's disease (PD) is a major neurodegenerative disorder characterized by a variety of non-motor symptoms in addition to the well-recognized motor dysfunctions that have commanded primary interest. We previously described a new PD mouse model based on heterozygous disruption of the B4galnt1 gene leading to partial deficiency of the GM1 family of gangliosides that manifested several nigrostriatal neuropathological features of PD as well as movement impairment. We now show this mouse also suffers three non-motor symptoms characteristic of PD involving the gastrointestinal, sympathetic cardiac, and cerebral cognitive systems. Treatment of these animals with a synthetic form of GM1 ganglioside, produced by transfected E. coli, proved ameliorative of these symptoms as well as the motor defect. These findings further suggest subnormal GM1 to be a systemic defect constituting a major risk factor in sporadic PD and indicate the B4galnt1(+/-) (HT) mouse to be a true neuropathological model that recapitulates both motor and non-motor lesions of this condition.

    Topics: Animals; Disease Models, Animal; Female; G(M1) Ganglioside; Gastrointestinal Diseases; Male; Memory Disorders; Mice; Mice, Inbred C57BL; Mice, Knockout; Mice, Transgenic; Motor Skills Disorders; N-Acetylgalactosaminyltransferases; Parkinson Disease

2020
Molecular basis for a new bovine model of Niemann-Pick type C disease.
    PloS one, 2020, Volume: 15, Issue:9

    Niemann-Pick type C disease is a lysosomal storage disease affecting primarily the nervous system that results in premature death. Here we present the first report and investigation of Niemann-Pick type C disease in Australian Angus/Angus-cross calves. After a preliminary diagnosis of Niemann-Pick type C, samples from two affected calves and two obligate carriers were analysed using single nucleotide polymorphism genotyping and homozygosity mapping, and NPC1 was considered as a positional candidate gene. A likely causal missense variant on chromosome 24 in the NPC1 gene (NM_174758.2:c.2969C>G) was identified by Sanger sequencing of cDNA. SIFT analysis, protein alignment and protein modelling predicted the variant to be deleterious to protein function. Segregation of the variant with disease was confirmed in two additional affected calves and two obligate carrier dams. Genotyping of 403 animals from the original herd identified an estimated allele frequency of 3.5%. The Niemann-Pick type C phenotype was additionally confirmed via biochemical analysis of Lysotracker Green, cholesterol, sphingosine and glycosphingolipids in fibroblast cell cultures originating from two affected calves. The identification of a novel missense variant for Niemann-Pick type C disease in Angus/Angus-cross cattle will enable improved breeding and management of this disease in at-risk populations. The results from this study offer a unique opportunity to further the knowledge of human Niemann-Pick type C disease through the potential availability of a bovine model of disease.

    Topics: Amino Acid Sequence; Animals; Base Sequence; Cattle; Cells, Cultured; Cholera Toxin; Cholesterol; Disease Models, Animal; DNA, Complementary; Fibroblasts; G(M1) Ganglioside; Homozygote; Mutation; Niemann-Pick C1 Protein; Niemann-Pick Disease, Type C; Phenotype; Polymorphism, Single Nucleotide; Polysaccharides; RNA, Messenger

2020
Visualisation of cholesterol and ganglioside GM1 in zebrafish models of Niemann-Pick type C disease and Smith-Lemli-Opitz syndrome using light sheet microscopy.
    Histochemistry and cell biology, 2020, Volume: 154, Issue:5

    Lysosomal storage diseases are the most common cause of neurodegeneration in children. They are characterised at the cellular level by the accumulation of storage material within lysosomes. There are very limited therapeutic options, and the search for novel therapies has been hampered as few good small animal models are available. Here, we describe the use of light sheet microscopy to assess lipid storage in drug and morpholino induced zebrafish models of two diseases of cholesterol homeostasis with lysosomal dysfunction: First, Niemann-Pick type C disease (NPC), caused by mutations in the lysosomal transmembrane protein NPC1, characterised by intralysosomal accumulation of cholesterol and several other lipids. Second, Smith-Lemli-Opitz syndrome (SLOS), caused by mutations in 7-dehydrocholesterol reductase, which catalyses the last step of cholesterol biosynthesis and is characterised by intralysosomal accumulation of dietary cholesterol. This is the first description of a zebrafish SLOS model. We find that zebrafish accurately model lysosomal storage and disease-specific phenotypes in both diseases. Increased cholesterol and ganglioside GM1 were observed in sections taken from NPC model fish, and decreased cholesterol in SLOS model fish, but these are of limited value as resolution is poor, and accurate anatomical comparisons difficult. Using light sheet microscopy, we were able to observe lipid changes in much greater detail and identified an unexpected accumulation of ganglioside GM1 in SLOS model fish. Our data demonstrate, for the first time in zebrafish, the immense potential that light sheet microscopy has in aiding the resolution of studies involving lysosomal and lipid disorders.

    Topics: Animals; Cholesterol; Disease Models, Animal; G(M1) Ganglioside; Lysosomes; Microscopy, Fluorescence; Niemann-Pick Disease, Type C; Smith-Lemli-Opitz Syndrome; Zebrafish

2020
GM1 Ganglioside Modifies α-Synuclein Toxicity and is Neuroprotective in a Rat α-Synuclein Model of Parkinson's Disease.
    Scientific reports, 2019, 06-10, Volume: 9, Issue:1

    While GM1 may interact with α-synuclein in vitro to inhibit aggregation, the ability of GM1 to protect against α-synuclein toxicity in vivo has not been investigated. We used targeted adeno-associated viral vector (AAV) overexpression of human mutant α-synuclein (A53T) in the rat substantia nigra (SN) to produce degeneration of SN dopamine neurons, loss of striatal dopamine levels, and behavioral impairment. Some animals received daily GM1 ganglioside administration for 6 weeks, beginning 24 hours after AAV-A53T administration or delayed start GM1 administration for 5 weeks beginning 3 weeks after AAV-A53T administration. Both types of GM1 administration protected against loss of SN dopamine neurons and striatal dopamine levels, reduced α-synuclein aggregation, and delayed start administration of GM1 reversed early appearing behavioral deficits. These results extend prior positive results in MPTP models, are consistent with the results of a small clinical study of GM1 in PD patients that showed slowing of symptom progression with chronic use, and argue for the continued refinement and development of GM1 as a potential disease modifying therapy for PD.

    Topics: alpha-Synuclein; Animals; Behavior, Animal; Corpus Striatum; Dependovirus; Disease Models, Animal; Dopamine; Dopaminergic Neurons; G(M1) Ganglioside; Gene Expression Regulation; Genetic Vectors; Humans; Neuroprotective Agents; Parkinson Disease; Rats; Substantia Nigra

2019
Induction of ganglioside synthesis in Drosophila brain accelerates assembly of amyloid β protein.
    Scientific reports, 2018, 05-29, Volume: 8, Issue:1

    The assembly and deposition of amyloid β protein (Aβ) is a fundamental event during the early stages of Alzheimer's disease (AD) and cerebral amyloid angiopathy. A growing body of evidence indicates that gangliosides form a pathological platform for the generation of ganglioside-bound Aβ, which facilitates the assembly of soluble Aβs; however, the molecular mechanisms underlying the binding of Aβ to gangliosides in the brain remain unclear due to the lack of an in vivo system that may address this issue. In insects, including the fruit fly Drosophila melanogaster, gangliosides are not intrinsically present at a detectable level. We herein demonstrate that ganglioside expression is inducible in Drosophila via the expression of transgenes of ganglioside synthesis enzymes and the feeding of exogenous sialic acid, and also that the induction of ganglioside synthesis significantly accelerates Aβ assembly in vivo. Our results support the hypothesis that gangliosides are responsible for Aβ assembly in vivo and also provide an opportunity to develop a valuable model for basic research as well as a therapeutic strategy for AD.

    Topics: Acetyltransferases; Alzheimer Disease; Amyloid beta-Peptides; Animals; Brain; Cell Membrane; Disease Models, Animal; Drosophila melanogaster; Drosophila Proteins; G(M1) Ganglioside; Galactosyltransferases; Gangliosides; Humans; N-Acetylneuraminic Acid; Protein Binding; Transgenes

2018
GM1 ganglioside prevents axonal regeneration inhibition and cognitive deficits in a mouse model of traumatic brain injury.
    Scientific reports, 2018, 09-06, Volume: 8, Issue:1

    Traumatic Brain Injury (TBI) is one of the most common causes of neurological damage in young populations. It has been previously suggested that one of the mechanisms that underlie brain injury is Axonal Outgrowth Inhibition (AOI) that is caused by altered composition of the gangliosides on the axon surface. In the present study, we have found a significant reduction of GM1 ganglioside levels in the cortex in a closed head traumatic brain injury model of a mouse, induced by a weight drop device. In addition, axonal regeneration in the brains of the injured mice was affected as seen by the expression of the axonal marker pNF-H and the growth cones (visualized by F-actin and β-III-tubulin). NeuN immunostaining revealed mTBI-induced damage to neuronal survival. Finally, as expected, spatial and visual memories (measured by the Y-maze and the Novel Object Recognition tests, respectively) were also damaged 7 and 30 days post injury. A single low dose of GM1 shortly after the injury (2 mg/kg; IP) prevented all of the deficits mentioned above. These results reveal additional insights into the neuroprotective characteristics of GM1 in prevention of biochemical, cellular and cognitive changes caused by trauma, and may suggest a potential intervention for mTBI.

    Topics: Animals; Axons; Brain Injuries, Traumatic; Cognition Disorders; Disease Models, Animal; G(M1) Ganglioside; Male; Mice; Mice, Inbred ICR; Neurodegenerative Diseases; Neuroprotective Agents

2018
Disease-modifying effects of ganglioside GM1 in Huntington's disease models.
    EMBO molecular medicine, 2017, Volume: 9, Issue:11

    Huntington's disease (HD) is a progressive neurodegenerative disorder characterized by motor, cognitive and psychiatric problems. Previous studies indicated that levels of brain gangliosides are lower than normal in HD models and that administration of exogenous ganglioside GM1 corrects motor dysfunction in the YAC128 mouse model of HD In this study, we provide evidence that intraventricular administration of GM1 has profound disease-modifying effects across HD mouse models with different genetic background. GM1 administration results in decreased levels of mutant huntingtin, the protein that causes HD, and in a wide array of beneficial effects that include changes in levels of DARPP32, ferritin, Iba1 and GFAP, modulation of dopamine and serotonin metabolism, and restoration of normal levels of glutamate, GABA, L-Ser and D-Ser. Treatment with GM1 slows down neurodegeneration, white matter atrophy and body weight loss in R6/2 mice. Motor functions are significantly improved in R6/2 mice and restored to normal in Q140 mice, including gait abnormalities that are often resistant to treatments. Psychiatric-like and cognitive dysfunctions are also ameliorated by GM1 administration in Q140 and YAC128 mice. The widespread benefits of GM1 administration, at molecular, cellular and behavioural levels, indicate that this ganglioside has strong therapeutic and disease-modifying potential in HD.

    Topics: Animals; Behavior, Animal; Body Weight; Brain; Calcium-Binding Proteins; Disease Models, Animal; Dopamine; Dopamine and cAMP-Regulated Phosphoprotein 32; Ferritins; G(M1) Ganglioside; gamma-Aminobutyric Acid; Glial Fibrillary Acidic Protein; Glutamic Acid; Huntingtin Protein; Huntington Disease; Infusions, Intraventricular; Mice; Mice, Transgenic; Microfilament Proteins; Serotonin; Survival Rate

2017
An in vivo study of hypoxia-inducible factor-1α signaling in ginsenoside Rg1-mediated brain repair after hypoxia/ischemia brain injury.
    Pediatric research, 2017, Volume: 81, Issue:1-1

    Hypoxia/ischemia (HI) brain injury is a common central nervous system insult in newborns. Studies have demonstrated bioactivity of ginsenoside Rg1 in increasing neural viability and promoting angiogenesis. However, there are few reports on roles of Rg1 in brain repair of neonatal HI, and the mechanisms involved are unclear.. a neonatal HI model was established by a modified Rice-Vannucci model (RVM) and pups received ginsenoside Rg1 or monosialotetrahexosyl ganglioside (GM1) treatment. Neurological function and pathologic damage of rats were evaluated. Cellular apoptosis was detected with Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay. Immunohistochemistry for von willebrand factor (vwf) was used to label micro vessels. Expression levels of hypoxia-inducible factor-1α (HIF-1α), vascular endothelial growth factor (VEGF), and cleaved caspase 3 (CC3) were detected by western blot.. Both Rg1 and GM1 reduced neurological impairment and pathologic damage after HI by enhancing neural survival. Rg1, but not GM1, could also facilitate angiogenesis after HI. These pharmacological effects of Rg1 may be attributed to regulation of expression level of VEGF and CC3 and HIF-1α signaling pathway was involved.. Rg1 plays a neuroprotective role in brain repair following neonatal HI, and HIF-1α is a potential target for therapeutic intervention in neonates with HI brain injury.

    Topics: Animals; Animals, Newborn; Apoptosis; Brain Injuries; Caspase 3; Disease Models, Animal; Female; G(M1) Ganglioside; Ginsenosides; Hypoxia-Inducible Factor 1, alpha Subunit; Hypoxia-Ischemia, Brain; Male; Neovascularization, Physiologic; Neurons; Neuroprotective Agents; Rats; Rats, Sprague-Dawley; Signal Transduction; Vascular Endothelial Growth Factor A

2017
IgG-degrading enzyme of Streptococcus pyogenes (IdeS) prevents disease progression and facilitates improvement in a rabbit model of Guillain-Barré syndrome.
    Experimental neurology, 2017, Volume: 291

    Autoantibodies binding to peripheral nerves followed by complement deposition and membrane attack complex formation results in nerve damage in Guillain-Barré syndrome (GBS). Strategies to remove the pathogenic autoantibodies or block the complement deposition benefit most patients with GBS. Immunoglobulin G-degrading enzyme of Streptococcus pyogenes (IdeS) is a cysteine protease which cleaves IgG antibodies into F(ab')

    Topics: Animals; Autoantibodies; Bacterial Proteins; Complement C3; Disease Models, Animal; Disease Progression; Enzyme-Linked Immunosorbent Assay; G(M1) Ganglioside; Guillain-Barre Syndrome; Immunoglobulin G; Neural Conduction; Rabbits; Ranvier's Nodes; Sodium Channels; Statistics, Nonparametric; Time Factors

2017
Therapeutic effects of GM1 on Parkinson's disease in rats and its mechanism.
    The International journal of neuroscience, 2016, Volume: 126, Issue:2

    To observe the effects of GM1 on apomorphine (APO)-induced rotational behavior and the expression of inflammatory factors in 6-hydroxydopamine-induced Parkinson's disease (PD) rat models.. Mature and healthy Wistar rats of either sex with body weight of 150-200 g were randomly divided into control group, PD+APO group and PD+APO+GM1 group (10 mg/kg of GM1, intraperitoneally, once a day, for 14 days; each group with 15 rats). PD rat models were prepared by injecting 6-hydroxydopamine into rat's right striatum, and then rotational behavior was induced by intraperitoneal injection of APO 7 days after operation. Rat rotational behavior was observed, and mRNA and protein levels of interleukin-1β (IL-1β) and interleukin-1Ra (IL-1Ra) were determined, respectively, by RT-PCR and Western blot.. Compared with PD+APO group, the rotational behavior was significantly relieved in PD+APO+GM1 group (p < 0.05). Compared with control group, mRNA and protein expressions of IL-1β in the striatum significantly increased in PD+APO group (p < 0.05). However, mRNA and protein expressions of IL-1β significantly decreased in PD+APO+GM1 group compared with PD+APO group (p < 0.05), but mRNA and protein expressions of IL-1β were also higher in PD+APO+GM1 group than in control group (p < 0.05). mRNA and protein expressions of IL-1Ra in the striatum were significantly higher in PD+APO+GM1 group than in PD+APO group (p < 0.05).. GM1 can inhibit inflammatory reaction through decreasing mRNA and protein expressions of IL-1β and increasing mRNA and protein expressions of IL-1Ra with the therapeutic effects on PD.

    Topics: Adrenergic Agents; Animals; Antiparkinson Agents; Apomorphine; Corpus Striatum; Disease Models, Animal; Dopamine Agonists; Female; G(M1) Ganglioside; Gene Expression Regulation; Interleukin 1 Receptor Antagonist Protein; Interleukin-1beta; Male; Oxidopamine; Parkinson Disease; Rats; Rats, Wistar; RNA, Messenger; Stereotyped Behavior

2016
Protection against Experimental Stroke by Ganglioside GM1 Is Associated with the Inhibition of Autophagy.
    PloS one, 2016, Volume: 11, Issue:1

    Ganglioside GM1, which is particularly abundant in the central nervous system (CNS), is closely associated with the protection against several CNS disorders. However, controversial findings have been reported on the role of GM1 following ischemic stroke. In the present study, using a rat middle cerebral artery occlusion (MCAO) model, we investigated whether GM1 can protect against ischemic brain injury and whether it targets the autophagy pathway. GM1 was delivered to Sprague-Dawley male rats at 3 doses (25 mg/kg, 50 mg/kg, 100 mg/kg) by intraperitoneal injection soon after reperfusion and then once daily for 2 days. The same volume of saline was given as a control. Tat-Beclin-1, a specific autophagy inducer, was administered by intraperitoneal injection at 24 and 48 hours post-MCAO. Infarction volume, mortality and neurological function were assessed at 72 hours after ischemic insult. Immunofluorescence and Western blotting were performed to determine the expression of autophagy-related proteins P62, LC3 and Beclin-1 in the penumbra area. No significant changes in mortality and physiological variables (heart rate, blood glucose levels and arterial blood gases) were observed between the different groups. However, MCAO resulted in enhanced conversion of LC3-I into LC3-II, P62 degradation, high levels of Beclin-1, a large area infarction (26.3±3.6%) and serious neurobehavioral deficits. GM1 (50 mg/kg) treatment significantly reduced the autophagy activation, neurobehavioral dysfunctions, and infarction volume (from 26.3% to 19.5%) without causing significant adverse side effects. However, this biological function could be abolished by Tat-Beclin-1.. GM1 demonstrated safe and robust neuroprotective effects that are associated with the inhibition of autophagy following experimental stroke.

    Topics: Animals; Apoptosis Regulatory Proteins; Autophagy; Beclin-1; Brain; Brain Ischemia; Disease Models, Animal; Drug Administration Schedule; G(M1) Ganglioside; Gene Expression Regulation; Gene Products, tat; Heat-Shock Proteins; Infarction, Middle Cerebral Artery; Injections, Intraperitoneal; Male; Microtubule-Associated Proteins; Neuroprotective Agents; Rats; Rats, Sprague-Dawley; Recombinant Fusion Proteins; Sequestosome-1 Protein; Signal Transduction; Stroke; Survival Analysis

2016
GM1 ganglioside reverses the cognitive deficits induced by MK801 in mice.
    Behavioural pharmacology, 2016, Volume: 27, Issue:5

    Cognitive deficits are core symptoms of schizophrenia, but effective treatments are still lacking. Previous studies have reported that the brain-derived neurotrophic factor (BDNF) signaling is closely involved in learning and memory. Monosialotetrahexosylganglioside (GM1) is a ganglioside with wide-ranging pharmacologic effects that enhances the BDNF signaling cascade. This study aimed to assess the effects of GM1 on schizophrenia-related cognitive impairments. A brief disruption of N-methyl-D-aspartate receptors with MK801 was used to generate the animal model for cognitive deficits in schizophrenia. It was found that MK801-treated mice showed significant deficits in memory ability compared with control mice in different behavior tests, and this was accompanied by decreased hippocampal BDNF signaling pathway. Consecutive administration of GM1 fully restored the MK801-induced cognitive deficits and the impaired BDNF signaling in the hippocampus. Furthermore, a BDNF system inhibitor abolished the effects of GM1 in the MK801 model. Taken together, our results show that GM1 could reverse the MK801-induced cognitive deficits, suggesting a potential usefulness of GM1 in treating the schizophrenia-related cognitive impairments.

