vx-765 has been researched along with Disease-Models--Animal* in 14 studies
14 other study(ies) available for vx-765 and Disease-Models--Animal
Article | Year |
---|---|
Pyroptosis inhibition improves the symptom of acute myocardial infarction.
Acute myocardial infarction (AMI), the leading cause of mortality worldwide, is a rapidly developing and irreversible disease. Therefore, proper prompt intervention at the early stage of AMI is crucial for its treatment. However, the molecular features in the early stage have not been clarified. Here, we constructed mouse AMI model and profiled transcriptomes and proteomes at the early stages of AMI progress. Immune system was extensively activated at 6-h AMI. Then, pyroptosis was activated at 24-h AMI. VX-765 treatment, a pyroptosis inhibitor, significantly reduced the infarct size and improved the function of cardiomyocytes. Besides, we identified that WIPI1, specifically expressed in heart, was significantly upregulated at 1 h after AMI. Moreover, WIPI1 expression is significantly higher in the peripheral blood of patients with AMI than healthy control. WIPI1 can serve as a potential early diagnostic biomarker for AMI. It likely decelerates AMI progress by activating autophagy pathways. These findings shed new light on gene expression dynamics in AMI progress, and present a potential early diagnostic marker and a candidate drug for clinical pre-treatment to prolong the optimal cure time. Topics: Animals; Autophagy-Related Proteins; Biomarkers; Dipeptides; Disease Models, Animal; Gene Expression Profiling; Gene Expression Regulation; Male; Mice, Inbred C57BL; Myocardial Infarction; Myocardium; para-Aminobenzoates; Proteome; Pyroptosis; Signal Transduction; Transcriptome | 2021 |
Therapeutic candidates for the Zika virus identified by a high-throughput screen for Zika protease inhibitors.
When Zika virus emerged as a public health emergency there were no drugs or vaccines approved for its prevention or treatment. We used a high-throughput screen for Zika virus protease inhibitors to identify several inhibitors of Zika virus infection. We expressed the NS2B-NS3 Zika virus protease and conducted a biochemical screen for small-molecule inhibitors. A quantitative structure-activity relationship model was employed to virtually screen ∼138,000 compounds, which increased the identification of active compounds, while decreasing screening time and resources. Candidate inhibitors were validated in several viral infection assays. Small molecules with favorable clinical profiles, especially the five-lipoxygenase-activating protein inhibitor, MK-591, inhibited the Zika virus protease and infection in neural stem cells. Members of the tetracycline family of antibiotics were more potent inhibitors of Zika virus infection than the protease, suggesting they may have multiple mechanisms of action. The most potent tetracycline, methacycline, reduced the amount of Zika virus present in the brain and the severity of Zika virus-induced motor deficits in an immunocompetent mouse model. As Food and Drug Administration-approved drugs, the tetracyclines could be quickly translated to the clinic. The compounds identified through our screening paradigm have the potential to be used as prophylactics for patients traveling to endemic regions or for the treatment of the neurological complications of Zika virus infection. Topics: Animals; Antiviral Agents; Artificial Intelligence; Chlorocebus aethiops; Disease Models, Animal; Drug Evaluation, Preclinical; High-Throughput Screening Assays; Immunocompetence; Inhibitory Concentration 50; Methacycline; Mice, Inbred C57BL; Protease Inhibitors; Quantitative Structure-Activity Relationship; Small Molecule Libraries; Vero Cells; Zika Virus; Zika Virus Infection | 2020 |
VX-765 attenuates atherosclerosis in ApoE deficient mice by modulating VSMCs pyroptosis.