    Topics: Animals; Brain-Derived Neurotrophic Factor; Cognition Disorders; Disease Models, Animal; Dizocilpine Maleate; G(M1) Ganglioside; Hippocampus; Male; Memory Disorders; Mice; Mice, Inbred C57BL; Receptors, N-Methyl-D-Aspartate; Schizophrenia; Signal Transduction

2016
GDNF signaling implemented by GM1 ganglioside; failure in Parkinson's disease and GM1-deficient murine model.
    Experimental neurology, 2015, Volume: 263

    GDNF is indispensible for adult catecholaminergic neuron survival, and failure of GDNF signaling has been linked to loss of dopaminergic neurons in Parkinson's disease (PD). This study demonstrates attenuated GDNF signaling in neurons deficient in ganglio-series gangliosides, and restoration of such signaling with LIGA20, a membrane permeable analog of GM1. GM1 is shown to associate in situ with GFRα1 and RET, the protein components of the GDNF receptor, this being necessary for assembly of the tripartite receptor complex. Mice wholly or partially deficient in GM1 due to disruption of the B4galnt1 gene developed PD symptoms based on behavioral and neuropathological criteria which were largely ameliorated by gene therapy with AAV2-GDNF and also with LIGA20 treatment. The nigral neurons of PD subjects that were severely deficient in GM1 showed subnormal levels of tyrosine phosphorylated RET. Also in PD brain, GM1 levels in the occipital cortex, a region of limited PD pathology, were significantly below age-matched controls, suggesting the possibility of systemic GM1 deficiency as a risk factor in PD. This would accord with our finding that mice with partial GM1 deficiency represent a faithful recapitulation of the human disease. Together with the previously demonstrated age-related decline of GM1 in human brain, this points to gradual development of subthreshold levels of GM1 in the brain of PD subjects below that required for effective GDNF signaling. This hypothesis offers a dramatically different explanation for the etiology of sporadic PD as a manifestation of acquired resistance to GDNF.

    Topics: Animals; Cell Line; Disease Models, Animal; G(M1) Ganglioside; Gangliosides; Gene Knockdown Techniques; Glial Cell Line-Derived Neurotrophic Factor; Humans; Immunoblotting; Immunohistochemistry; Mice; Parkinson Disease; Signal Transduction; Sphingosine

2015
Diabetes mellitus accelerates Aβ pathology in brain accompanied by enhanced GAβ generation in nonhuman primates.
    PloS one, 2015, Volume: 10, Issue:2

    Growing evidence suggests that diabetes mellitus (DM) is one of the strongest risk factors for developing Alzheimer's disease (AD). However, it remains unclear why DM accelerates AD pathology. In cynomolgus monkeys older than 25 years, senile plaques (SPs) are spontaneously and consistently observed in their brains, and neurofibrillary tangles are present at 32 years of age and older. In laboratory-housed monkeys, obesity is occasionally observed and frequently leads to development of type 2 DM. In the present study, we performed histopathological and biochemical analyses of brain tissue in cynomolgus monkeys with type 2 DM to clarify the relationship between DM and AD pathology. Here, we provide the evidence that DM accelerates Aβ pathology in vivo in nonhuman primates who had not undergone any genetic manipulation. In DM-affected monkey brains, SPs were observed in frontal and temporal lobe cortices, even in monkeys younger than 20 years. Biochemical analyses of brain revealed that the amount of GM1-ganglioside-bound Aβ (GAβ)--the endogenous seed for Aβ fibril formation in the brain--was clearly elevated in DM-affected monkeys. Furthermore, the level of Rab GTPases was also significantly increased in the brains of adult monkeys with DM, almost to the same levels as in aged monkeys. Intraneuronal accumulation of enlarged endosomes was also observed in DM-affected monkeys, suggesting that exacerbated endocytic disturbance may underlie the acceleration of Aβ pathology due to DM.

    Topics: Age Factors; Alzheimer Disease; Amyloid beta-Peptides; Amyloidosis; Animals; Brain; Cathepsin D; Cerebral Amyloid Angiopathy; Diabetes Mellitus; Diabetes Mellitus, Type 2; Disease Models, Animal; Endocytosis; Female; G(M1) Ganglioside; Immunohistochemistry; Macaca fascicularis; Phagosomes; Plaque, Amyloid; rab GTP-Binding Proteins

2015
Suppressed rate of carcinogenesis and decreases in tumour volume and lung metastasis in CXCL14/BRAK transgenic mice.
    Scientific reports, 2015, Mar-13, Volume: 5

    Cancer progression involves carcinogenesis, an increase in tumour size, and metastasis. Here, we investigated the effect of overexpressed CXC chemokine ligand 14 (CXCL14) on these processes by using CXCL14/BRAK (CXCL14) transgenic (Tg) mice. The rate of AOM/DSS-induced colorectal carcinogenesis in these mice was significantly lower compared with that for isogenic wild type C57BL/6 (Wt) mice. When tumour cells were injected into these mice, the size of the tumours that developed and the number of metastatic nodules in the lungs of the animals were always significantly lower in the Tg mice than in the Wt ones. Injection of anti-asialo-GM1 antibodies to the mice before and after injection of tumour cells attenuated the suppressing effects of CXCL14 on the tumor growth and metastasis, suggesting that NK cell activity played an important role during CXCL14-mediated suppression of tumour growth and metastasis. The importance of NK cells on the metastasis was also supported when CXCL14 was expressed in B16 melanoma cells. Further, the survival rates after tumour cell injection were significantly increased for the Tg mice. As these Tg mice showed no obvious abnormality, we propose that CXCL14 to be a promising molecular target for cancer suppression/prevention.

    Topics: Animals; Antigens, Ly; Autoantibodies; Cell Transformation, Neoplastic; Chemokines, CXC; Chronic Disease; Colitis; Disease Models, Animal; Female; G(M1) Ganglioside; Galactosylceramides; Killer Cells, Natural; Lung Neoplasms; Lymphocyte Depletion; Melanoma, Experimental; Mice; Mice, Transgenic; Neoplasms; NK Cell Lectin-Like Receptor Subfamily B; Tumor Burden

2015
The Possible Damaged Mechanism and the Preventive Effect of Monosialotetrahexosylganglioside in a Rat Model of Cerebral Ischemia-Reperfusion Injury.
    Journal of stroke and cerebrovascular diseases : the official journal of National Stroke Association, 2015, Volume: 24, Issue:7

    The pathogenesis of cerebral ischemia-reperfusion injury (CIRI) is not completely clear and therapies are limited now. Therefore, our study aimed to investigate the possible pathogenesis and preventive approach of CIRI through analyzing changes of aspartate (Asp), glutamate (Glu), mitochondrial calcium (MCa), calmodulin (CaM), and malondialdehyde (MDA) contents and ultramicropathology in hippocampus and cerebral cortex of ischemic susceptible injured regions and the effect of monosialotetrahexosylganglioside (GM1) in the rat model of CIRI.. Contents of Asp, Glu, MCa, CaM, and MDA in hippocampus and cerebral cortex tissues were measured by a high-performance liquid chromatography, atomic absorption spectrophotometer, and ordinary spectrophotometer, respectively, changes of ultramicrostructure in neurons of the hippocampus CA1 region and frontal cerebral cortex were observed by a transmission electron microscope.. Contents of Asp and Glu in hippocampus and cerebral cortex tissues of CIRI groups significantly decreased and contents of MCa, CaM, and MDA significantly increased than those in control groups, and the ultramicrostructure in neurons of the hippocampus CA1 region and frontal cerebral cortex revealed a significant damaged change, and GM1 significantly ameliorated changes of Asp, Glu, MCa, CaM, and MDA contents in hippocampus and ultrastructural changes in neurons of the hippocampus CA1 region and frontal cerebral cortex.. Our findings further support that the abnormal release and/or reuptake of excitatory amino acid neurotransmitters, the disordered calcium homeostasis and the excessive production and/or reduced elimination of reactive oxygen species contribute to the pathogenesis of CIRI, and GM1 can partially prevent these pathogenesises to exert the protective effect on CIRI.

    Topics: Animals; Biomarkers; Brain Ischemia; Calcium; Cerebral Cortex; Cytoprotection; Disease Models, Animal; Excitatory Amino Acids; G(M1) Ganglioside; Hippocampus; Homeostasis; Male; Neurons; Neuroprotective Agents; Oxidative Stress; Rats, Wistar; Reactive Oxygen Species; Reperfusion Injury; Time Factors

2015
GM1-Modified Lipoprotein-like Nanoparticle: Multifunctional Nanoplatform for the Combination Therapy of Alzheimer's Disease.
    ACS nano, 2015, Nov-24, Volume: 9, Issue:11

    Alzheimer's disease (AD) exerts a heavy health burden for modern society and has a complicated pathological background. The accumulation of extracellular β-amyloid (Aβ) is crucial in AD pathogenesis, and Aβ-initiated secondary pathological processes could independently lead to neuronal degeneration and pathogenesis in AD. Thus, the development of combination therapeutics that can not only accelerate Aβ clearance but also simultaneously protect neurons or inhibit other subsequent pathological cascade represents a promising strategy for AD intervention. Here, we designed a nanostructure, monosialotetrahexosylganglioside (GM1)-modified reconstituted high density lipoprotein (GM1-rHDL), that possesses antibody-like high binding affinity to Aβ, facilitates Aβ degradation by microglia, and Aβ efflux across the blood-brain barrier (BBB), displays high brain biodistribution efficiency following intranasal administration, and simultaneously allows the efficient loading of a neuroprotective peptide, NAP, as a nanoparticulate drug delivery system for the combination therapy of AD. The resulting multifunctional nanostructure, αNAP-GM1-rHDL, was found to be able to protect neurons from Aβ(1-42) oligomer/glutamic acid-induced cell toxicity better than GM1-rHDL in vitro and reduced Aβ deposition, ameliorated neurologic changes, and rescued memory loss more efficiently than both αNAP solution and GM1-rHDL in AD model mice following intranasal administration with no observable cytotoxicity noted. Taken together, this work presents direct experimental evidence of the rational design of a biomimetic nanostructure to serve as a safe and efficient multifunctional nanoplatform for the combination therapy of AD.

    Topics: Administration, Intranasal; Alzheimer Disease; Amino Acid Sequence; Amyloid beta-Peptides; Animals; Apolipoproteins E; Brain; Combined Modality Therapy; Disease Models, Animal; Endocytosis; G(M1) Ganglioside; Glutamates; Lipoproteins, HDL; Memory Disorders; Mice, Inbred ICR; Microglia; Molecular Sequence Data; Nanoparticles; Neuroprotection; Neuroprotective Agents; Particle Size; Peptide Fragments; Peptides; Rats, Sprague-Dawley; Static Electricity; Tissue Distribution

2015
Intraventricular Sialidase Administration Enhances GM1 Ganglioside Expression and Is Partially Neuroprotective in a Mouse Model of Parkinson's Disease.
    PloS one, 2015, Volume: 10, Issue:12

    Preclinical and clinical studies have previously shown that systemic administration of GM1 ganglioside has neuroprotective and neurorestorative properties in Parkinson's disease (PD) models and in PD patients. However, the clinical development of GM1 for PD has been hampered by its animal origin (GM1 used in previous studies was extracted from bovine brains), limited bioavailability, and limited blood brain barrier penetrance following systemic administration.. To assess an alternative therapeutic approach to systemic administration of brain-derived GM1 to enhance GM1 levels in the brain via enzymatic conversion of polysialogangliosides into GM1 and to assess the neuroprotective potential of this approach.. We used sialidase from Vibrio cholerae (VCS) to convert GD1a, GD1b and GT1b gangliosides to GM1. VCS was infused by osmotic minipump into the dorsal third ventricle in mice over a 4-week period. After the first week of infusion, animals received MPTP injections (20 mg/kg, s.c., twice daily, 4 hours apart, for 5 consecutive days) and were euthanized 2 weeks after the last injection.. VCS infusion resulted in the expected change in ganglioside expression with a significant increase in GM1 levels. VCS-treated animals showed significant sparing of striatal dopamine (DA) levels and substantia nigra DA neurons following MPTP administration, with the extent of sparing of DA neurons similar to that achieved with systemic GM1 administration.. The results suggest that enzymatic conversion of polysialogangliosides to GM1 may be a viable treatment strategy for increasing GM1 levels in the brain and exerting a neuroprotective effect on the damaged nigrostriatal DA system.

    Topics: Animals; Disease Models, Animal; Dopamine; G(M1) Ganglioside; Gene Expression Regulation; Infusions, Intraventricular; Male; Mice; Mice, Inbred C57BL; Neuraminidase; Neurons; Neuroprotective Agents; Parkinson Disease; Pars Compacta; Vibrio cholerae

2015
The effect of heme oxygenase on ganglioside redistribution within hepatocytes in experimental estrogen-induced cholestasis.
    Physiological research, 2014, Volume: 63, Issue:3

    Cholestasis is characterized by the elevation of serum total bile acids (TBA), which leads to the production of both free radicals and oxidative stress. Although they do not share the same mechanisms, membrane glycosphingolipids (GSL) and the antioxidant enzyme heme oxygenase-1 (HMOX1) both act against the pro-oxidative effect of TBA. The aim of the study was to assess the role of HMOX on GSL redistribution and composition within hepatocytes in the rat model of estrogen-induced cholestasis. Compared to the controls, an increase of total gangliosides in the liver homogenates of the cholestatic group (P=0.001) was detected; further, it paralleled along with the activation of their biosynthetic b-branch pathway (P<0.01). These effects were partially prevented by HMOX activation. Cholestasis was accompanied by a redistribution of GM1 ganglioside from the cytoplasm to the sinusoids; while HMOX activation led to the retention of GM1 in the cytoplasm (P=0.014). Our study shows that estrogen-induced cholestasis is followed by changes in the synthesis and/or distribution of GSL. These changes are not only triggered by the detergent power of accumulated TBA, but also by their pro-oxidant action. Increases in the antioxidant defenses might represent an important supportive therapeutic measure for patients with cholestatic liver disease.

    Topics: Animals; Antioxidants; Cholestasis; Disease Models, Animal; Enzyme Activation; Ethinyl Estradiol; Female; G(M1) Ganglioside; Heme Oxygenase (Decyclizing); Hepatocytes; Rats, Wistar

2014
Propofol and thiopental suppress amyloid fibril formation and GM1 ganglioside expression through the γ-aminobutyric acid A receptor.
    Anesthesiology, 2013, Volume: 118, Issue:6

    The incidence of Alzheimer disease may increase after surgical interventions. Amyloid β-protein (Aβ) fibrillogenesis, which is closely related to Alzheimer disease, is reportedly accelerated by exposure to anesthetics. However, the effects of GM1 ganglioside (GM1) on Αβ fibrillogenesis have not yet been reported. The current study was designed to examine whether the anesthetics propofol and thiopental are associated with Αβ assembly and GM1 expression on the neuronal cell surface.. PC12N cells and cultured neuronal cells were treated with propofol or thiopental, and GM1 expression in treated and untreated cells was determined by the specific binding of horseradish peroxidase-conjugated cholera toxin subunit B (n = 5). The effects of an inhibitor of the γ-aminobutyric acid A receptor was also examined (n= 5). In addition, the effects of the anesthetics on GM1 liposome-induced Αβ assembly were investigated (n = 5). Finally, the neurotoxicity of the assembled Αβ fibrils was studied by the lactate dehydrogenase release assay (n = 6).. Propofol (31.2 ± 4.7%) and thiopental (34.6 ± 10.5%) decreased GM1 expression on the cell surface through the γ-aminobutyric acid A receptor. The anesthetics inhibited Αβ fibril formation from soluble Αβ in cultured neurons. Moreover, propofol and thiopental suppressed GM1-induced fibril formation in a cell-free system (propofol, 75.8 ± 1.9%; thiopental, 83.6 ± 1.9%) and reduced the neurotoxicity of a mixture containing Aβ and GM1 liposomes (propofol, 35.3 ± 16.4%; thiopental, 21.3 ± 11.6%).. Propofol and thiopental have direct and indirect inhibitory effects on Αβ fibrillogenesis.

    Topics: Amyloid; Anesthetics, Intravenous; Animals; Cell Membrane; Cell-Free System; Cells, Cultured; Disease Models, Animal; G(M1) Ganglioside; Male; Mice; Neurons; PC12 Cells; Propofol; Receptors, GABA; Thiopental

2013
Alterations of membrane lipids and in gene expression of ganglioside metabolism in different brain structures in a mouse model of mucopolysaccharidosis type I (MPS I).
    Gene, 2013, Sep-15, Volume: 527, Issue:1

    Mucopolysaccharidosis I (MPS I) is a congenital disorder caused by the deficiency of α-l-iduronidase (IDUA), with the accumulation of glycosaminoglycans (GAGs) in the CNS. Although GAG toxicity is not fully understood, previous works suggest a GAG-induced alteration in neuronal membrane composition. This study is aimed to evaluate the levels and distribution of gangliosides and cholesterol in different brain regions (cortex, cerebellum, hippocampus and hypothalamus) in a model using IDUA knockout (KO) mice (C57BL/6). Lipids were extracted with chloroform-methanol and then total gangliosides and cholesterol were determined, followed by ganglioside profile analyses. While no changes in cholesterol content were observed, the results showed a tissue dependent ganglioside alteration in KO mice: a total ganglioside increase in cortex and cerebellum, and a selective presence of GM3, GM2 and GD3 gangliosides in the hippocampus and hypothalamus. To elucidate this, we evaluated gene expression of ganglioside synthesis (GM3, GD3 and GM2/GD2 synthases) and degradation of (Neuraminidase1) enzymes in the cerebellum and hippocampus by RT-sq-PCR. The results obtained with KO mice showed a reduced expression of GD3 and GM2/GD2 synthases and Neuraminidase1 in cerebellum; and a decrease in GM2/GD2 synthase and Neuraminidase1 in the hippocampus. These data suggest that the observed ganglioside changes result from a combined effect of GAGs on ganglioside biosynthesis and degradation.

    Topics: Animals; Brain; Cerebellum; Cerebral Cortex; Cholesterol; Disease Models, Animal; G(M1) Ganglioside; Gene Expression; Hippocampus; Hypothalamus; Membrane Lipids; Mice; Mice, Inbred C57BL; Mice, Knockout; Mucopolysaccharidosis I; N-Acetylgalactosaminyltransferases; Neuraminidase; Sialyltransferases

2013
Ganglioside GM1 reduces white matter damage in neonatal rats.
    Acta neurobiologiae experimentalis, 2013, Volume: 73, Issue:3

    This study investigated the neuronal protective effect of monosialotetrahexosylganglioside (GM1) on the hypoxia-ischemia white matter damage (WMD) in neonatal rats. Brain hypoxia-ischemia was induced by bilateral carotid artery occlusion in 4-day-old neonatal rats. Bilateral carotid artery occlusion (BCAO) was performed in rats in WMD and GM1 groups, while in sham group; the rat bilateral carotid arteries were merely exposed without occlusion. Immunohistochemical staining was used to determine the expression of myelin basic protein (MBP), glial fibrillary acidic protein (GFAP), and beta-amyloid precursor protein (beta-APP). In addition, suspension test, slope test, and open-field test were carried out on day 26 after BCAO to determine the neurobehavioral function. The percentage of MBP-positive cells was decreased while beta-APP-and GFAPpositive cells were increased in WMD group. After treated with GM1, the percentage of MBP-positive cells increased significantly than WMD rats at post-operation 72 h and day 7. GFAP-positive cells and beta-APP-positive cells decreased significantly in WMD group at post-operation 72 h, day 7 and 26. The suspension test, slope test, and open-field test showed that neurobehavioral function was improved in ganglioside GM1 group compared with WMD group. Taken together, our findings suggested that ganglioside GM1 treatment reduces hypoxia-ischemia induced impairment of the neurobehavioral function in WMD in neonatal rats.

    Topics: Amyloid beta-Protein Precursor; Animals; Animals, Newborn; Carotid Artery Diseases; Disease Models, Animal; Female; G(M1) Ganglioside; Gene Expression Regulation; Glial Fibrillary Acidic Protein; Leukoencephalopathies; Male; Myelin Basic Protein; Neurons; Rats; Rats, Sprague-Dawley; Time Factors

2013
Fuzheng Huayu inhibits carbon tetrachloride-induced liver fibrosis in mice through activating hepatic NK cells.
    Journal of ethnopharmacology, 2013, Jan-09, Volume: 145, Issue:1

    Fuzheng Huayu (FZHY) is a Chinese compound herbal preparation which consists of six Chinese herbs. This study examines the preventative effects of FZHY on liver fibrosis induced by carbon tetrachloride (CCl(4)) and explores its possible mechanisms of action.. Liver fibrosis was induced in male C57BL/6N mice by injecting a 10% CCl(4) solution intraperitoneal twice a week for six weeks. After 6 weeks of treatment, serum ALT and AST assay, liver tissue histological examination and immunostaining were carried out to examine the liver function and fibrosis degree. The expression levels of alpha-smooth muscle actin (SMA) were measured by quantitative real-time PCR and western blot. Hepatic natural killer (NK) cells were isolated from liver and evaluated by FACS.. Upon pathological examination, the FZHY-treated mice showed significantly reduced liver damage. The expression of α-SMA increased markedly upon treatment with CCl(4) and the increase was reversed by FZHY treatment. FZHY treatment also enhanced the activation of hepatic NK cells and the production of interferon-gamma (IFN-γ). The protective effects of FZHY were reversed in the mice that were depleted of NK cells by anti-ASGM-1 Ab treatment.. FZHY can efficiently inhibit CCl(4)-induced liver fibrosis. Furthermore, the depletion of NK cells attenuates the protective effects of FZHY. We conclude that FZHY could be an effective drug for liver fibrosis, and its mechanism of action involves the activation of hepatic NK cells.

    Topics: Actins; Alanine Transaminase; Animals; Aspartate Aminotransferases; Autoantibodies; Carbon Tetrachloride; Disease Models, Animal; Drugs, Chinese Herbal; G(M1) Ganglioside; Interferon-gamma; Killer Cells, Natural; Liver; Liver Cirrhosis; Male; Mice; Mice, Inbred C57BL

2013
Monosialoanglioside improves memory deficits and relieves oxidative stress in the hippocampus of rat model of Alzheimer's disease.
    Neurological sciences : official journal of the Italian Neurological Society and of the Italian Society of Clinical Neurophysiology, 2013, Volume: 34, Issue:8

    GM-1 ganglioside (GM-1) has been proposed as a new therapeutic agent against Alzheimer's disease (AD). Therefore, in this study we aimed to investigate the effects of GM1 on memory deficits and oxidative stress in the hippocampus of rat model of AD. Wistar rats were randomly divided into three groups (n = 15): control group, model group, and treatment group, which were injected with vehicle, Aβ1-40, and Aβ1-40 together with GM-1, respectively. Morris water maze test was performed to evaluate spatial learning and memory of the rats. Brain malondialdehyde (MDA) content was detected by biochemical assay, and 4-hydroxynonenal (4-HNE) level in the hippocampus was examined by immunohistochemistry. The results showed that learning and memory deficits were improved in treatment group compared to model group. Brain MDA content and 4-HNE level in hippocampus CA1 were much lower in treatment group than in model group. In summary, we demonstrate that GM-1 could improve spatial learning and memory deficits in rat model of AD, and this may be mediated by the inhibition of oxidative stress and lipid peroxidation in the neurons. These data suggest that GM-1 is a potential agent for AD treatment.

    Topics: Aldehydes; Alzheimer Disease; Animals; Disease Models, Animal; G(M1) Ganglioside; Hippocampus; Malondialdehyde; Maze Learning; Memory Disorders; Oxidative Stress; Rats; Rats, Wistar

2013
Anti-ganglioside antibody internalization attenuates motor nerve terminal injury in a mouse model of acute motor axonal neuropathy.
    The Journal of clinical investigation, 2012, Volume: 122, Issue:3

    In the Guillain-Barré syndrome subform acute motor axonal neuropathy (AMAN), Campylobacter jejuni enteritis triggers the production of anti-ganglioside Abs (AGAbs), leading to immune-mediated injury of distal motor nerves. An important question has been whether injury to the presynaptic neuron at the neuromuscular junction is a major factor in AMAN. Although disease modeling in mice exposed to AGAbs indicates that complement-mediated necrosis occurs extensively in the presynaptic axons, evidence in humans is more limited, in comparison to the extensive injury seen at nodes of Ranvier. We considered that rapid AGAb uptake at the motor nerve terminal membrane might attenuate complement-mediated injury. We found that PC12 rat neuronal cells rapidly internalized AGAb, which were trafficked to recycling endosomes and lysosomes. Consequently, complement-mediated cytotoxicity was attenuated. Importantly, we observed the same AGAb endocytosis and protection from cytotoxicity in live mouse nerve terminals. AGAb uptake was attenuated following membrane cholesterol depletion in vitro and ex vivo, indicating that this process may be dependent upon cholesterol-enriched microdomains. In contrast, we observed minimal AGAb uptake at nodes of Ranvier, and this structure thus remained vulnerable to complement-mediated injury. These results indicate that differential endocytic processing of AGAbs by different neuronal and glial membranes might be an important modulator of site-specific injury in acute AGAb-mediated Guillain-Barré syndrome subforms and their chronic counterparts.

    Topics: Animals; Antibodies, Anti-Idiotypic; Complement Activation; Disease Models, Animal; Endocytosis; Female; G(M1) Ganglioside; Gangliosides; Guillain-Barre Syndrome; Male; Mice; Mice, Inbred C57BL; Motor Neurons; Neuromuscular Junction; PC12 Cells; Ranvier's Nodes; Rats

2012
Ganglioside GM1 induces phosphorylation of mutant huntingtin and restores normal motor behavior in Huntington disease mice.
    Proceedings of the National Academy of Sciences of the United States of America, 2012, Feb-28, Volume: 109, Issue:9

    Huntington disease (HD) is a progressive neurodegenerative monogenic disorder caused by expansion of a polyglutamine stretch in the huntingtin (Htt) protein. Mutant huntingtin triggers neural dysfunction and death, mainly in the corpus striatum and cerebral cortex, resulting in pathognomonic motor symptoms, as well as cognitive and psychiatric decline. Currently, there is no effective treatment for HD. We report that intraventricular infusion of ganglioside GM1 induces phosphorylation of mutant huntingtin at specific serine amino acid residues that attenuate huntingtin toxicity, and restores normal motor function in already symptomatic HD mice. Thus, our studies have identified a potential therapy for HD that targets a posttranslational modification of mutant huntingtin with critical effects on disease pathogenesis.

    Topics: Animals; Codon; Corpus Striatum; Dimerization; Disease Models, Animal; Dopamine and cAMP-Regulated Phosphoprotein 32; Drug Evaluation, Preclinical; G(M1) Ganglioside; Huntingtin Protein; Infusion Pumps, Implantable; Infusions, Parenteral; Mice; Mice, Neurologic Mutants; Motor Activity; Nerve Tissue Proteins; Neurons; Nuclear Proteins; Phosphorylation; Phosphoserine; Protein Processing, Post-Translational; Psychomotor Performance

2012
Potentializing the effects of GM1 by hyperbaric oxygen therapy in acute experimental spinal cord lesion in rats.
    Spinal cord, 2010, Volume: 48, Issue:11

    Experimental, controlled, animal study.. To evaluate the effect of GM1 ganglioside, hyperbaric oxygen and both in combination, in the treatment of experimental spinal cord lesions in rats.. Brazil.. Thirty-two Wistar rats with spinal cord lesions were divided into four groups: one group received GM1 ganglioside, one was submitted to hyperbaric oxygen therapy (HBOT), the third received both treatments and the fourth received no treatment (control).. There were no significant differences between the groups in the histological analysis, for any of the variables (necrosis, hemorrhage, hyperemia, cystic degeneration, P>0.06). Neither were there any significant differences in the comparison of left and right sides in the functional tests (P>0.06 for all). No significant differences were found in the locomotor ratings, in the comparison of groups at 2, 7, 21 and 28 days after the surgical procedure. However, in the evaluation on day 14, group 3, which received the combined therapy, showed a significantly higher Basso Beattie and Bresnahan score than the other groups (P=0.015).. The therapeutic effect of GM1 in locomotor evaluation of rats submitted to spinal cord lesion is anticipated by HBOT.

    Topics: Acute Disease; Animals; Combined Modality Therapy; Disease Models, Animal; G(M1) Ganglioside; Hyperbaric Oxygenation; Male; Nerve Regeneration; Neuroprotective Agents; Rats; Rats, Wistar; Spinal Cord Injuries

2010
Monosialotetrahexosy-1 ganglioside attenuates diabetes-enhanced brain damage after transient forebrain ischemia and suppresses phosphorylation of ERK1/2 in the rat brain.
    Brain research, 2010, Jul-16, Volume: 1344

    Monosialotetrahexosy-1 ganglioside (GM1) has been shown to reduce brain damage induced by cerebral ischemia. The objective of this study is to determine whether GM1 is able to ameliorate hyperglycemia-exacerbated ischemic brain damage in hyperglycemia-recruited areas such as the hippocampal CA3 sub regions and the cingulated cortex. Histologic stainings of Haematoxylin and Eosin, Nissl body, the terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling (TUNEL) and phospho-ERK1/2 were performed on brain sections that have been subjected to 15 min of forebrain ischemia with reperfusion of 0, 1, 3, and 6h in normoglycemic, hyperglycemic and GM1-pretreated hyperglycemic groups. The results showed that GM1 ameliorated ischemic neuronal injuries in the CA3 area and cingulated cortex of the hyperglycemic animals after ischemia and reperfusion. Immunohistochemistry of phospho-ERK1/2 revealed that the neuroprotective effects of GM1 were associated with suppression of phospho-ERK1/2. The results suggest that GM1 attenuates diabetic-augmented ischemic neuronal injuries probably through suppression of ERK1/2 phosphorylation.

    Topics: Animals; Diabetes Mellitus, Experimental; Disease Models, Animal; G(M1) Ganglioside; Hippocampus; Hypoxia, Brain; In Situ Nick-End Labeling; Male; Mitogen-Activated Protein Kinase 3; Nerve Degeneration; Neurons; Neuroprotective Agents; Phosphorylation; Rats; Rats, Sprague-Dawley; Signal Transduction; Streptozocin; Time Factors

2010
The behavioral and immunological effect of GM-1 ganglioside on nerve root regeneration following C5 nerve root avulsion in a rat model.
    Journal of reconstructive microsurgery, 2009, Volume: 25, Issue:2

    Preganglionic nerve root avulsion precludes sensory return, but motor regeneration is possible with sparing of motoneurons. The effect of GM-1 ganglioside treatment was studied with parallel evaluation of the autoimmune response. Rats (N=64) received injections of either GM-1 ganglioside or saline for 30 days following either C5 root avulsion or a hemilaminectomy control. The Bertelli grooming test assessed functional return. Before sacrifice at 5 months, serum was collected for enzyme-linked immunoabsorbent assay testing. Only 44% of the rats treated with ganglioside had a good functional outcome compared with 50% for controls. Although 17% of the rats developed anti GM-1 antibodies, there was no functional or histological evidence of neuropathy in any of the rats. We conclude that ganglioside treatment did not enhance recovery from peripheral nerve injury. Although an immune response was present in some rats, no overt signs of neuropathy were observed.

    Topics: Animals; Cervical Vertebrae; Disease Models, Animal; Enzyme-Linked Immunosorbent Assay; G(M1) Ganglioside; Male; Nerve Regeneration; Radiculopathy; Rats; Rats, Sprague-Dawley; Recovery of Function

2009
Prevention of embryo loss in non-obese diabetic mice using adoptive ITGA2(+)ISG20(+) natural killer-cell transfer.
    Reproduction (Cambridge, England), 2009, Volume: 137, Issue:6

    Both regulatory T cells and regulatory natural killer (NK) cells may play essential roles in the maintenance of pregnancy. In this study, we show that a significantly high percentage of spontaneous embryo loss was observed in both allogeneic and syngeneic pregnant non-obese diabetic (NOD) mice. The percentage of embryo loss in allogeneic pregnant mice was further increased by the administration of anti-asialo ganglio-N-tetraosylceramide to deplete NK cells, but was decreased by the adoptive transfer of ITGA2(+)ISG20(+) (CD49b(+) CD25(+)) NK cells from normal mice. No such trend was observed in syngeneic pregnant NOD mice. The pattern of CXCR4 (specific receptor for CXCL12) expression on NK cells was analyzed and NK-cell migration was confirmed by in vitro and in vivo migratory assays. Since CXCL12 production by murine trophoblast cells was confirmed previously, our findings suggest that the recruitment of peripheral CXCR4-expressing ITGA2(+)ISG20(+) NK cells into pregnant uteri may be important in the regulation of feto-maternal tolerance.

    Topics: Adoptive Transfer; Animals; Cells, Cultured; Chemotaxis, Leukocyte; Diabetes Complications; Disease Models, Animal; Embryo Loss; Female; Forkhead Transcription Factors; G(M1) Ganglioside; Immune Tolerance; Integrin alpha2; Interleukin-2 Receptor alpha Subunit; Killer Cells, Natural; Male; Mice; Mice, Inbred BALB C; Mice, Inbred C57BL; Mice, Inbred NOD; Placenta; Pregnancy; Receptors, CXCR4; Transplantation, Homologous; Transplantation, Isogeneic

2009
Cloning, expression, purification and characterization of the cholera toxin B subunit and triple glutamic acid decarboxylase epitopes fusion protein in Escherichia coli.
    Protein expression and purification, 2009, Volume: 66, Issue:2

    Induction of specific immunological unresponsiveness by oral autoantigens such as glutamic acid decarboxylase 65 (GAD65) is termed oral tolerance and may be a potential therapy for autoimmune diabetes. However, the requirement for large amounts of protein will limit clinical testing of autoantigens, which are difficult to produce. Mucosal adjuvants such as cholera toxin B subunit (CTB) may lower the level of autoantigens required. Here we describe cloning, expression, purification and identification study of the CTB and triple GAD(531-545) epitopes fusion gene. The fusion gene was ligated via a flexible hinge tetrapeptide and expressed as a soluble protein in Escherichia coli BL21 (DE3) driven by the T7 promoter. We purified the recombination protein from the cell lysate and obtained approximately 2.5mg of CTB-GAD((531-545)3) per liter of culture with greater than 90% purity by a Ni-NTA resin column. The bacteria produced this protein as the pentameric form, which retained the GM1-ganglioside binding affinity and the native antigenicity of CTB and GAD65. Further studies revealed that oral administration of bacterial CTB-GAD((531-545)3) fusion protein showed the prominent reduction in pancreatic islet inflammation in non-obese diabetic mice. The results presented here demonstrate that the bacteria bioreactor is an ideal production system for an oral protein vaccine designed to develop immunological tolerance against autoimmune diabetes.

    Topics: Administration, Oral; Amino Acid Sequence; Animals; Autoantigens; Base Sequence; Bioreactors; Cholera Toxin; Chromatography, Affinity; Disease Models, Animal; Escherichia coli; Female; G(M1) Ganglioside; Glutamate Decarboxylase; Histocytochemistry; Immune Tolerance; Islets of Langerhans; Mice; Mice, Inbred NOD; Molecular Sequence Data; Recombinant Fusion Proteins

2009
GM1-ganglioside accumulation at the mitochondria-associated ER membranes links ER stress to Ca(2+)-dependent mitochondrial apoptosis.
    Molecular cell, 2009, Nov-13, Volume: 36, Issue:3

    Mitochondria-associated ER membranes, or MAMs, define the sites of endoplasmic reticulum/mitochondria juxtaposition that control Ca(2+) flux between these organelles. We found that in a mouse model of the human lysosomal storage disease GM1-gangliosidosis, GM1-ganglioside accumulates in the glycosphingolipid-enriched microdomain (GEM) fractions of MAMs, where it interacts with the phosphorylated form of IP3 receptor-1, influencing the activity of this channel. Ca(2+) depleted from the ER is then taken up by the mitochondria, leading to Ca(2+) overload in this organelle. The latter induces mitochondrial membrane permeabilization (MMP), opening of the permeability transition pore, and activation of the mitochondrial apoptotic pathway. This study identifies the GEMs as the sites of Ca(2+) diffusion between the ER and the mitochondria. We propose a new mechanism of Ca(2+)-mediated apoptotic signaling whereby GM1 accumulation at the GEMs alters Ca(2+) dynamics and acts as a molecular effector of both ER stress-induced and mitochondria-mediated apoptosis of neuronal cells.

    Topics: Adenosine Triphosphate; Animals; Apoptosis; beta-Galactosidase; Brain; Calcium; Cells, Cultured; Cytochromes c; Disease Models, Animal; Endoplasmic Reticulum; Fibroblasts; G(M1) Ganglioside; Gangliosidosis, GM1; Glycosphingolipids; Humans; Immunoblotting; Inositol 1,4,5-Trisphosphate Receptors; Membrane Microdomains; Membrane Potential, Mitochondrial; Mice; Mice, Knockout; Microscopy, Electron, Transmission; Mitochondria; Reactive Oxygen Species

2009
Effects of methylprednisolone and ganglioside GM-1 on a spinal lesion: a functional analysis.
    Clinics (Sao Paulo, Brazil), 2008, Volume: 63, Issue:3

    The pharmacological effects of methylprednisolone (MP) and ganglioside GM-1 on spinal injuries have been thoroughly investigated, but only a few studies have evaluated the interaction between these two drugs.. Twenty-four Wistar rats were subjected to contusive injury of the spinal cord produced by the NYU system. These animals were divided into four groups: group I was injected with MP; group II was injected with GM-1; group III was injected with MP together with GM-1; and group control received physiological serum. The animals were evaluated with regard to their recovery of locomotive function by means of the BBB test on the second, seventh and fourteenth days after receiving the contusive injury to the spinal cord. They were sacrificed on the fourteenth day.. This study demonstrated that the MP and GM-1 groups presented functional results that were better than those of the control group, although the enhanced recovery of group II (GM-1) relative to the control group was not statistically significant (p>0.05). The most notable recovery of locomotive function was observed in the group that received MP alone (p<0.05). The group that received MP together with GM-1 presented results that were better than those of the control group (p<0.05).. Administration of methylprednisolone alone or with GM-1 was shown to be effective for recovery of locomotive function. Combined administration of these drugs resulted in better outcomes than administration of methylprednisolone alone.

    Topics: Animals; Anti-Inflammatory Agents; Disease Models, Animal; Drug Evaluation, Preclinical; Drug Therapy, Combination; G(M1) Ganglioside; Male; Methylprednisolone; Motor Activity; Rats; Rats, Wistar; Recovery of Function; Spinal Cord Injuries; Statistics, Nonparametric; Treatment Outcome

2008
Renal gangliosides are involved in lead intoxication.
    Journal of applied toxicology : JAT, 2008, Volume: 28, Issue:2

    The biological effects of lead are well defined; however, neither the risk exposure level nor the subcellular mechanism of its action is completely clear. The present work was undertaken to investigate the effects of low level and long term lead exposure on the composition and expression of rat renal gangliosides. In order to identify ganglioside expression, frozen sections of kidneys were stained with monoclonal antibodies GMB16 (GM1 specific), GM28 (GM2 specific), AMR-10 (GM4 specific) and CDW 60 (9-O-Ac-GD3 specific). Strong reactivity was observed for GMB28, AMR-10 and CDW 60, while GMB16 developed only weak labelling in treated kidney compared with the control. The alterations in the expression of renal gangliosides observed by immunohistochemistry were accompanied by quantitative and qualitative changes in the thin layer chromatography of total gangliosides isolated from kidney tissues. Lead treatment produced a significant increase in 9-O-Ac GD3, a ganglioside involved in apoptotic processes. In agreement with this result, a significant decrease in the number of apoptotic glomerular cells was observed with the TUNEL assay. These findings lead us to suggest that alterations in renal gangliosides produced by low level lead exposure are associated with the apoptotic processes that take place in the kidney. These findings provide evidence that low level and long term lead exposure produces renal ganglioside alterations with urinary microalbumin excretion. The results suggest that lead levels within the limits of biological tolerance already cause molecular renal damage without clinical signs of toxicity.