Recent clinical evidences show that patients with atherosclerotic cardiovascular disease can benefit from a targeting IL-1β treatment. Caspase-1 is an important factor for pyroptosis and is responsible for mature and release of interleukin (IL)-1β. Here we investigated the effect of caspase-1 inhibitor VX-765 on atherosclerosis and vascular smooth muscle cells (VSMCs) pyroptosis.. Human carotid artery plaques and aortas from ApoE-/- mice which were gavaged with VX-765 or vehicle while fed with western diet were examined for plaque burden using Oil Red O staining and Immunohistochemistry staining. Dedifferentiated primary cultured mice VSMCs treated with oxidized low-density lipoprotein (OxLDL) were applied to examine cell pyroptosis.. The distribution of a-SMA and active pyroptotic indicators had a lot of overlaps near the necrotic core, at the lesion surface and in the intra-plaque hemorrhage area in human or mice plaque. In vitro studies further demonstrated that OxLDL induced VSMCs pyroptosis through activating NLRP3 inflammasome. What's more, VX-765 significantly inhibited the progression of established atheroma and the development of atherosclerosis, without substantially influence lipoprotein level in plasma. VX-765 also significantly reduced VSMCs pyroptosis and IL-1β processing induced by OxLDL.. VX-765 inhibits VSMCs pyroptosis during atherogenesis and targeting caspase-1 activity may be a potential treatment strategy for atherosclerotic diseases. Topics: Animals; Apolipoproteins E; Atherosclerosis; Dipeptides; Disease Models, Animal; Disease Progression; Humans; Male; Mice; Mice, Inbred C57BL; Mice, Knockout; Muscle, Smooth, Vascular; Myocytes, Smooth Muscle; para-Aminobenzoates; Pyroptosis | 2020 |
Pre-symptomatic Caspase-1 inhibitor delays cognitive decline in a mouse model of Alzheimer disease and aging.
Early therapeutic interventions are essential to prevent Alzheimer Disease (AD). The association of several inflammation-related genetic markers with AD and the early activation of pro-inflammatory pathways in AD suggest inflammation as a plausible therapeutic target. Inflammatory Caspase-1 has a significant impact on AD-like pathophysiology and Caspase-1 inhibitor, VX-765, reverses cognitive deficits in AD mouse models. Here, a one-month pre-symptomatic treatment of Swedish/Indiana mutant amyloid precursor protein (APP Topics: Aging; Alzheimer Disease; Amyloid beta-Peptides; Animals; Behavior, Animal; Cognitive Dysfunction; Cytokines; Dipeptides; Disease Models, Animal; Encephalitis; Female; Humans; Inflammation; Male; Memory Disorders; Mice; Mice, Inbred C57BL; Mice, Transgenic; para-Aminobenzoates; Serpins; Spatial Memory; Viral Proteins | 2020 |
Caspase-1 inhibition mediates neuroprotection in experimental stroke by polarizing M2 microglia/macrophage and suppressing NF-κB activation.
Stroke is a life-threatening neurological disease with limited therapeutic options. Inflammation is believed to be involved in the pathogenesis of ischemic stroke and contribute to the degree of brain injury. Vx-765 is a potent, selective, small-molecule caspase-1 inhibitor. Current studies have shown the anti-inflammatory properties of vx-765 in various disease; however, the impact of vx-765 on the ischemic stroke is still unclear. In the present study, we determine the neuroprotective effect of vx-765 in mice subjected to transient middle cerebral artery occlusion (MCAO). We found that caspase-1 inhibition by administration of vx-765 ameliorated cerebral injury in mice after ischemic stroke by reducing infarct volume and ameliorating the neurological deficits. Mechanistically, we showed that the contribution of vx-765 to ischemic injuries may be associated with reducing microglial activation, and downregulating the production of associated pro inflammatory cytokines including IL-1β, TNF-α, and iNOS, as well as upregulating anti-inflammatory cytokines such as TGF-β and YM-1. Additionally, vx-765 altered the phenotype of microglia via switching the microglia polarization toward M2 phenotype, as demonstrably related to inhibition of the NF-κB activation. Our findings indicate that vx-765 protects against MCAO injury and attenuated microglia mediated neuroinflammation primarily by shifting microglia polarization from M1 phenotype toward M2 phenotype. Vx-765 might be a potential therapeutic drug for ameliorating ischemic stroke. Topics: Animals; Caspase 1; Caspase Inhibitors; Dipeptides; Disease Models, Animal; Infarction, Middle Cerebral Artery; Macrophage Activation; Macrophages; Male; Mice, Inbred C57BL; Microglia; Neuroprotection; Neuroprotective Agents; NF-kappa B; para-Aminobenzoates; Stroke | 2019 |
The Caspase 1 Inhibitor VX-765 Protects the Isolated Rat Heart via the RISK Pathway.