    Topics: Albuminuria; Animals; Apoptosis; Body Weight; Chromatography, Thin Layer; Disease Models, Animal; Eating; G(M1) Ganglioside; G(M2) Ganglioside; Gangliosides; Immunohistochemistry; In Situ Nick-End Labeling; Kidney; Kidney Diseases; Lead Poisoning; Male; Organometallic Compounds; Rats; Rats, Wistar; Time Factors

2008
Natural killer cells are required for accelerated type 1 diabetes driven by interferon-beta.
    Clinical and experimental immunology, 2008, Volume: 151, Issue:3

    The destruction of beta cells by the islet infiltrating lymphocytes causes type 1 diabetes. Transgenic mice models expressing interferon (IFN)-beta in beta cells, in the non-obese diabetic (NOD) strain and in a diabetes-free, major histocompatibility complex-matched, homologous strain, the non-obese resistant (NOR) mice, developed accelerated type 1 diabetes after 3 weeks of age. Our aim was to determine if natural killer (NK) cells could affect the acceleration of the disease. We determined the amount of NK cells in the pancreas, spleen and lymph nodes from NOD rat insulin promoter (RIP)-IFN-beta mice. Pancreatic cytokines were assessed by quantitative real-time polymerase chain reaction and protein arrays. To confirm the relevance of NK cells in the acceleration of autoimmune diabetes this subset was depleted with anti-asialo GM1 antibodies. An increase of intrapancreatic NK cells characterized the accelerated onset of diabetes both in NOD and NOR RIP-IFN-beta transgenic models. Cytokines involved in NK function and migration were found to be hyperexpressed in the pancreas from accelerated diabetic mice. Interestingly, the depletion of NK cells in vivo abolished completely the acceleration of diabetes. NK cells connect innate to adaptive immunity and might play a role in autoimmunity. We report here that NK cells are required critically in the pancreas for accelerated diabetes. This model links inflammation to acceleration of beta cell-specific autoimmunity mediated by NK cells.

    Topics: Animals; B-Lymphocytes; Cytokines; Diabetes Mellitus, Type 1; Disease Models, Animal; Disease Progression; G(M1) Ganglioside; Interferon-beta; Islets of Langerhans; Killer Cells, Natural; Lymph Nodes; Lymphocyte Subsets; Mice; Mice, Inbred NOD

2008
Enhanced autophagy and mitochondrial aberrations in murine G(M1)-gangliosidosis.
    Biochemical and biophysical research communications, 2008, Mar-14, Volume: 367, Issue:3

    G(M1)-gangliosidosis is an autosomal recessive lysosomal lipid storage disorder, caused by mutations of the lysosomal beta-galactosidase (beta-gal) and results in the accumulation of G(M1). The underlying mechanisms of neurodegeneration are poorly understood. Here we demonstrate increased autophagy in beta-gal-deficient (beta-gal(-/-)) mouse brains as evidenced by elevation of LC3-II and beclin-1 levels. Activation of autophagy in the beta-gal(-/-) brain was found to be accompanied with enhanced Akt-mTOR and Erk signaling. In addition, the mitochondrial cytochrome c oxidase activity was significantly decreased in brains and cultured astrocytes from beta-gal(-/-) mouse. Mitochondria isolated from beta-gal(-/-) astrocytes were morphologically abnormal and had a decreased membrane potential. These cells were more sensitive to oxidative stress than wild type cells and this sensitivity was suppressed by ATP, an autophagy inhibitor 3-methyladenine and a pan-caspase inhibitor z-VAD-fmk. These results suggest activation of autophagy leading to mitochondrial dysfunction in the brain of G(M1)-gangliosidosis.

    Topics: Adenine; Adenosine Triphosphate; Animals; Apoptosis Regulatory Proteins; Astrocytes; Autophagy; Beclin-1; beta-Galactosidase; Brain; Cells, Cultured; Disease Models, Animal; Electron Transport Complex IV; Enzyme Inhibitors; G(M1) Ganglioside; Gangliosidosis, GM1; Lysosomes; Mice; Mice, Knockout; Microtubule-Associated Proteins; Mitochondria; Paraquat; Protein Kinases; Proteins; Signal Transduction; TOR Serine-Threonine Kinases

2008
Generation and characterization of recombinant feline beta-galactosidase for preclinical enzyme replacement therapy studies in GM1 gangliosidosis.
    Metabolic brain disease, 2008, Volume: 23, Issue:2

    Lysosomal beta-galactosidase is required for the degradation of GM1 ganglioside and other glycolipids and glycoproteins with a terminal galactose moiety. Deficiency of this enzyme leads to the lysosomal storage disorder, GM1 gangliosidosis, marked by severe neurodegeneration resulting in premature death. As a step towards preclinical studies for enzyme replacement therapy in an animal model of GM1 gangliosidosis, a feline beta-galactosidase cDNA was cloned into a mammalian expression vector and subsequently expressed in Chinese hamster ovary (CHO-K1) cells. The enzyme secreted into culture medium exhibited specific activity on two synthetic substrates as well as on the native beta-galactosidase substrate, GM1 ganglioside. The enzyme was purified from transfected CHO-K1 cell culture medium by chromatography on PATG-agarose. The affinity-purified enzyme preparation consisted mainly of the protein with approximate molecular weight of 94 kDa and displayed immunoreactivity with antibodies raised against a 16-mer synthetic peptide corresponding to C-terminal amino acid sequence deduced from the feline beta-galactosidase cDNA.

    Topics: Animals; Antibody Specificity; beta-Galactosidase; Cats; CHO Cells; Chromatography, Agarose; Cloning, Molecular; Cricetinae; Cricetulus; Culture Media, Conditioned; Disease Models, Animal; DNA, Complementary; G(M1) Ganglioside; Gangliosidosis, GM1; Genetic Therapy; Genetic Vectors; Molecular Weight; Protein Structure, Tertiary; Recombinant Proteins; Transfection

2008
Cerebrospinal fluid biomarkers showing neurodegeneration in dogs with GM1 gangliosidosis: possible use for assessment of a therapeutic regimen.
    Brain research, 2007, Feb-16, Volume: 1133, Issue:1

    The present study investigated cerebrospinal fluid (CSF) biomarkers for estimating degeneration of the central nervous system (CNS) in experimental dogs with GM1 gangliosidosis and preliminarily evaluated the efficacy of long-term glucocorticoid therapy for GM1 gangliosidosis using the biomarkers identified here. GM1 gangliosidosis, a lysosomal storage disease that affects the brain and multiple systemic organs, is due to an autosomal recessively inherited deficiency of acid beta-galactosidase activity. Pathogenesis of GM1 gangliosidosis may include neuronal apoptosis and abnormal axoplasmic transport and inflammatory response, which are perhaps consequent to massive neuronal storage of GM1 ganglioside. In the present study, we assessed some possible CSF biomarkers, such as GM1 ganglioside, aspartate aminotransferase (AST), lactate dehydrogenase (LDH), neuron-specific enolase (NSE) and myelin basic protein (MBP). Periodic studies demonstrated that GM1 ganglioside concentration, activities of AST and LDH, and concentrations of NSE and MBP in CSF were significantly higher in dogs with GM1 gangliosidosis than those in control dogs, and their changes were well related with the months of age and clinical course. In conclusion, GM1 ganglioside, AST, LDH, NSE and MBP could be utilized as CSF biomarkers showing CNS degeneration in dogs with GM1 gangliosidosis to evaluate the efficacy of novel therapies proposed for this disease. In addition, we preliminarily treated an affected dog with long-term oral administration of prednisolone and evaluated the efficacy of this therapeutic trial using CSF biomarkers determined in the present study. However, this treatment did not change either the clinical course or the CSF biomarkers of the affected dog, suggesting that glucocorticoid therapy would not be effective for treating GM1 gangliosidosis.

    Topics: Animals; Anti-Inflammatory Agents; Aspartate Aminotransferases; Biomarkers; Brain; Cerebrospinal Fluid Proteins; Disease Models, Animal; Dogs; G(M1) Ganglioside; Gangliosidosis, GM1; L-Lactate Dehydrogenase; Myelin Basic Protein; Nerve Degeneration; Phosphopyruvate Hydratase; Predictive Value of Tests; Prednisolone; Treatment Outcome; Up-Regulation

2007
Anti-diarrheal effect of Galla Chinensis on the Escherichia coli heat-labile enterotoxin and ganglioside interaction.
    Journal of ethnopharmacology, 2006, Feb-20, Volume: 103, Issue:3

    Enterotoxigenic Escherichia coli (ETEC) is the most frequently isolated enteropathogen, accounting for approximately 210 million diarrhea episodes annually. ETEC-induced diarrhea is initiated by the binding of B subunit of heat-labile enterotoxin (LTB) to the ganglioside G(M1) on the surface of intestinal epithelial cell. Therefore, we evaluated the inhibitory effects of 297 Chinese medicinal herbs on the LTB and G(M1) interaction by G(M1)-enzyme-linked immunosorbent assay. Galla Chinensis extract (GCE) exhibited anti-LT-induced diarrheal effect in the patent mouse gut assay, with IC50 value of 4.7+/-1.3 mg/ml. GCE also inhibited the binding of LTB to G(M1), suggesting that GCE suppressed the LT-induced fluid accumulation by blocking the binding of LTB to G(M1). Furthermore, the ethyl acetate (EA) soluble fraction was the most active fraction of Galla Chinensis that inhibiting the binding of LTB to G(M1) with an IC50 value of 153.6+/-3.4 microg/ml. The major components of the EA fraction should be phenolic derivatives according to a thin-layer chromatography analysis. Gallic acid, the major component of EA fraction, blocked the binding of LTB to G(M1), resulting in the suppression of LT-induced diarrhea. In conclusion, these data suggested that Galla Chinensis and gallic acid might be potent drugs for the treatment of LT-induced diarrhea.

    Topics: Animals; Antidiarrheals; Bacterial Toxins; Binding, Competitive; Diarrhea; Disease Models, Animal; Dose-Response Relationship, Drug; Drug Interactions; Drugs, Chinese Herbal; Enterotoxins; Escherichia coli Proteins; Female; G(M1) Ganglioside; Gallic Acid; Mice; Mice, Inbred BALB C; Plant Tumors; Rhus

2006
Experimental axonopathy induced by immunization with Campylobacter jejuni lipopolysaccharide from a patient with Guillain-Barré syndrome.
    Journal of neuroimmunology, 2006, Volume: 174, Issue:1-2

    New Zealand white rabbits were immunized with a lipopolysaccharide (LPS) extracted from a Campylobacter jejuni HS:19 strain isolated from a GBS patient expressing GM1 and GD1a-like epitopes, Freund's adjuvant (group I) and Freund's adjuvant plus keyhole lympet hemocyanin (KLH) (group II). Both groups showed high titers of anti-LPS and anti-GM1 and lower titers of anti-GD1b and anti-GD1a antibodies. Weakness and axonal degeneration in sciatic nerves was detected in 1/11 of group I and 6/7 of group II. This model replicates, at least in part, the pathogenetic process hypothesized in the human axonal GBS with antiganglioside antibodies post C. jejuni infection and indicates that KLH plays an additional role in neuropathy induction.

    Topics: Animals; Antibodies, Bacterial; Blotting, Western; Campylobacter jejuni; Disease Models, Animal; Electrophoresis, Polyacrylamide Gel; Enzyme-Linked Immunosorbent Assay; Epitopes; G(M1) Ganglioside; Gangliosides; Gene Expression; Guillain-Barre Syndrome; Humans; Immunization; Lipopolysaccharides; Male; Polyradiculoneuropathy; Rabbits; Time Factors

2006
High affinity of anti-GM1 antibodies is associated with disease onset in experimental neuropathy.
    Journal of neuroscience research, 2006, Volume: 84, Issue:5

    High antibody affinity has been proposed as a disease determinant factor in neuropathies associated with anti-GM1 antibodies. An experimental model of Guillain-Barré syndrome, induced by immunization of rabbits with bovine brain gangliosides or GM1, was described recently (Yuki et al. [2001] Ann. Neurol. 49:712-720). We searched plasma from these rabbits, taken at disease onset and 1 or 2 weeks prior to onset, for the presence of high-affinity anti-GM1 IgG antibodies. Affinity was estimated by soluble antigen binding inhibition. High-affinity antibodies (binding inhibition by 10(-9) M GM1) were detected at disease onset but not before. No such difference was found for other antibody parameters such as titer, fine specificity, and population distribution. These findings support the proposed role of high affinity as an important factor in disease induction by anti-GM1 antibodies.

    Topics: Animals; Antibody Affinity; Antibody Specificity; Autoantibodies; Binding Sites, Antibody; Cattle; Chromatography, Thin Layer; Disease Models, Animal; Enzyme-Linked Immunosorbent Assay; G(M1) Ganglioside; Guillain-Barre Syndrome; Rabbits; Time Factors

2006
a-Series gangliosides mediate the effects of advanced glycation end products on pericyte and mesangial cell proliferation: a common mediator for retinal and renal microangiopathy?
    Diabetes, 2005, Volume: 54, Issue:1

    Advanced glycation end products (AGEs) are involved in the development of microvascular complications, including alterations of retinal pericyte and renal mesangial cell growth occurring during diabetic retinopathy and diabetic nephropathy, respectively. Because gangliosides are implicated in the regulation of cell proliferation, we hypothesized that AGEs could exert cellular effects in part by modulating ganglioside levels. Results of the present study indicate that AGEs caused an inhibition of both bovine retinal pericyte (BRP) and rat renal mesangial cell (RMC) proliferation, associated with an increase of a-series gangliosides consecutive to GM3 synthase activity increase and GD3 synthase activity inhibition. Similar modifications were also found in the renal cortex of diabetic db/db mice compared with controls. Treatment of BRP and RMC with exogenous a-series gangliosides decreased proliferation and blockade of a-series gangliosides with specific antibodies partially protecting the two cell types from the AGE-induced proliferation decrease. Further, inhibition of GM3 synthase using specific SiRNA partially reversed the AGE effects on mesangial cell proliferation. These results suggest that a-series gangliosides are mediators of the adverse AGE effects on BRP and RMC proliferation. They also raise the hypothesis of common mechanisms involved in the development of diabetic retinopathy and diabetic nephropathy.

    Topics: Animals; Cattle; Cell Division; Cells, Cultured; Diabetes Mellitus, Type 1; Diabetic Nephropathies; Diabetic Retinopathy; Disease Models, Animal; G(M1) Ganglioside; G(M3) Ganglioside; Gangliosides; Glomerular Mesangium; Glycation End Products, Advanced; Kidney Cortex; Mice; Microcirculation; Pericytes; Rats; Retinal Vessels; RNA, Small Interfering; Sialyltransferases

2005
G(M1)-ganglioside degradation and biosynthesis in human and murine G(M1)-gangliosidosis.
    Clinica chimica acta; international journal of clinical chemistry, 2005, Volume: 354, Issue:1-2

    Gangliosides are building blocks of cell membranes and their biosynthesis and degradation have been extensively studied in the past. Regulation of the metabolism of these glycolipids controls fundamental cell functions. G(M1)-gangliosidosis, a neurodegenerative glycosphingolipid storage disease, is caused by deficiency of lysosomal beta-galactosidase with consequent disruption of the normal degradative pathway of G(M1)-ganglioside. We studied the impact of G(M1)-ganglioside accumulation on its biosynthetic enzyme in cells and tissues from human patients and from the G(M1)-gangliosidosis mouse model.. We tested the qualitative and quantitative pattern of gangliosides by thin layer chromatography and N-acetylneuraminic acid dosage, respectively. Regulation of G(M1)-ganglioside biosynthesis was evaluated by G(M1) synthase assay in human and murine samples.. G(M1)-ganglioside accumulation has an inhibitory effect on the human but not on the mouse G(M1) synthase. We present evidence that G(M1) synthase activity in human and murine cells are regulated by different mechanisms.. Alternative pathways in the mouse may account for these results and possibly explain some of the phenotypical differences between the human and mouse forms of this disorder.

    Topics: Animals; Brain; Chromatography, Thin Layer; Disease Models, Animal; Fibroblasts; G(M1) Ganglioside; Gangliosidosis, GM1; Hexosyltransferases; Humans; Mice; Mice, Inbred Strains; Phenotype

2005
Brucella melitensis infection associated with Guillain-Barré syndrome through molecular mimicry of host structures.
    FEMS immunology and medical microbiology, 2005, Aug-01, Volume: 45, Issue:2

    Brucella melitensis is a facultative intracellular bacterium that can survive inside macrophages and the causative agent of brucellosis. In the present study, we found that a lipooligosaccharide of B. melitensis has a GM1 ganglioside-like structure and shows a strong antibody response in mice. The cholera toxin B subunit, which binds to GM1 ganglioside specifically, reacted with the surface of B. melitensis. Immunization with B. melitensis induced the production of anti-GM1 ganglioside antibodies in mice and serum from immunized mice showed a cross-reaction with Guillain-Barré syndrome (GBS)-associated Campylobacter jejuni, but not non-GBS-associated C. jejuni. When B. melitensis was treated with a neuraminidase, antibody responses disappeared. B. melitensis immunization induced the production of anti-GM1 ganglioside antibodies in BALB/c mice but not in C57BL/6 and ddY mice, and for BALB/c mice, immunization with B. melitensis induced much greater production of anti-GM1 ganglioside than GBS-associated C. jejuni. Flaccid limb weakness was observed in B. melitensis immunized mice. These results suggest that B. melitensis is a new etiological agent for GBS and that immunological responses between it and GBS-associated C. jejuni in the mouse model may be different.

    Topics: Animals; Antibodies, Bacterial; Brucella melitensis; Brucellosis; Campylobacter jejuni; Cholera Toxin; Cross Reactions; Disease Models, Animal; G(M1) Ganglioside; Guillain-Barre Syndrome; Humans; Immunization; In Vitro Techniques; Lipopolysaccharides; Mice; Mice, Inbred BALB C; Mice, Inbred C57BL; Molecular Mimicry

2005
GM1 ganglioside prevented the decline of hippocampal neurogenesis associated with D-galactose.
    Neuroreport, 2005, Aug-22, Volume: 16, Issue:12

    Hippocampal neurogenesis is thought to play a functional role in hippocampal-dependent learning and memory. The neurogenic capability of the hippocampus declines with age and may be associated with a decline in cognitive function. In the present study, an established model of ageing in mice was used to test the protective effects of GM1 ganglioside on hippocampal neurogenesis. This model uses D-galactose treatment to cause neuronal injury and reduced neurogenesis. GM1 significantly increased the proliferation, long-term survival and neuronal differentiation of hippocampal progenitors that had been injured with D-galactose. This study demonstrates that GM1 can protect hippocampal neurogenesis from D-galactose injury. Therefore, GM1 may protect neurogenesis in the ageing brain.

    Topics: Analysis of Variance; Animals; Bromodeoxyuridine; Cell Count; Cell Death; Cell Proliferation; Disease Models, Animal; Doublecortin Domain Proteins; G(M1) Ganglioside; Galactose; Hippocampus; Immunohistochemistry; In Situ Nick-End Labeling; Male; Mice; Mice, Inbred C57BL; Microtubule-Associated Proteins; Nerve Degeneration; Neuropeptides; Neuroprotective Agents; Phosphopyruvate Hydratase; Random Allocation; Regeneration

2005
CDP-choline liposomes provide significant reduction in infarction over free CDP-choline in stroke.
    Brain research, 2005, Oct-05, Volume: 1058, Issue:1-2

    Cytidine-5'-diphosphocholine (CDP-choline, Citicoline, Somazina) is in clinical use (intravenous administration) for stroke treatment in Europe and Japan, while USA phase III stroke clinical trials (oral administration) were disappointing. Others showed that CDP-choline liposomes significantly increased brain uptake over the free drug in cerebral ischemia models. Liposomes were formulated as DPPC, DPPS, cholesterol, GM(1) ganglioside; 7/4/7/1.57 molar ratio or 35.8/20.4/35.8/8.0 mol%. GM(1) ganglioside confers long-circulating properties to the liposomes by suppressing phagocytosis. CDP-choline liposomes deliver the agent intact to the brain, circumventing the rate-limiting, cytidine triphosphate:phosphocholine cytidylyltransferase in phosphatidylcholine synthesis. Our data show that CDP-choline liposomes significantly ( P < 0.01) decreased cerebral infarction (by 62%) compared to the equivalent dose of free CDP-choline (by 26%) after 1 h focal cerebral ischemia and 24 h reperfusion in spontaneously hypertensive rats. Beneficial effects of CDP-choline liposomes in stroke may derive from a synergistic effect between the phospholipid components of the liposomes and the encapsulated CDP-choline.

    Topics: Animals; Brain; Brain Infarction; Brain Ischemia; Cholesterol; Choline; Cytidine Diphosphate Choline; Disease Models, Animal; Drug Combinations; Drug Synergism; G(M1) Ganglioside; Liposomes; Male; Phagocytosis; Phosphatidylcholines; Rats; Rats, Inbred SHR; Reperfusion Injury; Stroke; Treatment Outcome

2005
Natural killer (NK) cells play a critical role in the early innate immune response to Chlamydophila abortus infection in mice.
    Journal of comparative pathology, 2004, Volume: 130, Issue:1

    Chlamydophila abortus, the aetiological agent of ovine enzootic abortion, induces a strong inflammatory reaction that leads to the T helper cell (Th1) specific immune response necessary for the clearance of infection. Because the role of natural killer (NK) cells during the first stages of this response has received little attention, this study focused on determining the function of these cells in a mouse model of infection. The location of NK cells in the liver and spleen of infected mice was examined immunohistochemically with an anti-Ly49G monoclonal antibody. The number of NK cells increased during the infection both in spleen and liver. In subsequent experiments, an anti-asialo GM1 polyclonal antibody was injected to deplete the NK cells. NK-depleted mice showed a substantial increase in their susceptibility to C. abortus infection, with high mortality rates and an increased burden of bacteria in the liver. Histopathological studies showed that inflammatory foci, composed mainly of neutrophils, were greater in size and number in depleted mice, while numerous chlamydial inclusions were associated with the foci. Serum concentrations of IFN-gamma, a key cytokine in the control of C. abortus infection, were substantially reduced in the NK-depleted mice. To establish the relationship between NK cells and other components of the innate immune response, neutrophils were depleted with the RB6-8C5 antibody. These cells were shown to be crucial in the recruitment of NK cells to the inflammatory foci.

    Topics: Animals; Chlamydophila; Chlamydophila Infections; Disease Models, Animal; G(M1) Ganglioside; Killer Cells, Natural; Liver; Lymphocyte Depletion; Mice; Mice, Inbred C57BL; Neutrophils; Spleen

2004
Interferon alpha 2b decreases hepatic micrometastasis in a murine model of ocular melanoma by activation of intrinsic hepatic natural killer cells.
    Investigative ophthalmology & visual science, 2004, Volume: 45, Issue:7

    To investigate in a murine model the mechanism by which micrometastatic melanoma, which spreads from the eye to the liver, is controlled by interferon (IFN)-alpha 2b.. Major histocompatibility (MHC) class I antigen (H-2, all haplotypes) expression in three murine melanoma cell lines (Queens, B16LS9, B16F10) was determined by flow cytometric immunophenotyping. The cell lines were heterotopically inoculated into the posterior compartments (PCs) of C57Bl/6 mice, and the mice were given intraperitoneal (IP) injections of IFN-alpha 2b or PBS for 1 or 4 days before enucleation at 7 days after inoculation. Groups of mice were made NK deficient or depleted with subcutaneous (s.c.) injection of anti-asialo GM1. The mice were killed at 28 days or 56 days (survival experiment) after inoculation, and the number of hepatic micrometastases was histologically determined. NK cells were isolated from the spleen and liver at necropsy, and propidium iodide labeled target-specific cytolysis was determined by flow cytometry. The micrometastases were evaluated for apoptosis and proliferation with TUNEL and MIB1 immunostaining, respectively, and TUNEL-to-MIB1 ratios were determined. Hepatic NK cells were immunostained with CD49b.. MHC class I antigen was expressed in the three cell lines in the order of Queens < B16LS9 < B16F10. All cell lines grew, were confined to the PC, and formed hepatic micrometastases. A decrease in micrometastases, an increase in target-specific cytolysis, and an increase in survival correlated with decreased HLA class I expression by the melanoma cells. The IFN-alpha 2b treatment resulted in a boost of intrinsic hepatic NK cells, demonstrated in NK-deficient but not NK-depleted mice. The treatment effect corresponded to increased apoptosis (TUNEL)-proliferation (MIB1) ratios in the micrometastases. Immunostaining demonstrated an increased number of intrahepatic NK cells associated with the micrometastases in treated groups.. Neoadjuvant IFN-alpha 2b results in decreased hepatic micrometastasis and increased survival time through increased intrinsic hepatic NK cell-mediated tumor apoptosis in a murine model of metastatic ocular melanoma.

    Topics: Animals; Antineoplastic Agents; Apoptosis; Cytotoxicity, Immunologic; Disease Models, Animal; Female; Flow Cytometry; G(M1) Ganglioside; Histocompatibility Antigens Class I; Immunophenotyping; In Situ Nick-End Labeling; Injections, Intraperitoneal; Interferon alpha-2; Interferon-alpha; Killer Cells, Natural; Liver; Liver Neoplasms, Experimental; Lymphocyte Activation; Lymphocyte Depletion; Melanoma, Experimental; Mice; Mice, Inbred C57BL; Recombinant Proteins; Spleen; Uveal Neoplasms

2004
An anti-ganglioside antibody-secreting hybridoma induces neuropathy in mice.
    Annals of neurology, 2004, Volume: 56, Issue:2

    Immune responses against gangliosides are strongly implicated in the pathogenesis of some variants of Guillain-Barré syndrome (GBS). For example, IgG antibodies against GM1, GD1a, and related gangliosides are frequently present in patients with post-Campylobacter acute motor axonal neuropathy (AMAN) variant of GBS, and immunization of rabbits with GM1 has produced a model of AMAN. However, the role of anti-ganglioside antibodies in GBS continues to be debated because of lack of a passive transfer model. We recently have raised several monoclonal IgG anti-ganglioside antibodies. We passively transfer these antibodies by intraperitoneal hybridoma implantation and by systemic administration of purified anti-ganglioside antibodies in mice. Approximately half the animals implanted with an intraperitoneal clone of anti-ganglioside antibody-secreting hybridoma developed a patchy, predominantly axonal neuropathy affecting a small proportion of nerve fibers. In contrast to hybridoma implantation, passive transfer with systemically administered anti-ganglioside antibodies did not cause nerve fiber degeneration despite high titre circulating antibodies. Blood-nerve barrier studies indicate that animals implanted with hybridoma had leaky blood-nerve barrier compared to mice that received systemically administered anti-ganglioside antibodies. Our findings suggest that in addition to circulating antibodies, factors such as antibody accessibility and nerve fiber resistance to antibody-mediated injury play a role in the development of neuropathy.

    Topics: Animals; Antibodies, Monoclonal; Antibody Formation; Blood-Brain Barrier; Blotting, Western; Capillary Permeability; Disease Models, Animal; G(M1) Ganglioside; Gangliosides; Hybridomas; Immunoglobulin G; Immunohistochemistry; Ki-67 Antigen; Male; Mice; Mice, Inbred Strains; Nerve Degeneration; Peripheral Nerves; Peripheral Nervous System Diseases; Species Specificity; Spinal Cord

2004
GM1-ganglioside-mediated activation of the unfolded protein response causes neuronal death in a neurodegenerative gangliosidosis.
    Molecular cell, 2004, Sep-10, Volume: 15, Issue:5

    GM1-ganglioside (GM1) is a major sialoglycolipid of neuronal membranes that, among other functions, modulates calcium homeostasis. Excessive accumulation of GM1 due to deficiency of lysosomal beta-galactosidase (beta-gal) characterizes the neurodegenerative disease GM1-gangliosidosis, but whether the accumulation of GM1 is directly responsible for CNS pathogenesis was unknown. Here we demonstrate that activation of an unfolded protein response (UPR) associated with the upregulation of BiP and CHOP and the activation of JNK2 and caspase-12 leads to neuronal apoptosis in the mouse model of GM1-gangliosidosis. GM1 loading of wild-type neurospheres recapitulated the phenotype of beta-gal-/- cells and activated this pathway by depleting ER calcium stores, which ultimately culminated in apoptosis. Activation of UPR pathways did not occur in mice double deficient for beta-gal and ganglioside synthase, beta-gal-/-/GalNAcT-/-, which do not accumulate GM1. These findings suggest that the UPR can be induced by accumulation of the sialoglycolipid GM1 and this causes a novel mechanism of neuronal apoptosis.

    Topics: Animals; Animals, Newborn; Apoptosis; beta-Galactosidase; Calcium; Caspase 12; Caspases; CCAAT-Enhancer-Binding Proteins; Cell Death; Cells, Cultured; Disease Models, Animal; Endoplasmic Reticulum Chaperone BiP; G(M1) Ganglioside; Gangliosidosis, GM1; Heat-Shock Proteins; Mice; Mice, Knockout; Mitogen-Activated Protein Kinase 9; Mitogen-Activated Protein Kinases; Molecular Chaperones; N-Acetylgalactosaminyltransferases; Nerve Degeneration; Neurons; Polypeptide N-acetylgalactosaminyltransferase; Protein Folding; Transcription Factor CHOP; Transcription Factors

2004
Beta 2 microglobulin knockout mice are resistant to lethal intraabdominal sepsis.
    American journal of respiratory and critical care medicine, 2003, Jun-15, Volume: 167, Issue:12

    beta 2 microglobulin knockout (beta2M-/-) mice lack CD8+ T and natural killer T cells. We hypothesized that beta 2M-/- mice are resistant to lethal intraabdominal sepsis. To test this hypothesis, mortality, cytokine production, and physiologic function were assessed in beta 2M-/- mice during sepsis caused by cecal ligation and puncture (CLP). beta 2M-/- mice survived significantly longer than wild-type mice after CLP but ultimately exhibited 100% mortality. Treatment of beta 2M-/- mice with anti-asialoGM1 to deplete natural killer cells conferred greater than 70% long-term survival. Compared with wild-type mice, beta 2M-/- mice treated with anti-asialoGM1 produced decreased amounts of proinflammatory cytokines and did not exhibit hypothermia or metabolic acidosis after CLP. Adoptive transfer of CD8+ T and natural killer cells into beta 2M-/- mice treated with anti-asialoGM1 re-established CLP-induced mortality. CD8 knockout mice treated with anti-asialoGM1, which are specifically deficient in CD8+ T and natural killer cells, exhibited 40% long-term survival after CLP. Furthermore, treatment of wild-type mice with antibodies to CD8 and asialoGM1 conferred a significant survival benefit compared with wild-type mice treated with nonspecific IgG. These findings demonstrate that beta 2M-/- mice treated with anti-asialoGM1 are resistant to CLP-induced mortality and that depletion of CD8+ T and natural killer cells largely accounts for the survival benefit observed in these mice.

    Topics: Adoptive Transfer; Animals; beta 2-Microglobulin; CD8-Positive T-Lymphocytes; Cecum; Disease Models, Animal; Female; G(M1) Ganglioside; Immunity, Innate; Inflammation; Killer Cells, Natural; Ligation; Lymphopenia; Mice; Mice, Inbred C57BL; Mice, Knockout; Peritonitis; Sepsis; Survival Analysis

2003
Induction of conduction block by Campylobacter jejuni lipopolysaccharides and focal neural insult.
    Journal of the neurological sciences, 2003, Sep-15, Volume: 213, Issue:1-2

    A systemic exposure to gram negative LPS have caused transient conduction abnormalities in a certain strain of rats probably associated with the action of cytokines secreted by macrophages. Our previous studies demonstrated that anti-GM1 antibodies induced in rats by the cross-reactive Cj-LPS, caused no conduction abnormalities. We designed the present study to evaluate the effect of systemic exposure to Cj-LPS on nerve conduction after a focal minor neural trauma. Female Lewis rats were sensitized against KLH by repetitive subcutaneous injections. After 28 days rats were intraneurally injected with saline in the right sciatic nerve and concomitantly with intraperitoneal Cj-LPS. Sciatic nerve conduction studies were performed on days 0, 1, 2, 3, and 7 after injections. Nerve conduction blocks developed in all the rats (n=10) which received an intraneural injection of saline concomitantly with the systemic Cj-LPS exposure, before titers of anti-ganglioside antibodies were detected. We conclude that humoral factors (possibly cytokines), other than antibodies are secreted by lymphocytes and macrophages stimulated by gram negative LPS, and cause functional conduction abnormalities when the blood-nerve barrier is disrupted.

    Topics: Action Potentials; Animals; Antibodies; Campylobacter jejuni; Cross Reactions; Disease Models, Animal; Electric Stimulation; Enzyme-Linked Immunosorbent Assay; Female; G(M1) Ganglioside; Hemocyanins; Lipopolysaccharides; Neural Conduction; Neuritis, Autoimmune, Experimental; Rats; Rats, Inbred Lew; Sciatic Nerve; Time Factors

2003
IP-10 and Mig facilitate accumulation of T cells in the virus-infected liver.
    Cellular immunology, 2002, Volume: 219, Issue:1

    Viral infection of the liver causes accumulation of T cells in the infected organ, raising the question as to the signals that mediate this response. Employing an adenovirus induced hepatitis model in mice, we show that IP-10 and Mig are essential for T cell recruitment and that induction of the two chemokines occurs concomitant to production of IFNgamma. It is shown that while IFNgamma induces IP-10 and Mig in hepatocytes, for optimal chemokine induction, a co-stimulatory signal mediated by cross-linking of Fas on hepatocytes is required. Moreover, cross-linking of Fas by injection of anti-Fas antibody into mice triggers induction of IP-10 and Mig in the liver. The cells providing the two signals are shown to express NK1.1 and AsGM1; elimination of these cells leads to inhibition of IFNgamma and chemokine transcript induction. The conclusion is drawn that both NK cells and T cells provide the two signals for induction of IP-10 and Mig in the liver.

    Topics: Adenoviridae Infections; Animals; Antibodies; Antigens; Antigens, Ly; Antigens, Surface; Cell Line; Chemokine CXCL10; Chemokine CXCL9; Chemokines, CXC; Disease Models, Animal; fas Receptor; Female; G(M1) Ganglioside; Hepatitis, Viral, Animal; Intercellular Signaling Peptides and Proteins; Interferon-gamma; Killer Cells, Natural; Lectins, C-Type; Liver; Lymphocyte Subsets; Mice; Mice, Inbred C57BL; Mice, SCID; NK Cell Lectin-Like Receptor Subfamily B; Proteins; Signal Transduction; T-Lymphocytes; Up-Regulation

2002
Pseudomonas aeruginosa infection of respiratory epithelium in a cystic fibrosis xenograft model.
    The Journal of infectious diseases, 2001, Mar-15, Volume: 183, Issue:6

    Pulmonary infection with Pseudomonas aeruginosa in patients with cystic fibrosis (CF) causes a chronic destructive bronchitis. A xenograft model was used to study the susceptibility of the CF respiratory epithelium to P. aeruginosa strain PAK and the virulence of certain mutants. Despite an early trend toward increased susceptibility, colonization of CF xenografts (ID(95), 62 colony-forming units [cfu]) was not statistically different (P=.5) than in xenografts with normal respiratory cells (ID(95), 1.2x10(3) cfu). Infection severity in 12 CF xenografts (mean polymorphonuclear leukocyte [PMNL] density, 1.88x10(6)+/-1.75x10(6)/xenograft) was similar to that in 16 non-CF xenografts (3.19x10(6)+/-2.45x10(6) PMNL/xenograft; P=.38), despite slightly greater bacterial density in the CF xenografts (mean, 1.57+/-2.73x10(6) cfu/xenograft) versus xenografts with normal epithelium (mean, 1.03+/-1.3x10(6) cfu/xenograft). P. aeruginosa mutants pilA and fliF, but not rpoN, colonized normal respiratory xenografts, indicating that colonization and infection in this model depend on an uncharacterized RpoN-controlled gene. This model appears to be suitable for genetic study of P. aeruginosa virulence but not of the CF respiratory tract's unique susceptibility.

    Topics: Animals; Colony Count, Microbial; Cystic Fibrosis; Disease Models, Animal; Epitopes; Female; G(M1) Ganglioside; Humans; Mice; Mice, Inbred BALB C; Mice, Nude; Mutation; N-Acetylneuraminic Acid; Pseudomonas aeruginosa; Pseudomonas Infections; Respiratory Mucosa; Transplantation, Heterologous

2001
Variants of the Guillain Barré syndrome: progress toward fulfilling "Koch's postulates".
    Annals of neurology, 2001, Volume: 49, Issue:6

    Topics: Animals; Antibodies; Antibody Specificity; Cattle; Disease Models, Animal; G(M1) Ganglioside; Guillain-Barre Syndrome; Immunization; Peripheral Nerves; Phenotype; Rabbits

2001
Animal model of axonal Guillain-Barré syndrome induced by sensitization with GM1 ganglioside.
    Annals of neurology, 2001, Volume: 49, Issue:6

    Some humans develop the axonal form of Guillain-Barré syndrome after receiving bovine brain ganglioside. On sensitization with the ganglioside mixture, all of a group of rabbits injected developed high anti-GM1 IgG antibody titers, flaccid limb weakness of acute onset, and a monophasic illness course. Pathological findings for the peripheral nerves showed predominant Wallerian-like degeneration, with neither lymphocytic infiltration nor demyelination. IgG was deposited on the axons of the anterior roots, and GM1 was proved to be present on the axons of peripheral nerves. Sensitization with purified GM1 also induced axonal neuropathy, indicating that GM1 was the immunogen in the mixture. A model of human axonal Guillain-Barré syndrome has been established that uses inoculation with a bovine brain ganglioside mixture or isolated GM1. This model may help to clarify the molecular pathogenesis of the syndrome and to develop new treatments for it.

    Topics: Animals; Antibodies; Antibody Specificity; Autoantibodies; Axons; Cattle; Chemotaxis, Leukocyte; Disease Models, Animal; G(M1) Ganglioside; Guillain-Barre Syndrome; Immunization; Immunoglobulin G; Immunoglobulin M; Immunohistochemistry; Male; Muscle Weakness; Peripheral Nerves; Rabbits; Wallerian Degeneration

2001
Campylobacter jejuni lipopolysaccharides from Guillain-Barré syndrome patients induce IgG anti-GM1 antibodies in rabbits.
    Journal of neuroimmunology, 2000, May-01, Volume: 104, Issue:2

    Lipopolysaccharides (LPS) from Campylobacter jejuni strains isolated from patients with Guillain-Barré syndrome (GBS) display molecular mimicry with GM1. We immunized rabbits with C. jejuni LPS from GBS-associated strains containing a GM1-like epitope. All animals produced high titre anti-LPS antibodies that were cross-reactive with GM1. We conclude that C. jejuni strains from GBS patients are able to induce antibodies that cross-react with gangliosides and LPS. This study further confirms the role of molecular mimicry in the induction of anti-ganglioside antibodies in GBS patients.

    Topics: Animals; Antibodies; Antibody Formation; Campylobacter jejuni; Disease Models, Animal; Epitopes; G(M1) Ganglioside; G(M2) Ganglioside; Guillain-Barre Syndrome; Humans; Immunization; Immunoglobulin G; Immunoglobulin M; Lipopolysaccharides; Rabbits; Time Factors

2000
Systemic treatment with GM1 ganglioside improves survival and function of cryopreserved embryonic midbrain grafted to the 6-hydroxydopamine-lesioned rat striatum.
    Experimental neurology, 2000, Volume: 164, Issue:1

    Cryopreservation may allow long-term storage of embryonic ventral mesencephalon (VM) for neural transplantation. We investigated whether the ganglioside GM1 or the lazaroid tirilazad mesylate (U-74006F) could improve survival of grafts derived from cryopreserved VM in a rat model of Parkinson's disease. VM was dissected from rat embryos (E14-E15), frozen and stored in liquid nitrogen under controlled conditions, thawed, dissociated, and then grafted into the 6-hydroxydopamine-lesioned rat striatum. In Experiment I, VM fragments were exposed in vitro either to GM1 (100 microM) or to lazaroid (0.3 microM) during all preparative steps. In Experiment II, rats receiving GM1-pretreated VM were, in addition, treated systematically with GM1 (30 mg/kg) daily for 3.5 weeks. Rats grafted with untreated cryopreserved or fresh VM were used as controls, respectively. Rats receiving fresh VM control grafts showed complete recovery from lesion-induced rotations after 6 weeks whereas rats grafted with cryopreserved VM (untreated or pretreated) did not recover. Cryografts contained significantly less (18%, control; 23%, GM1; and 12%, lazaroid) tyrosine hydroxylase-positive cells compared to fresh grafts (1415 +/- 153; mean +/- SEM). Graft volume was also significantly smaller after cryopreservation. In contrast, with additional systemic GM1 treatment cryografts contained almost the same number of tyrosine hydroxylase-positive cells (376 +/- 85) as fresh grafts (404 +/- 56), which was significantly more than that of untreated cryografts (147 +/- 20), showed a significantly larger volume (0.15 mm(3)) compared to that of untreated grafts (0.08 mm(3)) (fresh controls, 0.19 mm(3)), and induced significant and complete functional recovery in the rotation test. In conclusion, systemic treatment of rats with GM1 improved the low survival and functional inefficacy of grafts derived from cryopreserved VM whereas tissue pretreatment alone with either GM1 or lazaroid was not effective.