Protecting the heart from ischaemia-reperfusion (IR) injury is a major goal in patients presenting with an acute myocardial infarction. Pyroptosis is a novel form of cell death in which caspase 1 is activated and cleaves interleukin 1β. VX-785 is a highly selective, prodrug caspase 1 inhibitor that is also clinically available. It has been shown to be protective against acute IR in vivo rat model, and therefore might be a promising possibility for future cardioprotective therapy. However, it is not known whether protection by VX-765 involves the reperfusion injury salvage kinase (RISK) pathway. We therefore investigated whether VX-765 protects the isolated, perfused rat heart via the PI3K/Akt pathway and whether protection was additive with ischaemic preconditioning (IPC).. Langendorff-perfused rat hearts were subject to ischaemia and reperfusion injury in the presence of 30 μM VX-765, with precedent IPC, or the combination of VX-765 and IPC.. VX-765 reduced infarct size (28 vs 48% control; P < 0.05) to a similar extent as IPC (30%; P < 0.05). The PI3 kinase inhibitor, wortmannin, abolished the protective effect of VX-765. Importantly in the model used, we were unable to show additive protection with VX-765 + IPC.. The caspase 1 inhibitor, VX-765, was able to reduce myocardial infarction in a model of IR injury. However, the addition of IPC did not demonstrate any further protection. Topics: Animals; Caspase 1; Caspase Inhibitors; Cytoprotection; Dipeptides; Disease Models, Animal; Isolated Heart Preparation; Male; Myocardial Infarction; Myocardial Reperfusion Injury; Myocardium; para-Aminobenzoates; Phosphatidylinositol 3-Kinase; Proto-Oncogene Proteins c-akt; Pyroptosis; Rats, Sprague-Dawley; Signal Transduction | 2018 |
Caspase-1 inhibition alleviates cognitive impairment and neuropathology in an Alzheimer's disease mouse model.
Alzheimer's disease (AD) is an intractable progressive neurodegenerative disease characterized by cognitive decline and dementia. An inflammatory neurodegenerative pathway, involving Caspase-1 activation, is associated with human age-dependent cognitive impairment and several classical AD brain pathologies. Here, we show that the nontoxic and blood-brain barrier permeable small molecule Caspase-1 inhibitor VX-765 dose-dependently reverses episodic and spatial memory impairment, and hyperactivity in the J20 mouse model of AD. Cessation of VX-765 results in the reappearance of memory deficits in the mice after 1 month and recommencement of treatment re-establishes normal cognition. VX-765 prevents progressive amyloid beta peptide deposition, reverses brain inflammation, and normalizes synaptophysin protein levels in mouse hippocampus. Consistent with these findings, Caspase-1 null J20 mice are protected from episodic and spatial memory deficits, neuroinflammation and Aβ accumulation. These results provide in vivo proof of concept for Caspase-1 inhibition against AD cognitive deficits and pathologies. Topics: Alzheimer Disease; Amyloid beta-Peptides; Animals; Brain; Caspase 1; Caspase Inhibitors; Cognition; Cognition Disorders; Dipeptides; Disease Models, Animal; Humans; Memory; Memory Disorders; Mice, Knockout; para-Aminobenzoates | 2018 |
Preclinical Screening for Treatments for Infantile Spasms in the Multiple Hit Rat Model of Infantile Spasms: An Update.
Topics: Animals; Anticonvulsants; Dipeptides; Disease Models, Animal; Dose-Response Relationship, Drug; Doxorubicin; Drug Evaluation, Preclinical; Estradiol; GABA Antagonists; Humans; Infant; Lipopolysaccharides; Male; Organophosphorus Compounds; para-Aminobenzoates; Random Allocation; Rats; Rats, Sprague-Dawley; Spasms, Infantile; Treatment Outcome | 2017 |
Blockade of the IL-1R1/TLR4 pathway mediates disease-modification therapeutic effects in a model of acquired epilepsy.