    Topics: Animals; Brain Tissue Transplantation; Cell Count; Cell Survival; Corpus Striatum; Cryopreservation; Disease Models, Animal; Female; Fetal Tissue Transplantation; G(M1) Ganglioside; Graft Survival; Mesencephalon; Motor Activity; Neurons; Neuroprotective Agents; Oxidopamine; Parkinson Disease, Secondary; Pregnatrienes; Rats; Rats, Sprague-Dawley; Recovery of Function; Tyrosine 3-Monooxygenase

2000
Human peripheral blood lymphocyte severe combined immunodeficiency (hu-PBL SCID) models of toxoplasmosis.
    Immunology and cell biology, 2000, Volume: 78, Issue:6

    Toxoplasmosis is a potentially fatal opportunistic infection of immunocompromised hosts. Improved animal models of toxoplasmosis are needed to more nearly approximate conditions that occur in immunocompromised humans. The development of models of toxoplasmosis using human peripheral blood lymphocytes (hu-PBL) transplanted into severe combined immunodeficiency (SCID) mice is described here. Transplantation of hu-PBL into SCID mice without prior conditioning of the mice resulted in detectable differences in quantitative histological scores of brain inflammation due to Toxoplasma gondii infection, but did not alter mortality when compared to SCID mouse controls. The lack of detectable differences in survival were due to inadequate engraftment of hu-PBL, as assessed by flow cytometry. Unconditioned hu-PBL SCID mice had low titre T. gondii-specific antibody detectable after infection. When pretransplantation conditioning with irradiation and antiasialo GM 1 (n-glucolyl neuraminic acid) antibody was used, prolonged hu-PBL engraftment was observed in SCID mice, which was associated with worsened histopathology and usually impaired survival when compared with SCID mouse controls. When pretransplantation conditioning with irradiation, antiasialo GM antibody and polyethylene glycol-conjugated IL-2 was used, prolonged hu-PBL engraftment was also documented, but this did not affect survival from T. gondii infection when compared with similarly conditioned SCID mouse controls. The latter conditioning protocol resulted in hu-PBL SCID mice producing high titre T. gondii-specific antibody after infection. Conditioned hu-PBL SCID mice had evidence of increased T. gondii-induced inflammatory scores when compared with conditioned SCID mice. These models show promise for the study of the pathogenesis of toxoplasmosis and conditioned hu-PBL SCID mice may have applications for the evaluation of novel therapies for toxoplasmosis in immunocompromised humans.

    Topics: Acute Disease; Animals; Antibodies; Antibodies, Protozoan; Chronic Disease; Cytotoxicity Tests, Immunologic; Disease Models, Animal; Flow Cytometry; G(M1) Ganglioside; Humans; Killer Cells, Natural; Liver; Lymphocyte Count; Lymphocyte Transfusion; Mice; Mice, SCID; Spleen; Survival Rate; Toxoplasma; Toxoplasmosis, Animal; Transplantation Conditioning; Whole-Body Irradiation

2000
GM1 produces attenuation of short-term memory deficits in Hebb-Williams maze performance after unilateral entorhinal cortex lesions.
    Physiology & behavior, 1999, Volume: 66, Issue:3

    The Hebb-Williams maze was used to examine spatial abilities of adult male Sprague-Dawley rats with unilateral electrolytic entorhinal cortex lesions. The injured rats were treated for 14 days with either saline or ganglioside GM1. Testing was begun 7 weeks following injury, and involved 12 maze problems with independent configurations, with immediate starting replacement used for the six trials per problem. Compared to sham-operated counterparts, the rats with lesion plus saline treatment were impaired in total number of errors, initial entry errors, and repeat errors over 12 consecutive problems. GM1-treated rats showed improved performance, making significantly fewer total and repeat errors, indicating that this substance may be potentially useful as therapy after entorhinal cortex injury.

    Topics: Analysis of Variance; Animals; Brain Injuries; Cues; Disease Models, Animal; Entorhinal Cortex; G(M1) Ganglioside; Male; Maze Learning; Memory Disorders; Memory, Short-Term; Neuronal Plasticity; Neuroprotective Agents; Orientation; Rats; Rats, Sprague-Dawley

1999
Intravitreal injection of ganglioside GM1 after ischemia reduces retinal damage in rats.
    Stroke, 1997, Volume: 28, Issue:3

    Gangliosides are normal components of cell membranes and contribute to structural rigidity and membrane function. They have been shown to protect against various insults in the brain. We have shown previously that GM1 administered intraperitoneally before the induction of retinal ischemia provides a protective effect. This study evaluates the protective effect of GM1 administered intravitreally after ischemia on retinal lesions.. We induced retinal ischemia unilaterally in Long-Evans rats by increasing intraocular pressure to 160 mm Hg for 60 minutes. GM1 (20 microL x 10(-5) mol/L) or saline (20 microL) was injected into the vitreous 15 minutes after ischemia, and the postischemic survival time was either 8 or 15 days. The degree of retinal damage was assessed by histopathological study.. Retinal ischemia led to reductions in thickness and cell number, principally in the inner retinal layers (39% to 80%) and to a lesser extent in the outer retinal layers (26% to 45%). Postischemic treatment with intravitreally injected GM1 conferred significant protection against retinal ischemic damage after both 8 and 15 days of survival time. After 8 days of reperfusion, the ischemia-induced loss in overall retinal thickness was reduced by 15% and those of the inner nuclear and plexiform layers by 44% and 17%, respectively. Ischemic-induced ganglion cell and inner nuclear cell density losses were reduced by 37% and 27%, respectively. After 15 days of reperfusion, approximately the same statistically significant differences could be observed in comparison with the 15-day saline-injected group.. GM1 protects the rat retina from pressure-induced ischemic injury when given intravitreally after the insult. The protection provided by GM1 after initiation of retinal damage could be of therapeutic interest.

    Topics: Animals; Disease Models, Animal; Eye; G(M1) Ganglioside; Neuroprotective Agents; Rats; Rats, Inbred Strains; Reperfusion Injury; Retina; Sodium Chloride; Time Factors; Vitreous Body

1997
Generalized CNS disease and massive GM1-ganglioside accumulation in mice defective in lysosomal acid beta-galactosidase.
    Human molecular genetics, 1997, Volume: 6, Issue:2

    Human GM1-gangliosidosis is caused by a genetic deficiency of lysosomal acid beta-galactosidase (beta-gal). The disease manifests itself either as an infantile, juvenile or adult form and is primarily a neurological disorder with progressive brain dysfunction. A mouse model lacking a functional beta-gal gene has been generated by homologous recombination and embryonic stem cell technology. Tissues from affected mice are devoid of beta-gal mRNA and totally deficient in GM1-ganglioside-hydrolyzing capacity. Storage material was already conspicuous in the brain at 3 weeks. By 5 weeks, extensive storage of periodic acid Schiff-positive material was observed in neurons throughout the brain and spinal cord. Consistent with the neuropathology, abnormal accumulation of GM1-ganglioside in the brain progressed from twice to almost five times the normal amount during the period from 3 weeks to 3.5 months. Despite the accumulation of brain GM1-ganglioside at the level equal to or exceeding that seen in gravely ill human patients, these mice show no overt clinical phenotype up to 4-5 months. However, tremor, ataxia and abnormal gait become apparent in older mice. Thus, the beta-gal-deficient mice appear to mimic closely the pathological, biochemical and clinical abnormalities of the human disease.

    Topics: Animals; beta-Galactosidase; Brain; Central Nervous System Diseases; Disease Models, Animal; G(M1) Ganglioside; Gangliosides; Gangliosidosis, GM1; Glycosphingolipids; Humans; Mice; Mice, Inbred C57BL

1997
A model of human anti-T-cell monoclonal antibody therapy in SCID mice engrafted with human peripheral blood lymphocytes.
    Clinical transplantation, 1997, Volume: 11, Issue:5 Pt 2

    A chimeric severe combined immunodeficient mouse engrafted with human peripheral blood (hu-PBL-SCID) model has been developed to test anti-T-cell monoclonal antibody (mAb) effects on systemic symptoms of the host and the survival of human skin grafts. To obtain consistent engraftment without lethal acute graft-versus-host disease (GVHD), SCID mice were pretreated with a combination of total body irradiation (2.5 Gy, day 0) and anti-asialo GM1 (anti-mouse natural killer cell) antiserum (50 micrograms i.p., day 3) before the intraperitoneal injection of 40-50 X 10(6) human PBL on day 4. With this protocol, the engraftment rate was 82% with 5-98% human CD45-positive cells in the peripheral blood. Mortality at 30 days was 0% in the mice bearing 5-50% human cells compared with 70% in those with more than 50%. Using hu-PBL-SCID mice with 5-50% human cells in their peripheral blood, we demonstrated the following results: 1) Human T cells isolated from these mice proliferated in response to immobilized OKT3 stimulation in vitro. 2) Hu-PBL-SCID mice but not normal SCID mice were able to reject human skin grafts in vivo 16-21 days after grafting. 3) Both OKT3 (anti-human CD3 mAb) and T10B9 (anti-human alpha beta T-cell receptor mAb) treatment prevented human skin graft rejection in hu-PBL-SCID mice. 4) OKT3 but not T10B9 induced first dose reactions characterized by hypothermia and hypoactivity which were consistently observed within 90 min of intravenous injection into hu-PBL-SCID mice. 5) Human cytokines were detected in the serum of the hu-PBL-SCID mice treated with anti-T-cell mAbs. The close similarity of these responses to human clinical mAb immunosuppressive therapy suggests that the hu-PBL-SCID mouse model may be an excellent tool for investigating the immunosuppression, side effects, and mechanism of action of agents that are specific for human and higher apes and not reactive with lower animals.

    Topics: Acute Disease; Animals; Antibodies, Monoclonal; Cell Division; Cytokines; Disease Models, Animal; G(M1) Ganglioside; Graft Survival; Graft vs Host Disease; Hematopoietic Stem Cell Transplantation; Humans; Hypothermia; Immune Sera; Immunoglobulin M; Immunosuppressive Agents; Leukocytes; Lymphocyte Activation; Mice; Mice, SCID; Muromonab-CD3; Receptors, Antigen, T-Cell, alpha-beta; Skin Transplantation; Survival Rate; T-Lymphocytes; Transplantation, Heterologous; Whole-Body Irradiation

1997
Effects of monosialoganglioside on a new model of tardive dyskinesia.
    Progress in neuro-psychopharmacology & biological psychiatry, 1997, Volume: 21, Issue:7

    1- The effects of monosialoganglioside GM1 were studied on a new model of tardive dyskinesia, i.e., the frequency of spontaneous tongue protrusions in rats repeatedly treated with reserpine. 2- Rats were co-treated with vehicle (VEH) or reserpine (RES) (0.1 mg/kg, s.c., every other day) and saline (SAL) or GM1 (5 mg/kg, i.p., every day) for 30 days and observed for tongue protrusions on days 10, 20 and 30. 3- During each test day animals of the RES + SAL group exhibited an increase in tongue protrusions relative to rats of the VEH + SAL group. However, rats of the RES + GM1 group showed an increased frequency of tongue protrusions only on day 10, when compared to animals of the VEH + SAL group. There were no significant differences in tongue protrusion frequency between the VEH + GM1 and the VEH + SAL groups. 4- These results differ from previous studies which reported a facilitatory effect of GM1 co-administration on conventional behavioral animal models of tardive dyskinesia. The possibility is raised that GM1 attenuates the reserpine-induced increase in tongue protrusions through its protective effect on glutamate/oxidative stress neurotoxicity.

    Topics: Animals; Anti-Inflammatory Agents, Non-Steroidal; Antipsychotic Agents; Disease Models, Animal; Dyskinesia, Drug-Induced; Free Radicals; G(M1) Ganglioside; Male; Motor Activity; Oxidative Stress; Random Allocation; Rats; Rats, Wistar; Receptors, Dopamine; Reserpine; Tongue

1997
Monosialoganglioside GM1 reduces ischemia--reperfusion-induced injury in the rat retina.
    Investigative ophthalmology & visual science, 1996, Volume: 37, Issue:2

    Gangliosides are normal components of cell membranes, contribute to structural rigidity and membrane function, and have been shown to protect against various insults to the brain. This study evaluates the effect of exogenously administered monosialoganglioside GM1 on retinal damage induced by transient retinal ischemia and reperfusion.. Retinal ischemia was induced unilaterally in Long Evans rats by increasing intraocular pressure to 160 mm Hg for 60 minutes. GM1 (30 mg/kg, intraperitoneally) or buffer controls were administered at 48 hours, and 15 minutes before ischemia, and survival time after ischemia was either 8 or 15 days. The degree of retinal damage was assessed by histopathologic study according to Hughes' quantification of ischemic damage.. Retinal ischemia led to significant reductions in thickness and cell number, principally in the inner retinal layers (30% to 80%), and to a lesser extent in the outer retinal layers (18% to 42%). Pretreatment with intraperitoneally injected monosialoganglioside GM1 conferred significant protection against retinal ischemic damage either 8 or 15 days after ischemic survival time. After 8 days reperfusion, the ischemic-induced loss in overall retinal thickness was reduced by 70%, and those of the inner nuclear and plexiform layers were reduced by 77% and 44%, respectively. Ischemic-induced ganglion cell, inner nuclear, and outer nuclear layer cell density losses were reduced by 45%, 40%, and 57%, respectively. After 15 days of reperfusion, approximately the same statistically significant differences could be observed in comparison with the 15-day ischemic--reperfusion group.. Monosialoganglioside GM1 protects the rat retina from pressure-induced ischemic injury when administered intraperitoneally 2 days before insult. This protection afforded by GM1 can be observed even after 8 days or 15 days of reperfusion.

    Topics: Animals; Apoptosis; Cell Count; Cell Death; Disease Models, Animal; G(M1) Ganglioside; Injections, Intraperitoneal; Ischemia; Rats; Reperfusion; Reperfusion Injury; Retina; Retinal Artery; Retinal Diseases

1996
Dextran sulfate sodium (DSS) induced experimental colitis in immunodeficient mice: effects in CD4(+) -cell depleted, athymic and NK-cell depleted SCID mice.
    Inflammation research : official journal of the European Histamine Research Society ... [et al.], 1996, Volume: 45, Issue:4

    Administration of dextran sulfate to mice, given in the drinking water results in acute or subacute colonic inflammation, depending on the administration protocol. This colonic inflammation exhibits ulceration, healing and repair, and a therapeutic response that makes it valuable for the study of mechanisms that could act in the pathogenesis of human ulcerative colitis, a disease thought to have an immunologically dependent pathogenesis. To investigate if immunological mechanisms were involved in the induction of colonic inflammation in this model, mice with different degrees of immunodeficiency were used. It was shown that dextran sulfate induced colitis could be induced in Balb/c mice depleted of CD4(+) helper T cells by treatment with monoclonal antibodies preceded by adult thymectomy. The depletion of CD4(+) was verified by flow cytometric analysis. Furthermore, the colonic inflammation could equally be induced in athymic CD-1 nu/nu mice lacking thymus-derived T cells, in T and B-cell deficient SCID mice, and also in SCID mice depleted of NK cells by treatment with anti-asialo GM1 antibodies. The NK-cell depletion was verified by measuring spleen NK-cell activity. The resulting colonic inflammation in all these types of deficient mice was qualitatively comparable, as shown by clinical and histological appearance. These results indicate that the presence of functional T, B and NK cells is not crucial for the induction of dextran sulfate colitis in mice.

    Topics: Administration, Oral; Analysis of Variance; Animals; Antibodies, Monoclonal; Antiviral Agents; B-Lymphocytes; Body Weight; Colitis, Ulcerative; Colon; Dextran Sulfate; Disease Models, Animal; Female; Flow Cytometry; Fluorescent Antibody Technique, Direct; G(M1) Ganglioside; Immunoglobulin G; Killer Cells, Natural; Lymphoid Tissue; Mice; Mice, Inbred BALB C; Mice, Nude; Mice, SCID; Organ Size; Specific Pathogen-Free Organisms; T-Lymphocytes, Helper-Inducer; Thymectomy

1996
Novel metastasis model of human lung cancer in SCID mice depleted of NK cells.
    International journal of cancer, 1996, Jul-17, Volume: 67, Issue:2

    Metastasis is a critical problem in the treatment of human lung cancer. Thus, a suitable animal model of metastasis of human lung cancer is required for in vivo biological and preclinical studies. In this study, we tried to establish a suitable model for this, using SCID mice. Neither human SCLC H69/VP cells (5 x 10(6)) nor squamous-cell carcinoma RERF-LC-AI cells (1 x 10(6)), injected through a tail vein, formed metastases in untreated SCID mice. Pre-treatment of SCID mice with anti-asialo GM1 serum resulted in only a few metastases of H69/VP cells, but pre-treatment with anti-mouse IL-2 receptor beta chain Ab (TM-beta 1) resulted in numerous lymph-node metastases 56 days after tumor inoculation. H69/VP-M cells, an in vivo-selected variant line, formed significant numbers of lymph-node metastases even in SCID mice pre-treated with anti-asialo GM1 serum. SCID mice depleted of NK cells by treatment with TM-beta 1 showed different patterns of metastasis when inoculated intravenously with the 2 different human lung cancer cell lines (H69/VP and RERF-LC-AI cells): H69/VP cells formed metastases mainly in systemic lymph nodes and the liver, whereas RERF-LC-AI cells formed metastases mainly in the liver and kidneys, with only a few in lymph nodes. A histopathological study showed that the metastatic colonies consisted of cancer cells. The numbers of metastatic colonies formed by the 2 cell lines increased with the number of cells inoculated. TM-beta 1 treatment of SCID mice efficiently removed NK cells from peripheral blood for at least 6 weeks, whereas, after treatment of the mice with anti-asialo GM1 serum, NK cells were recovered within 9 days. These findings suggest that NK-cell-depleted SCID mice may be useful as a model in biological and pre-clinical studies on metastasis of human lung cancer.

    Topics: Animals; Antibodies; Disease Models, Animal; G(M1) Ganglioside; Humans; Immune Sera; Killer Cells, Natural; Liver Neoplasms; Lung Neoplasms; Lymphatic Metastasis; Male; Mice; Mice, SCID; Neoplasm Metastasis; Receptors, Interleukin-2; Tumor Cells, Cultured

1996
Inability to establish ectopic endometrium in a natural killer cell-deficient murine model. Immunologic, histologic and histochemical assessment.
    The Journal of reproductive medicine, 1996, Volume: 41, Issue:11

    To investigate what effect natural killer (NK) cells have on the implantation of heterologous endometrial scrapings.. Anti-asialo GM1 (AA-GM1) anti-sera have been shown to eliminate NK cell activity in various strains of rats and mice. Either AA-GM1 antibodies (+) or rabbit antiglobulin (-) was administered to beige mice (NK cell deficient) or beige control mice (not NK cell deficient of the same strain). The heterologous endometrial scrapings were prepared by scraping seven pairs of uterine horns from normal mice of the same strain. Beige and normal mice were then injected intraperitoneally every 3 days with the heterologous endometrial scraping and antibodies for a period of 50 days. The four experimental groups (n = 10 per group) can be summarized as being beige (+), beige (-), normal (+) and normal (-).. There was no evidence of ectopic endometrial tissue in any of the four test groups by histologic examination or by using immunohistochemical staining techniques. Histologic evidence of an impaired immune response was clearly demonstrated in the beige mice receiving AA-GM1 antibodies.. Using this model, a deficiency of NK cell activity did not appear to enhance the implantation of endometrial tissue on the abdominal peritoneum of mice.