We recently discovered that forebrain activation of the IL-1 receptor/Toll-like receptor (IL-1R1/TLR4) innate immunity signal plays a pivotal role in neuronal hyperexcitability underlying seizures in rodents. Since this pathway is activated in neurons and glia in human epileptogenic foci, it represents a potential target for developing drugs interfering with the mechanisms of epileptogenesis that lead to spontaneous seizures. The lack of such drugs represents a major unmet clinical need. We tested therefore novel therapies inhibiting the IL-1R1/TLR4 signaling in an established murine model of acquired epilepsy. We used an epigenetic approach by injecting a synthetic mimic of micro(mi)RNA-146a that impairs IL1R1/TLR4 signal transduction, or we blocked receptor activation with antiinflammatory drugs. Both interventions when transiently applied to mice after epilepsy onset, prevented disease progression and dramatically reduced chronic seizure recurrence, while the anticonvulsant drug carbamazepine was ineffective. We conclude that IL-1R1/TLR4 is a novel potential therapeutic target for attaining disease-modifications in patients with diagnosed epilepsy. Topics: Animals; Anti-Inflammatory Agents, Non-Steroidal; Anticonvulsants; Carbamazepine; Cyanobacteria; Dipeptides; Disease Models, Animal; Epilepsy; Hippocampus; Kainic Acid; Lipopolysaccharides; Male; Mice, Inbred C57BL; MicroRNAs; Oligonucleotides; para-Aminobenzoates; Random Allocation; Receptors, Interleukin-1 Type I; Time Factors; Toll-Like Receptor 4 | 2017 |
Reducing C-terminal truncation mitigates synucleinopathy and neurodegeneration in a transgenic model of multiple system atrophy.
Multiple system atrophy (MSA) is a sporadic orphan neurodegenerative disorder. No treatment is currently available to slow down the aggressive neurodegenerative process, and patients die within a few years after disease onset. The cytopathological hallmark of MSA is the accumulation of alpha-synuclein (α-syn) aggregates in affected oligodendrocytes. Several studies point to α-syn oligomerization and aggregation as a mediator of neurotoxicity in synucleinopathies including MSA. C-terminal truncation by the inflammatory protease caspase-1 has recently been implicated in the mechanisms that promote aggregation of α-syn in vitro and in neuronal cell models of α-syn toxicity. We present here an in vivo proof of concept of the ability of the caspase-1 inhibitor prodrug VX-765 to mitigate α-syn pathology and to mediate neuroprotection in proteolipid protein α-syn (PLP-SYN) mice, a transgenic mouse model of MSA. PLP-SYN and age-matched wild-type mice were treated for a period of 11 wk with VX-765 or placebo. VX-765 prevented motor deficits in PLP-SYN mice compared with placebo controls. More importantly, VX-765 was able to limit the progressive toxicity of α-syn aggregation by reducing its load in the striatum of PLP-SYN mice. Not only did VX-765 reduce truncated α-syn, but it also decreased its monomeric and oligomeric forms. Finally, VX-765 showed neuroprotective effects by preserving tyrosine hydroxylase-positive neurons in the substantia nigra of PLP-SYN mice. In conclusion, our results suggest that VX-765, a drug that was well tolerated in a 6 wk-long phase II trial in patients with epilepsy, is a promising candidate to achieve disease modification in synucleinopathies by limiting α-syn accumulation. Topics: alpha-Synuclein; Animals; Caspase 1; Clinical Trials as Topic; Corpus Striatum; Dipeptides; Disease Models, Animal; Gene Expression Regulation; Humans; Mice; Mice, Transgenic; Multiple System Atrophy; Neurons; Oligodendroglia; para-Aminobenzoates; Protein Aggregates; Proteolysis; Signal Transduction; Substantia Nigra; Tyrosine 3-Monooxygenase | 2016 |
NLRP3 Inflammasome Mediates Chronic Mild Stress-Induced Depression in Mice via Neuroinflammation.