    Topics: Animals; Antibodies; Disease Models, Animal; Endometriosis; Endometrium; Female; G(M1) Ganglioside; Glycoproteins; Humans; Immunohistochemistry; Immunosuppression Therapy; Intermediate Filament Proteins; Killer Cells, Natural; Mice; Mice, Inbred C57BL; Peritoneum; Serpins; Transplantation, Heterotopic

1996
Antiviral activity of biological response modifiers in a murine model of AIDS. Requirement for augmentation of natural killer cell activity and synergy with oral AZT.
    International journal of immunopharmacology, 1996, Volume: 18, Issue:11

    We employed the Rauscher murine leukemia virus (RMuLV) as a murine retrovirus model of AIDS, to test biological response modifiers (BRM) and antiviral agents for potential therapeutic activity against the human immunodeficiency virus (HIV). We examined the relationship between the augmentation of natural killer (NK) cell activity and antiviral efficacy of a series of BRM, most of which are known inducers of interferon, in this model. Poly [I,C]-LC, MVE-2, and CL 246,738, but not Ampligen, soluble glucan, or 7-thia-8-oxoguanosine, consistently produced antiviral activity. In addition, the combination of suboptimal doses of oral 3'-azido-3'-deoxythymidine (AZT) (in drinking water) and poly [I,C]-LC produced a synergistic antiviral effect. With all the BRM tested, a consistent pattern emerged, namely that antiviral activity always correlated with the augmentation of splenic NK cell activity in infected animals. For instance, poly [I,C]-LC boosted NK activity much more in infected mice treated therapeutically (treatment initiated after infection) than prophylactically (treatment initiated before infection), and it had greater antiviral activity therapeutically than prophylactically. For the BRM tested, antiviral activity did not occur without augmentation of NK activity in infected mice. In contrast, augmentation of NK activity in uninfected mice bore no relationship to antiviral activity. Furthermore, elimination of NK cells by treating mice with anti-asialo GM1 abolished the antiviral activity of poly [I,C]-LC. Although splenic NK activity was ablated by anti-asialo GM1, serum interferon levels were not affected by this treatment. These results point to a causal connection between the augmentation of NK cell activity and the antiviral efficacy of these BRM in this murine AIDS model. NK cells thus appear to play a key role in resistance to this retrovirus, as has been suggested for HIV.

    Topics: Acquired Immunodeficiency Syndrome; Acridines; Animals; Antibodies; Antiviral Agents; Asialoglycoproteins; Disease Models, Animal; Drug Synergism; Female; G(M1) Ganglioside; Glucans; Guanosine; Immunologic Factors; In Vitro Techniques; Killer Cells, Natural; Lymphocyte Depletion; Mice; Mice, Inbred BALB C; Poly I-C; Poly U; Pyran Copolymer; Rabbits; Rauscher Virus; Specific Pathogen-Free Organisms; Viral Plaque Assay; Zidovudine

1996
Stress-triggered abortion: inhibition of protective suppression and promotion of tumor necrosis factor-alpha (TNF-alpha) release as a mechanism triggering resorptions in mice.
    American journal of reproductive immunology (New York, N.Y. : 1989), 1995, Volume: 33, Issue:1

    Stress adversely affects pregnancy outcome and has been implicated as an abortogen in both animals and humans. However, the mechanisms whereby stress aborts are largely unknown. Alloimmunization can prevent stress-triggered abortion, and immunization is known to increase transforming growth factor-beta 2 (TGF-beta 2)-related suppressive activity.. To investigate these mechanisms, DBA/2J males were mated to CBA/J or C3H/HeJ females, and the pregnant females were exposed to ultrasonic sound stress for a period of 24 h between day 4.5 to 8.5 of pregnancy.. Ultrasonic stress significantly elevated the resorption rate with a peak effect on day 5.5 in the CBA/J females and on day 4.5 in the LPS-resistant C3H/HeJ females. The tumor necrosis factor-alpha (TNF-alpha) release from the decidua was also elevated and the TGF-beta 2-mediated suppressive activity was significantly decreased. The resorption rate only increased when the TNF-alpha/TGF-beta 2 ratio was increased compared to the control.. These data suggest that stress may inhibit protective suppressor mechanisms and promote secretion of abortogenic cytokines such as TNF-alpha. Possible mechanisms are discussed.

    Topics: Abortion, Spontaneous; Animals; Decidua; Disease Models, Animal; Female; Fetal Resorption; G(M1) Ganglioside; Immunization; Isoantigens; Male; Mice; Mice, Inbred Strains; Noise; Pest Control; Pregnancy; Pregnancy Complications; Psychoneuroimmunology; Stress, Physiological; Transforming Growth Factor beta; Tumor Necrosis Factor-alpha; Ultrasonics

1995
Limitations of the severe combined immunodeficiency (SCID) mouse model for study of human B-cell responses.
    Scandinavian journal of immunology, 1995, Volume: 41, Issue:4

    Mice lacking functional T and B lymphocytes offer an in vivo animal model for the study of human immune functions. We have attempted to optimize the reconstitution of severe combined immunodeficiency (SCID) mice with human peripheral blood lymphocytes (PBL) using radiation, anti-asialo GM1 antibody or cyclophosphamide (Cy) treatment of the mice and in vitro stimulation of human PBL with interleukin (IL)-2 prior to their transfer to the mice. Total human IgG and tetanus-toxoid (TT)-specific human IgG responses of the mice were used as parameters of successful reconstitution. Treatment of the mice with anti-asialo GM1 antibody significantly enhanced total human IgG levels, but not TT-specific antibody responses, whereas irradiation or Cy treatment of the mice had no effect on human antibody production. In vitro treatment of human PBL with IL-2 prior to engraftment significantly decreased total human IgG responses of human PBL-grafted SCID mice. The immune responses of individual mice within a group were highly variable, which constitutes a major disadvantage of this model.

    Topics: Animals; Antibodies; B-Lymphocytes; Cyclophosphamide; Disease Models, Animal; G(M1) Ganglioside; Humans; Immunoglobulin G; Interleukin-2; Mice; Mice, SCID; Recombinant Proteins; Severe Combined Immunodeficiency; Tetanus Toxoid; Whole-Body Irradiation

1995
GM1 ganglioside rescues substantia nigra pars compacta neurons and increases dopamine synthesis in residual nigrostriatal dopaminergic neurons in MPTP-treated mice.
    Journal of neuroscience research, 1995, Sep-01, Volume: 42, Issue:1

    GM1 ganglioside has been shown to stimulate recovery of the damaged dopamine system under a number of different circumstances. In addition to rescue of damaged dopamine neurons, the present study assessed the ability of GM1 to enhance the synthesis of dopamine in remaining nigrostriatal neurons following 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) exposure. There was a significantly greater accumulation of L-dopa 30 min after aromatic amino acid decarboxylase inhibition with NSD-1015 (100 mg/kg) and an increase in the ratio of L-dopa to dopamine in MPTP+GM1-treated mice than in mice that received only MPTP. This effect of GM1 on dopamine synthesis was dependent upon the degree of initial damage to the nigrostriatal dopamine system. That is, the GM1 effect on dopamine synthesis could not be demonstrated in mice with greater than 95% striatal dopamine loss and 75% substantia nigra dopamine neuron loss. These results suggest that in addition to previously reported effects of GM1 on rescue and repair of dopaminergic neurons, GM1 may also have the ability to enhance dopamine synthesis in residual dopaminergic neurons. Direct effects on dopamine neurochemistry may contribute to functional improvement seen after GM1 treatment in various models of parkinsonism.

    Topics: 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine; Animals; Corpus Striatum; Disease Models, Animal; Dopamine; G(M1) Ganglioside; Levodopa; Mice; Mice, Inbred C57BL; Neurons; Parkinson Disease; Substantia Nigra

1995
[Facilitation of hepatic metastasis by surgical stress in mice].
    Nihon Rinsho Men'eki Gakkai kaishi = Japanese journal of clinical immunology, 1995, Volume: 18, Issue:5

    An experimental model for hepatic metastasis with a transplantation route of free-pedicled subcutaneous-embedded spleen was established in BALB/c mice. Colon-26 tumor cells to produce hepatic metastasis were inoculated into the spleen and the influence of surgical stress by means of a 20-min exposure of the abdominal cavity on the incidence of hepatic metastasis was examined. Hepatic metastasis was more promoted by the surgical stress in order when it was given on the same day, the 7th day and the 3rd day of the inoculation. Administration, without surgical stress, of ASGM 1, a specific inhibitor of the natural killer activity, also facilitated the hepatic disease. Administration of OK-432 prior to the surgical stress or ASGM 1 was at least partly effective for prevention of the hepatic metastasis and prolonged the survival of the inoculated mice. Preoperative immunotherapy utilizing OK-432 might be a possible means to prevent hepatic metastasis triggered in colorectal surgery for cancer.

    Topics: Animals; Antibodies; Antineoplastic Agents; Colonic Neoplasms; Cytotoxicity, Immunologic; Disease Models, Animal; G(M1) Ganglioside; Killer Cells, Natural; Liver Neoplasms; Male; Mice; Mice, Inbred BALB C; Neoplasm Transplantation; Neoplastic Cells, Circulating; Picibanil; Stress, Physiological; Surgical Procedures, Operative

1995
Auditory brainstem responses of CBA/J mice with neonatal conductive hearing losses and treatment with GM1 ganglioside.
    Hearing research, 1995, Volume: 87, Issue:1-2

    Exogenous administration of GM1 ganglioside to CBA/J mice with a neonatal conductive hearing loss ameliorates the atrophy of spiral ganglion neurons, ventral cochlear nucleus neurons, and ventral cochlear nucleus volume. The present investigation demonstrates the extent of a conductive loss caused by atresia and tests the hypothesis that GM1 ganglioside treatment will ameliorate the conductive hearing loss. Auditory brainstem responses were recorded from four groups of seven mice each: two groups received daily subcutaneous injections of saline (one group had normal hearing; the other had a conductive hearing loss); the other two groups received daily subcutaneous injections of GM1 ganglioside (one group had normal hearing; the other had a conductive hearing loss). In mice with a conductive loss, decreases in hearing sensitivity were greatest at high frequencies. The decreases were determined by comparing mean ABR thresholds of the conductive loss mice with those of normal hearing mice. The conductive hearing loss induced in the mice in this study was similar to that seen in humans with congenital aural atresias. GM1 ganglioside treatment had no significant effect on ABR wave I thresholds or latencies in either group.

    Topics: Acoustic Stimulation; Animals; Auditory Threshold; Cochlear Nucleus; Disease Models, Animal; Evoked Potentials, Auditory, Brain Stem; G(M1) Ganglioside; Hearing Loss, Conductive; Mice; Mice, Inbred CBA; Neurons; Spiral Ganglion; Tympanic Membrane

1995
The B cell repertoire in experimental allergic neuritis involves multiple myelin proteins and GM1.
    Journal of the neurological sciences, 1994, Volume: 125, Issue:2

    Experimental allergic neuritis (EAN) is a T cell mediated disease associated with inflammation and demyelination of peripheral nerves. EAN is an experimental model of Guillain-Barré syndrome. The peripheral nerve myelin components P2 and P0 represent major neuritogens, but the diversity and quantity of B cell responses in EAN are unknown. Lewis rats were immunized with bovine peripheral nerve myelin (BPM), and levels of B cells secreting IgM and IgG antibodies to BPM, P2 and P0, the glycolipid GM1 and five peptides of myelin-associated glycoprotein (MAG) were determined. Already on day 7 post-immunization (p.i.), i.e. before the onset of clinical EAN, lymph nodes contained elevated levels of cells secreting IgM antibodies of all specificities examined. Maximum numbers of IgG antibodies secreting cells were generally reached at the height of clinical disease. The numbers of cells secreting IgG antibodies to BPM, P2, P0, GM1 and MAG peptides were also elevated before disease onset, but they were mostly higher than those of IgM antibodies and they reached their maximum only after recovery. The results imply that EAN is associated with strong B cell responses to all myelin antigens under study without restriction to any immunodominant myelin component or MAG peptides.

    Topics: Amino Acid Sequence; Animals; Autoantibodies; Autoimmune Diseases; B-Lymphocytes; Cattle; Disease Models, Animal; G(M1) Ganglioside; Immunization; Immunoglobulin G; Immunoglobulin M; Lymph Nodes; Male; Molecular Sequence Data; Myelin Basic Protein; Myelin P0 Protein; Myelin P2 Protein; Myelin Proteins; Myelin Sheath; Myelin-Associated Glycoprotein; Neuritis, Autoimmune, Experimental; Peptide Fragments; Polyradiculoneuropathy; Rats; Rats, Inbred Lew; Spleen

1994
The role of natural killer cells in resistance to coccidiosis: investigations in a murine model.
    Clinical and experimental immunology, 1994, Volume: 97, Issue:2

    Natural killer (NK) activity, detected by the lysis of Yac-1 target cells, was examined in splenic and mesenteric lymph node (MLN) cells throughout the course of infection with Eimeria vermiformis in BALB/c and C57B1/6 (B6) mice. These strains are, respectively, relatively resistant and susceptible to primary infections, which render them equally, and completely, resistant to challenge. Resting levels of NK activity were higher in B6 than in BALB/c, and B6 responded earlier in the course of infection than BALB/c, but splenic peak values were higher in BALB/c; the pattern of response in MLN cells was similar in both strains, but the peak was higher in BALB/c. At the time (7 days p.i.) of peak NK response in BALB/c mice there was, depending upon the choice of NK-resistant/lymphokine-activated killer (LAK)-sensitive target cells, either little (P388D1), or no (P815) splenic LAK activity. Challenge of immunized BALB/c mice did not evoke a detectable NK response. Although the higher NK activity in BALB/c mice correlated with greater control of primary infection, depletion of NK activity (demonstrated in splenic cells) in vivo by treatment with anti-asialo GM1 antibodies did not greatly affect the course of infection. Furthermore, this treatment did not augment the exacerbation of infection produced by treatment with anti-interferon-gamma (IFN-gamma) MoAb, indicating that, at least in this system, NK cells are not a fundamentally important source of this controlling cytokine of eimerian infections. The results suggest that NK cells may not greatly influence the outcome of coccidial infections.

    Topics: Animals; Coccidiosis; Disease Models, Animal; Eimeria; Female; G(M1) Ganglioside; Immunity, Innate; Interferon-gamma; Killer Cells, Lymphokine-Activated; Killer Cells, Natural; Mice; Mice, Inbred BALB C; Mice, Inbred C57BL

1994
The ganglioside GM1 decreases autotomy but not substance P depletion in a peripheral mononeuropathy rat model.
    Anesthesia and analgesia, 1993, Volume: 77, Issue:3

    The effect of the ganglioside GM1 on autotomy, a nociceptive behavioral marker for neuropathic pain, and substance P depletion was determined in a rat model of peripheral mononeuropathy, sciatic cryoneurolysis (SCN). SCN is produced by the application of a cryoprobe to the common sciatic nerve using a freeze-thaw-freeze cycle. Due to structural sparing of the nerve, regenerative processes are not precluded. After this peripheral nerve insult, behavioral and neurochemical changes occur that support the use of SCN as a neuropathic pain model. These changes include: autotomy with coincident transient weight loss and paling of eye color suggestive of increased sympathetic activity, spontaneous nociceptive behaviors, touch-evoked allodynia, prolonged mechanical allodynia, ipsilateral decrease of immunoreactive substance P, and increases in spinal cord dynorphin expression. Incidence and severity of autotomy were assessed after the intraperitoneal administration of GM1 (1, 10, and 20 mg/kg) or saline injected daily for 2 days before SCN, the day of surgery, and for 14 days after surgery. In a subset of two rats from each treatment group, transcardiac perfusion was performed and spinal cords were processed for substance P immunoreactivity. GM1 at 10 and 20 mg/kg doses significantly attenuated autotomy as compared with saline-treated rats (P = 0.007 and 0.0001, respectively). However, GM1, at the doses studied, failed to alter the spinal substance P depletion 21 days after SCN. These results indicate that the ganglioside GM1 may have a role in the clinical management of neuropathic pain after peripheral nerve injury.

    Topics: Animals; Behavior, Animal; Disease Models, Animal; G(M1) Ganglioside; Male; Pain; Peripheral Nervous System Diseases; Rats; Rats, Sprague-Dawley; Sciatic Nerve; Self Mutilation; Spinal Cord; Substance P

1993
Serologic responses to nerve antigens in sooty mangabey monkeys with experimental leprosy.
    International journal of leprosy and other mycobacterial diseases : official organ of the International Leprosy Association, 1993, Volume: 61, Issue:2

    Eight sooty mangabey monkeys were inoculated intravenously and intradermally with varying doses of Mycobacterium leprae from 4.8 x 10(7) to 4.8 x 10(10). Serum samples were obtained from the animals at intervals of about 3 months for 90 months, and were examined for IgM and IgG antibodies to nerve antigens, including ceramide, galactocerebroside (GC), and asialo-GM1 (AGM1), using an enzyme-linked immunosorbent assay (ELISA). The serological results were then compared with clinical findings, particularly nerve involvement. Of 8 mangabey monkeys inoculated with M. leprae, 7 animals had clinical leprosy; 6 of them had nerve damage, including neurologic deformities in 4 monkeys and nerve enlargement in 2. Median time for the initial signs of leprosy was 10 months postinoculation (p.i.), a range from 4 to 35 months. In contrast, nerve damage was noted rather late, about 35 to 86 months p.i. (median 54 months). The major immunoglobulin class to ceramide, GC, and AGM1 antigens was IgM, and the antibody responses to the nerve antigens appeared from 15 to 63 months p.i. (median 37 months). Antineural antibodies were thus detectable about 18 months (range -2 to 60 months) prior to observable nerve damage. In addition, elevation of antineural antibody levels were predictive of clinical exacerbation of the disease and neuritic damage. This study suggests that antineural antibodies are produced during the course of M. leprae infection and may be indicative of nerve damage, such as neurological deformities or nerve enlargement, in leprosy patients.

    Topics: Animals; Autoantigens; Brain Diseases; Ceramides; Cercocebus atys; Disease Models, Animal; Enzyme-Linked Immunosorbent Assay; G(M1) Ganglioside; Galactosylceramides; Immunoglobulin G; Immunoglobulin M; Leprosy, Lepromatous; Mycobacterium leprae; Nerve Tissue Proteins

1993
Antitumor effect of thymosin alpha 1/interleukin-2 or thymosin alpha 1/interferon alpha,beta following cyclophosphamide in mice injected with highly metastatic Friend erythroleukemia cells.
    Journal of immunotherapy with emphasis on tumor immunology : official journal of the Society for Biological Therapy, 1993, Volume: 13, Issue:1

    We investigated the effects of the systemic administration of thymosin alpha 1 plus relatively low doses of human recombinant interleukin-2 or very low doses of interferon alpha,beta in untreated and cyclophosphamide (CY)-treated DBA/2 mice challenged either subcutaneously or intravenously (i.v.) with Friend erythroleukemia cells (FLC). Both treatments resulted in the complete regression of subcutaneous tumor and cured a significative percentage of mice. They also increased the survival time of mice i.v. injected with large numbers of FLC. Neither immunotherapy alone nor CY, alone or in combination with single cytokines, produced similar effects. The antitumor action of these combined chemoimmunotherapy protocols seems to involve activation of the immune response since (a) a synergistic increase of the cytotoxicity of spleen cells was demonstrated in treated mice; (b) selective in vivo depletion of asialo-GM1, CD4, or CD8-positive cells abrogated this antitumor activity; and (c) a high lymphoid cell infiltration was found at the tumor site and in the livers of treated mice.

    Topics: Animals; Antineoplastic Combined Chemotherapy Protocols; CD4 Antigens; CD8 Antigens; Cyclophosphamide; Cytotoxicity, Immunologic; Disease Models, Animal; Friend murine leukemia virus; G(M1) Ganglioside; Interferon-alpha; Interferon-beta; Interleukin-2; Leukemia, Erythroblastic, Acute; Leukemia, Experimental; Male; Mice; Mice, Inbred DBA; Thymalfasin; Thymosin; Tumor Virus Infections

1993
Generation and control of metastasis in experimental tumor systems; inhibition of experimental metastases by a tilorone analogue.
    International journal of cancer, 1993, May-28, Volume: 54, Issue:3

    The role of the chemical compound RMI 10,874DA (3,6-bis[2-(dimethylamino)-ethoxyl]-9H-xanthene-9-one dihydrochloride) in the abrogation of the metastatic spread of tumor cells was studied. Pre-treatment of BALB/c mice with the RMI 10,874DA compound (referred to below as tilorone analogue) completely eliminated lung colonization of an H-2-negative (GR9.B9) MCA-induced fibrosarcoma clone in an experimental metastasis assay. Other murine tumors, including H-2-positive and H-2-negative chemically induced fibrosarcoma clones and B16 melanoma, were also sensitive to the treatment; orally administered tilorone analogue given one day before the i.v. injection of tumor cells markedly inhibited lung colonization. The effect was not due to direct toxicity of tilorone analogue on tumor cells, but instead it was dependent on NK cells; this was suggested by the finding that anti-asialo GM, treatment of mice abrogated the effect of tilorone analogue. Kinetic studies of splenic NK activity in tilorone-treated mice showed a rapid boosting of NK-cell activity, the greatest stimulation occurring the day before removal of splenocytes for 51Cr-release assay against YAC-I target cells. These kinetics correlated with the inhibition of in vivo lung colonization after tilorone analogue treatment. Inhibition of experimental tumor metastasis was dose-dependent and was observed when animals were treated the day before or the day after tumor-cell injection. Furthermore, repeated treatment of mice with this tilorone analogue significantly reduced lung colonization.

    Topics: Animals; Antineoplastic Agents; Disease Models, Animal; Dose-Response Relationship, Drug; G(M1) Ganglioside; Immune Sera; Killer Cells, Natural; Lung Neoplasms; Mice; Mice, Inbred BALB C; Mice, Inbred C57BL; Neoplasm Metastasis; Neoplasms, Experimental; Spleen; Tilorone; Time Factors; Xanthenes; Xanthones

1993
GM1 reduces injury-induced metabolic deficits and degeneration in the rat optic nerve.
    Investigative ophthalmology & visual science, 1992, Volume: 33, Issue:13

    This study demonstrates the earliest reported effects of GM1 treatment on crush-injured axons of the mammalian optic nerve. GM1, administered intraperitoneally immediately after injury, was found to reduce the injury-induced metabolic deficit in nerve activity within 2 hr of injury, as measured by changes in the nicotine-amine adenine dinucleotide redox state. After 4 wk, transmission electron microscopy 1 mm distal to the site of injury revealed a sevenfold increase in axonal survival in GM1-treated compared to untreated injured nerves. These results emphasize the beneficial effect of GM1 on injured optic nerves as well as the correlation between immediate and long-term consequences of the injury. Thus, these results have implications for treating damaged optic nerves.

    Topics: Animals; Axons; Cell Count; Cell Survival; Disease Models, Animal; G(M1) Ganglioside; Injections, Intraperitoneal; Male; NAD; Nerve Degeneration; Optic Nerve; Optic Nerve Injuries; Rats; Rats, Sprague-Dawley; Spectrometry, Fluorescence

1992
[Immunological mechanism of pregnancy and miscarriage--a study with a murine spontaneous miscarriage model].
    Nihon Sanka Fujinka Gakkai zasshi, 1992, Volume: 44, Issue:2

    In order to investigate the immunological mechanisms of pregnancy, fluorocytometric and immunohistochemical analysis of the cells was performed in the placenta and spleen of a murine spontaneous miscarriage model (CBA/J x DBA/2) and control (CBA/J x BALB/c). There was a significant difference between the miscarriage rate for the miscarriage model and that for the control, even though H-2 in these two group is matched. The analysis also was performed in a miscarriage model immunized with male splenocytes. Moreover, the effect of gamma-interferon, a potentiator of NK cell activity, on pregnancy was examined. Interferon treatment increased the miscarriage rate. In pregnancy, the number of splenocyte positive Asialo-GM1 or LFA-1 decreased and the intensity of these antigens decreased, as well. Interleukin-2R positive cell increased in number as well as intensity. In the miscarriage model group successfully treated by immunization, the number of Asialo-GM1 positive cells and L3T4 positive cells decreased, whereas they increased in the unsuccessfully treated group. Asialo-GM1 positive cells in the placenta of successful pregnancy decreased in number, and those in miscarried pregnancy increased. In conclusion, the success of the immunization treatment for habitual abortion depends on how to suppress NK cell activity in a linkage with the helper T-cell.

    Topics: Abortion, Habitual; Animals; Disease Models, Animal; Embryo Implantation; Female; G(M1) Ganglioside; Glycosphingolipids; Immunohistochemistry; Interferon-gamma; Interleukin-2; Killer Cells, Natural; Mice; Mice, Inbred BALB C; Mice, Inbred DBA; Placenta; Pregnancy; Pregnancy, Animal; T-Lymphocytes, Helper-Inducer

1992
Canine GM1-gangliosidosis. A clinical, morphologic, histochemical, and biochemical comparison of two different models.
    The American journal of pathology, 1992, Volume: 140, Issue:3

    The clinical, morphologic, histochemical, and biochemical features of GM1-gangliosidosis in two canine models, English Springer Spaniel (ESS) and Portuguese Water Dog (PWD), have been compared. The disease onset, its clinical course, and survival period of the affected dogs were similar in both models. Skeletal dysplasia was noted radiographically at 2 months of age, whereas at 4 1/2 months of age there was progressive neurologic impairment. However, dwarfism and coarse facial features were seen only in ESS. Both models had similar deficiency in activity of lysosomal beta-galactosidase, but possessed a normal protein activator for GM1-beta-galactosidase. Both models stored GM1-ganglioside, asialo-GM1, and oligosaccharides in brain. Furthermore, only the PWD stored glycoproteins containing polylactosaminoglycans in visceral organs, and neither model stored them in the brain. Morphologically, both models demonstrated similar storage material in multiple tissues and cell types. The ultrastructure of the storage material was cell-type specific and identical in both models. However, some differences in the lectin staining pattern were noted. Our clinical, biochemical, and histochemical findings indicate that PWD and ESS may represent two different mutations of the beta-galactosidase gene. Moreover, the authors conclude that it is difficult, and inappropriate, to apply the human classification of GM1-gangliosidosis (i.e. infantile, juvenile, and adult forms) to these canine models.

    Topics: Amniotic Fluid; Animals; Carbohydrate Metabolism; Disease Models, Animal; Dogs; G(M1) Ganglioside; Gangliosidoses; Lipid Metabolism; Microscopy, Electron; Placenta; Umbilical Cord

1992
An EEG analysis of drug effects after mild head injury in mice.
    Life sciences, 1992, Volume: 51, Issue:3

    An electroencephalographic (EEG) and behavioral model of head injury in unanesthetized, free moving mice has been used to test the effects of TRH and GM1. In our experimental conditions a mechanical head injury capable of inducing loss of righting reflex for 2 to 60 sec, also induces a consistent decrease of the total power of the spectrum of EEG and a decrease of the power of fast beta band (20-40 Hz) for at least 120 min. TRH, injected after trauma in dose of 10 mg/kg, caused improvement of EEG total power of the spectrum. GM1 in high (30 mg/kg) but not in low dose (5 mg/kg) caused more rapid restoration of both the total power and fast as well as slow beta band power. These results suggest that GM1 has favorable effects on post-concussive neurophysiological symptoms in head injured animals.

    Topics: Animals; Craniocerebral Trauma; Disease Models, Animal; Electroencephalography; G(M1) Ganglioside; Male; Mice; Thyrotropin-Releasing Hormone

1992
[Recovery of nigrostriatal and mesolimbic dopaminergic system following administration of ganglioside in MPTP-treated mice].
    No to shinkei = Brain and nerve, 1990, Volume: 42, Issue:11

    The effects of systemic injection of GM 1 ganglioside on dopaminergic (DA) nigrostriatal and mesolimbic system following 1-methyl-4-phenyl-1, 2,3,6-tetrahydropyridine (MPTP) have been studied in C 57 BL/6 mice. MPTP treatment (4 x 20 mg/kg i.p. given 12 hr apart) resulted in significant depletion of DA concentration in the major terminal fields of the nigrostriatal and mesolimbic DA systems, i.e. dorsal striatum, ventral striatum, nucleus accumbens and olfactory tubercle 5 weeks after treatment in young (2 month old) mice. In aging (12 month old) mice treated with MPTP, significant depletion of DA concentration was observed in the cell body regions, i.e. substantia nigra and ventral tegmental area in addition to the major terminal fields, suggesting that the effect of MPTP is more widespread in aging mice. Although GM 1 ganglioside treatment (30 mg/kg, i.p. daily for 5 weeks) partially restored DA concentration in every major terminal field in young mice, such an apparent recovery was not seen in aging mice. GM 1 ganglioside treatment also reduced the increased 3,4-dihydroxyphenylacetic acid (DOPAC)/DA ratio following MPTP injection in the striatum of young mice, but such an effect was not observed in aging mice. We conclude that DA nigrostriatal and mesolimbic system in aging mice demonstrates reduced regenerative capacity following MPTP depletion compared with young mice, and the beneficial effect of GM 1 ganglioside for the recovery of DA nigrostriatal and mesolimbic system neurons declines with age.

    Topics: 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine; Adrenergic Fibers; Age Factors; Animals; Corpus Striatum; Disease Models, Animal; Dopamine; G(M1) Ganglioside; Limbic System; Mice; Mice, Inbred C57BL; Parkinson Disease, Secondary; Substantia Nigra

1990
Reduction of acute graft-versus-host disease-related mortality and cytotoxic T lymphocyte induction after pretreatment of the recipient with anti-asialo GM1 antibody in the murine P-to-F1 model.
    Transplantation proceedings, 1987, Volume: 19, Issue:1 Pt 3

    Topics: Animals; Antibodies; Cytotoxicity, Immunologic; Disease Models, Animal; G(M1) Ganglioside; Glycosphingolipids; Graft vs Host Disease; Killer Cells, Natural; Mice; Mice, Inbred CBA; Mice, Inbred Strains; Reference Values; Spleen; T-Lymphocytes, Cytotoxic

1987
Increased acetylcholine synthesis and release in brains of cats with GM1 gangliosidosis.
    Journal of neurochemistry, 1986, Volume: 46, Issue:5

    Cholinergic processes were measured in motor cortex, hippocampus, and striatum of cats in the terminal stages of GM1 gangliosidosis and compared to those of control cats. The greatest difference observed was elevation in the rate of K+-stimulated release of acetylcholine (ACh) from brain slices prepared from affected cats. The K+-stimulated release of endogenous ACh was increased by 31-43% and of newly synthesized ACh by 19-80% in brain slices from different brain regions. All regions that were examined were affected but the greatest effects occurred in cortex. The rate of synthesis of ACh was elevated in cortical and hippocampal slices. Choline acetyltransferase activity in brain regions of cats with GM1 gangliosidosis was not significantly different from that in controls, whereas high-affinity choline transport in cortical synaptosomes was elevated. Muscarinic receptor binding sites were reduced in the cortex, hippocampus, and striatum of GM1 mutant cats, whereas the apparent affinity was not altered. These results indicate that there are major alterations of cholinergic function in the brains of cats with GM1 gangliosidosis.

    Topics: Acetylcholine; Animals; Biological Transport; Brain; Cats; Choline; Choline O-Acetyltransferase; Corpus Striatum; Disease Models, Animal; G(M1) Ganglioside; Gangliosidoses; Hippocampus; Motor Cortex; Potassium; Receptors, Muscarinic

1986
Natural killer (NK) cells and graft-versus-host disease (GVHD): no correlation between the NK cell levels and GVHD in the murine P----F1 model.
    Immunology, 1985, Volume: 54, Issue:1

    Graft-versus-host disease (GVHD) was induced in (CBA X C57BL/6) F1 mice by i.v. injection of 50 X 10(6) parental spleen cells. The GVHD induced an enhanced NK (anti-YAC-1) cytotoxicity during the first 2 weeks after the spleen cell transfusion. This cytotoxic activity was shown to be mediated by asialo GM1-positive, partially Thy-1-positive and nylon-wool (NW) non-adherent cells, thus being classical NK cells. Depletion of NK-cell activity from donor and/or recipient mice with anti-asialo GM1 antibody prior to the spleen cell transfer did not prevent the GVHD as judged by the splenomegaly assay. Also, when NK activity was potentiated with polyinosinic-polycytidylic acid (pIC), no effect on the GVHD was seen. These data suggest that NK cells are not crucial for the development of GVHD in this model.

    Topics: Animals; Antibodies; Cytotoxicity, Immunologic; Disease Models, Animal; G(M1) Ganglioside; Glycosphingolipids; Graft vs Host Disease; Killer Cells, Natural; Leukocyte Count; Male; Mice; Poly I-C

1985
GM1 gangliosidosis, type 2: ocular clinicopathologic correlation.
    Graefe's archive for clinical and experimental ophthalmology = Albrecht von Graefes Archiv fur klinische und experimentelle Ophthalmologie, 1984, Volume: 222, Issue:2

    The clinical and pathological manifestations of a case of juvenile GM1 gangliosidosis are presented and the pathological findings compared with those previously reported for GM1 gangliosidosis in man and in animal models. The most striking finding in the present case was the marked degeneration of the retinal ganglion cell and nerve fiber layers. Although such extensive ganglion cell loss was not observed in any of the other cases reviewed, the presence of multimembranous inclusion bodies in retinal ganglion cells strongly suggests that the pathological process was similar in all cases. Much remains to be learned about the function of gangliosides in the healthy retina and about the pathophysiological consequences of deranged ganglioside metabolism. The many parallels, including those observed in pathological studies, between the human and animal forms of GM1 gangliosidosis allow an optimistic appraisal of the value of further research using the animal models.

    Topics: Animals; Atrophy; Cats; Cattle; Conjunctiva; Cornea; Disease Models, Animal; Dogs; Epithelium; Eye; Eye Diseases; Female; G(M1) Ganglioside; Gangliosidoses; Humans; Infant; Retina; Retinal Ganglion Cells

1984
Neurotransmitter chemistry in feline GM1 gangliosidosis: a model for human ganglioside storage disease.
    Annals of neurology, 1982, Volume: 12, Issue:1

    Assays for synaptosomal high-affinity uptake activity (glutamate, gamma-aminobutyric acid, norepinephrine), neurotransmitter synthesizing enzymes (choline acetyltransferase, glutamate decarboxylase, tyrosine hydroxylase), and endogenous neurotransmitters were performed in cats with advanced inherited GM1 gangliosidosis. A significant reduction in uptake activity, ranging from 24 to 77% of control, was demonstrated in motor, occipital, and cerebellar brain regions. This reduction was unassociated with comparable alterations in neurotransmitter levels or synthesizing enzyme activity. We hypothesize that the defect of neurotransmitter inactivation is part of an overall abnormality of synaptic membrane function that could contribute to the neurological symptoms seen in the hereditary gangliosidoses.

    Topics: Animals; Brain; Cats; Cerebellum; Disease Models, Animal; Frontal Lobe; G(M1) Ganglioside; gamma-Aminobutyric Acid; Gangliosidoses; Glutamates; Glutamic Acid; Humans; Neurotransmitter Agents; Norepinephrine; Occipital Lobe; Synaptosomes

1982
Canine GM1 gangliosidosis. An ultrastructural and biochemical study.
    Journal of neuropathology and experimental neurology, 1982, Volume: 41, Issue:6

    The ultrastructural and biochemical features of canine GM1 gangliosidosis were studied. beta-Galactosidase activity assayed using both skin fibroblast tissue culture strains and fresh skin revealed enzyme activities in three groups (normals, heterozygotes, and homozygotes) corresponding to an autosomal recessive inheritance. The concentration of ganglioside GM1 was greatly increased in cerebral gray matter and kidney. A striking elevation of tissue oligosaccharides was found in liver, kidney, and spleen. Most neurons in the cerebral cortex and deep gray matter were filled by spherical lamellated inclusions. Hepatocytes contained vacuoles with an amorphous granular material which may correspond to the accumulation of galactose-oligosaccharides determined chemically. The disease in dogs has features similar to both the infantile and juvenile form of human GM1 gangliosidosis.

    Topics: Animals; Brain; Brain Chemistry; Disease Models, Animal; Dog Diseases; Dogs; Female; G(M1) Ganglioside; Gangliosidoses; Heterozygote; Homozygote; Humans; Liver; Male

1982
An immunological model of epilepsy: seizures induced by antibodies to GM1 ganglioside.
    Epilepsia, 1981, Volume: 22, Issue:2

    Following observations that the intracerebral injection into rats of antiserum to brain gangliosides resulted in recurrent epileptiform activity and that seizure activity was not seen if antibodies were removed by absorption of the antiserum with pure GM1 ganglioside, a study was undertaken to establish characteristics of the immunological agents used to produce this model of epilepsy. It was determined that the potencies (antibody titers with GM1 ganglioside) of antiganglioside sera can be correlated with the intensities of epileptiform activity they induce; that immunoglobulin fractions from antiganglioside sera are even more effective biologically than the antisera; and that antibodies to GM1 ganglioside purified by affinity chromatography can also induce recurrent epileptiform discharges but are not as effective as either native antiserum or immunoglobulin fractions.

    Topics: Animals; Disease Models, Animal; Electroencephalography; Epilepsy; G(M1) Ganglioside; Gangliosides; Immune Sera; Immunoglobulins; Rats; Seizures

1981
Enzyme replacement using liposome carriers in feline Gm1 gangliosidosis fibroblasts.
    Nature, 1978, Oct-26, Volume: 275, Issue:5682

    Topics: Animals; beta-Galactosidase; Biological Transport; Cats; Disease Models, Animal; Fibroblasts; G(M1) Ganglioside; Galactosidases; Gangliosidoses; Humans; Liposomes; Lysosomes

1978
Fine structure of meganeurites and secondary growth processes in feline GM1-gangliosidosis.
    Brain research, 1978, Mar-17, Volume: 143, Issue:1

    Electron microscope studies were carried out on neurons of the hippocampal formation in a feline mutant with beta-galactosidase deficiency and GMI-gangliosidosis. Fusiform processes with characteristics similar to meganeurites of Golgi studies were identified between cell bodies and axons of pyramidal and granule cells. The presence of dense material subjacent to the plasma membrane at the meganeurite-axon junction provides evidence that meganeurites form at the axon-hillock region and displace the initial axonal segment distally. Meganeurites of hippocampal neurons exhibited pleomorphic secondary processes with fine structural features of growth cones. Spines and spine-synapses were abundant on perikarya and meganeurites. Numerous membranous cytoplasmic bodies (MCBs) were encountered amongst otherwise normally appearing organelles of the cell body. MCBs were densely packed in meganeurites except near their peripheral area. They were less common in dendrites and rare in synapses of the neuropil. The observations provide further support for the view that meganeurites of mature cortical neurons in ganglioside storage diseases have embryonic growth characteristics.

    Topics: Animals; Axons; Cat Diseases; Cats; Dendrites; Disease Models, Animal; Endoplasmic Reticulum; G(M1) Ganglioside; Galactosidases; Gangliosidoses; Hippocampus; Humans; Neurons; Pyramidal Tracts; Synapses

1978
Meganeurites and other aberrant processes of neurons in feline GM1-gangliosidosis: a Golgi study.
    Brain research, 1978, Mar-17, Volume: 143, Issue:1

    Golgi studies were carried out on neurons in several forebrain structures of young adult mutant cats with inherited beta-galactosidase deficiency and neurobehavioral deterioration due to GM1-ganglioside storage disease. Meganeurites similar to those observed in several human gangliosidoses were present on small and medium pyramidal neurons, granule cells of the fascia dentata and spiny neurons of the caudate nucleus. Large and giant pyramidal cells of the motor cortex exhibited prominent somatic spines but lacked meganeurites. Cortical non-pyramidal neurons and aspiny caudate cells were relatively normal in appearance although they showed variable increases in cell body diameter. The range of morphological alterations in different types of cortical neurons in feline GM1-gangliosidosis was identical to that found in human ganglioside storage diseases. Neurite outgrowth from meganeurites was particularly prominent in the feline mutant. The extensive proliferation of neurites confined to meganeurites indicates that the latter have growth properties typical of embryonic neuronal elements. The demonstration of neurite outgrowth from meganeurites of mature cortical neurons in feline GM1-gangliosidosis suggests a possible role for gangliosides in neurite formation during neuronal differentiation and synaptogenesis.

    Topics: Animals; Axons; Cat Diseases; Cats; Caudate Nucleus; Cerebral Cortex; Disease Models, Animal; G(M1) Ganglioside; Galactosidases; Gangliosidoses; Hippocampus; Humans; Motor Cortex; Neurons; Pyramidal Tracts

1978