Evidence from both clinical and experimental research indicates that the immune-brain interaction plays a pivotal role in the pathophysiology of depression. A multi-protein complex of the innate immune system, the NLRP3 inflammasome regulates cleavage and secretion of proinflammatory cytokine interleukin-1β. The inflammasome detects various pathogen-associated molecule patterns and damage-associated molecule patterns, which then leads to a series of immune-inflammatory reactions.. To explore the role of inflammasome activation in the underlying biological mechanisms of depression, we established a mouse model of depression with unpredictable chronic mild stress.. Mice subjected to chronic mild stress for 4 weeks had significantly higher serum corticosterone levels, serum interleukin-1β levels, and hippocampal active interleukin-1β protein levels. They also displayed depressive-like symptoms, including decreased sucrose preference and increased immobility time. Moreover, the hippocampi of chronic mild stress-exposed mice had significantly higher activity of caspase-1, which accompanied by higher protein levels of NLRP3 and the apoptotic speck-containing protein with a card. Pretreatment with the NLRP3 inflammasome inhibitor VX-765 decreased serum and hippocampal levels of interleukin-1β protein and significantly moderated the depressive-like behaviors induced by chronic mild stress.. These data suggest the NLRP3 inflammasome mediates stress-induced depression via immune activation. Future procedures targeting the NLRP3 inflammasome may have promising effects in the prevention and treatment of depression. Topics: Animals; Anti-Inflammatory Agents, Non-Steroidal; Antidepressive Agents; Carrier Proteins; Caspase 1; Chronic Disease; Corticosterone; Depressive Disorder; Dietary Sucrose; Dipeptides; Disease Models, Animal; Food Preferences; Hippocampus; Inflammation; Interleukin-1beta; Male; Mice, Inbred BALB C; Motor Activity; Neuroimmunomodulation; NLR Family, Pyrin Domain-Containing 3 Protein; para-Aminobenzoates; Random Allocation; Stress, Psychological; Uncertainty | 2015 |
Pharmacological blockade of IL-1β/IL-1 receptor type 1 axis during epileptogenesis provides neuroprotection in two rat models of temporal lobe epilepsy.
We studied whether pharmacological blockade of the IL-1β-mediated signaling, rapidly activated in forebrain by epileptogenic injuries, affords neuroprotection in two different rat models of status epilepticus (SE). As secondary outcome, we measured treatment's effect on SE-induced epileptogenesis. IL-1β signaling was blocked by systemic administration of two antiinflammatory drugs, namely human recombinant IL-1 receptor antagonist (anakinra), the naturally occurring and clinically used competitive IL-1 receptor type 1 antagonist, and VX-765 a specific non-peptide inhibitor of IL-1β cleavage and release. Antiinflammatory drugs were given 60min after antiepileptic (AED) drug-controlled SE induced by pilocarpine, or 180min after unrestrained electrical SE, for 7days using a protocol yielding therapeutic drug levels in brain. This drug combination significantly decreased both IL-1β expression in astrocytes and cell loss in rat forebrain. Neuroprotection and the antiinflammatory effect were more pronounced in the electrical SE model. Onset of epilepsy, and frequency and duration of seizures 3months after electrical SE were not significantly modified. Transcriptomic analysis in the hippocampus showed that the combined treatment did not affect the broad inflammatory response induced by SE during epileptogenesis. In particular, the treatment did not prevent the induction of the complement system and Toll-like receptors, both contributing to cell loss and seizure generation. We conclude that the IL-1β signaling represents an important target for reducing cell loss after SE. The data highlight a new class of clinically tested agents affording neuroprotection after a delayed post-injury intervention. Earlier blockade of this rapid onset inflammatory pathway during SE, or concomitant treatment with antiinflammatory drugs targeting additional components of the broad inflammatory response to SE, or co-treatment with AEDs, is likely to be required for optimizing beneficial outcomes. Topics: Animals; Cell Death; Cerebral Cortex; Dipeptides; Disease Models, Animal; Electric Stimulation; Epilepsy, Temporal Lobe; Female; Glial Fibrillary Acidic Protein; Hippocampus; Humans; Interleukin 1 Receptor Antagonist Protein; Interleukin-1beta; Lithium; Male; para-Aminobenzoates; Pilocarpine; Rats; Rats, Sprague-Dawley; Receptors, Interleukin-1 Type I | 2013 |
IL-1β is induced in reactive astrocytes in the somatosensory cortex of rats with genetic absence epilepsy at the onset of spike-and-wave discharges, and contributes to their occurrence.
Interleukin (IL)-1β plays a crucial role in the mechanisms of limbic seizures in rodent models of temporal lobe epilepsy. We addressed whether activation of the IL-1β signaling occurs in rats with genetic absence epilepsy (GAERS) during the development of spike-and-wave discharges (SWDs). Moreover, we studied whether inhibition of IL-1β biosynthesis in GAERS could affect SWD activity. IL-1β expression and glia activation were studied by immunocytochemistry in the forebrain of GAERS at postnatal days (PN)14, PN20, and PN90 and in age-matched non-epileptic control Wistar rats. In PN14 GAERS, when no SWDs have developed yet, IL-1β immunostaining was undetectable, and astrocytes and microglia showed a resting phenotype similar to control Wistar rats. In 3 out of 9 PN20 GAERS, IL-1β was observed in activated astrocytes of the somatosensory cortex; the cytokine expression was associated with the occurrence of immature-type of SWDs. In all adult PN90 GAERS, when mature SWDs are established, IL-1β was observed in reactive astrocytes of the somatosensory cortex but not in adjacent cortical areas or in extra-cortical regions. An age-dependent c-fos activation was found in the somatosensory cortex of GAERS with maximal levels reached in PN90 rats; c-fos was also induced in some thalamic nuclei in PN20 and PN90 GAERS. Inhibition of IL-1β biosynthesis in PN90 GAERS by 4-day systemic administration of a specific ICE/Caspase-1 blocker, significantly reduced both SWD number and duration. These results show that IL-1β is induced in reactive astrocytes of the somatosensory cortex of GAERS at the onset of SWDs. IL-1β has pro-ictogenic properties in this model, and thus it may play a contributing role in the mechanisms underlying the occurrence of absence seizures. Topics: 4-Aminobenzoic Acid; Age Factors; Analysis of Variance; Animals; Animals, Newborn; Astrocytes; Brain Waves; Cell Count; Dipeptides; Disease Models, Animal; Electroencephalography; Enzyme Inhibitors; Epilepsy, Absence; Gene Expression Regulation, Developmental; Glial Fibrillary Acidic Protein; Interleukin-1beta; Male; para-Aminobenzoates; Proto-Oncogene Proteins c-fos; Rats; Rats, Mutant Strains; Rats, Wistar; Somatosensory Cortex; Statistics, Nonparametric; Thalamic Nuclei | 2011 |
Interleukin Converting Enzyme inhibition impairs kindling epileptogenesis in rats by blocking astrocytic IL-1beta production.
An enhanced production of IL-1beta in glia is a typical feature of epileptogenic tissue in experimental models and in human drug-refractory epilepsy. We show here that the selective inhibition of Interleukin Converting Enzyme (ICE), which cleaves the biologically active form of IL-1beta using VX-765, blocks kindling development in rats by preventing IL-1beta increase in forebrain astrocytes, without interfering with glia activation. The average afterdischarge duration was not altered significantly by VX-765. Up to 24 h after kindling completion and drug washout, kindled seizures could not be evoked in treated rats. VX-765 did not affect seizures or afterdischarge duration in fully kindled rats. These data indicate an antiepileptogenic effect mediated by ICE inhibition and suggest that specific anti-IL-1beta pharmacological strategies can be envisaged to interfere with epileptogenic mechanisms. Topics: 4-Aminobenzoic Acid; Animals; Anticonvulsants; Astrocytes; Caspase 1; Caspase Inhibitors; Dipeptides; Disease Models, Animal; Enzyme Inhibitors; Epilepsy; Interleukin-1beta; Kindling, Neurologic; Male; para-Aminobenzoates; Prosencephalon; Rats; Rats, Sprague-Dawley | 2008 |