Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Int J Mol Sci ; 25(10)2024 May 17.
Article in English | MEDLINE | ID: mdl-38791515

ABSTRACT

Myocardial necrosis following the successful reperfusion of a coronary artery occluded by thrombus in a patient presenting with ST-elevation myocardial infarction (STEMI) continues to be a serious problem, despite the multiple attempts to attenuate the necrosis with agents that have shown promise in pre-clinical investigations. Possible reasons include confounding clinical risk factors, the delayed application of protective agents, poorly designed pre-clinical investigations, the possible effects of routinely administered agents that might unknowingly already have protected the myocardium or that might have blocked protection, and the biological differences of the myocardium in humans and experimental animals. A better understanding of the pathobiology of myocardial infarction is needed to stem this reperfusion injury. P2Y12 receptor antagonists minimize platelet aggregation and are currently part of the standard treatment to prevent thrombus formation and propagation in STEMI protocols. Serendipitously, these P2Y12 antagonists also dramatically attenuate reperfusion injury in experimental animals and are presumed to provide a similar protection in STEMI patients. However, additional protective agents are needed to further diminish reperfusion injury. It is possible to achieve additive protection if the added intervention protects by a mechanism different from that of P2Y12 antagonists. Inflammation is now recognized to be a critical factor in the complex intracellular response to ischemia and reperfusion that leads to tissue necrosis. Interference with cardiomyocyte inflammasome assembly and activation has shown great promise in attenuating reperfusion injury in pre-clinical animal models. And the blockade of the executioner protease caspase-1, indeed, supplements the protection already seen after the administration of P2Y12 antagonists. Importantly, protective interventions must be applied in the first minutes of reperfusion, if protection is to be achieved. The promise of such a combination of protective strategies provides hope that the successful attenuation of reperfusion injury is attainable.


Subject(s)
Inflammation , Myocardial Reperfusion Injury , NLR Family, Pyrin Domain-Containing 3 Protein , Purinergic P2Y Receptor Antagonists , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Humans , Purinergic P2Y Receptor Antagonists/pharmacology , Purinergic P2Y Receptor Antagonists/therapeutic use , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Inflammation/drug therapy , Inflammation/pathology , Inflammation/metabolism , Receptors, Purinergic P2Y12/metabolism
2.
Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi ; 38(1): 74-81, 2024 Jan 15.
Article in Chinese | MEDLINE | ID: mdl-38225845

ABSTRACT

Objective: To investigate the effects and underlying mechanisms of VX765 on osteoarthritis (OA) and chondrocytes inflammation in rats. Methods: Chondrocytes were isolated from the knee joints of 4-week-old Sprague Dawley (SD) rats. The third-generation cells were subjected to cell counting kit 8 (CCK-8) analysis to assess the impact of various concentrations (0, 1, 5, 10, 20, 50, 100 µmol/L) of VX765 on rat chondrocyte activity. An in vitro lipopolysaccharide (LPS) induced cell inflammation model was employed, dividing cells into control group, LPS group, VX765 concentration 1 group and VX765 concentration 2 group without obvious cytotoxicity. Western blot, real-time fluorescence quantitative PCR, and ELISA were conducted to measure the expression levels of inflammatory factors-transforming growth factor ß 1 (TGF-ß 1), interleukin 6 (IL-6), and tumor necrosis factor α (TNF-α). Additionally, Western blot and immunofluorescence staining were employed to assess the expressions of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase 1 (HO-1). Thirty-two SD rats were randomly assigned to sham surgery group (group A), OA group (group B), OA+VX765 (50 mg/kg) group (group C), and OA+VX765 (100 mg/kg) group (group D), with 8 rats in each group. Group A underwent a sham operation with a medial incision, while groups B to D underwent additional transverse incisions to the medial collateral ligament and anterior cruciate ligament, with removal of the medial meniscus. One week post-surgery, groups C and D were orally administered 50 mg/kg and 100 mg/kg VX765, respectively, while groups A and B received an equivalent volume of saline. Histopathological examination using HE and safranin-fast green staining was performed, and Mankin scoring was utilized for evaluation. Immunohistochemical staining technique was employed to analyze the expressions of matrix metalloproteinase 13 (MMP-13) and collagen type Ⅱ. Results: The CCK-8 assay indicated a significant decrease in cell viability at VX765 concentrations exceeding 10 µmol/L ( P<0.05), so 4 µmol/L and 8 µmol/L VX765 without obvious cytotoxicity were selected for subsequent experiments. Following LPS induction, the expressions of TGF-ß 1, IL-6, and TNF-α in cells significantly increased when compared with the control group ( P<0.05). However, intervention with 4 µmol/L and 8 µmol/L VX765 led to a significant decrease in expression compared to the LPS group ( P<0.05). Western blot and immunofluorescence staining demonstrated a significant upregulation of Nrf2 pathway-related molecules Nrf2 and HO-1 protein expressions by VX765 ( P<0.05), indicating Nrf2 pathway activation. Histopathological examination of rat knee joint tissues and immunohistochemical staining revealed that, compared to group B, treatment with VX765 in groups C and D improved joint structural damage in rat OA, alleviated inflammatory reactions, downregulated MMP-13 expression, and increased collagen type Ⅱ expression. Conclusion: VX765 can improve rat OA and reduce chondrocyte inflammation, possibly through the activation of the Nrf2 pathway.


Subject(s)
Chondrocytes , Dipeptides , Osteoarthritis , para-Aminobenzoates , Rats , Animals , Chondrocytes/metabolism , Matrix Metalloproteinase 13/metabolism , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/metabolism , Collagen Type II/metabolism , Interleukin-6 , Lipopolysaccharides/pharmacology , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/pharmacology , Inflammation/drug therapy , Osteoarthritis/metabolism , Transforming Growth Factor beta1/metabolism
3.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-1009112

ABSTRACT

OBJECTIVE@#To investigate the effects and underlying mechanisms of VX765 on osteoarthritis (OA) and chondrocytes inflammation in rats.@*METHODS@#Chondrocytes were isolated from the knee joints of 4-week-old Sprague Dawley (SD) rats. The third-generation cells were subjected to cell counting kit 8 (CCK-8) analysis to assess the impact of various concentrations (0, 1, 5, 10, 20, 50, 100 μmol/L) of VX765 on rat chondrocyte activity. An in vitro lipopolysaccharide (LPS) induced cell inflammation model was employed, dividing cells into control group, LPS group, VX765 concentration 1 group and VX765 concentration 2 group without obvious cytotoxicity. Western blot, real-time fluorescence quantitative PCR, and ELISA were conducted to measure the expression levels of inflammatory factors-transforming growth factor β 1 (TGF-β 1), interleukin 6 (IL-6), and tumor necrosis factor α (TNF-α). Additionally, Western blot and immunofluorescence staining were employed to assess the expressions of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase 1 (HO-1). Thirty-two SD rats were randomly assigned to sham surgery group (group A), OA group (group B), OA+VX765 (50 mg/kg) group (group C), and OA+VX765 (100 mg/kg) group (group D), with 8 rats in each group. Group A underwent a sham operation with a medial incision, while groups B to D underwent additional transverse incisions to the medial collateral ligament and anterior cruciate ligament, with removal of the medial meniscus. One week post-surgery, groups C and D were orally administered 50 mg/kg and 100 mg/kg VX765, respectively, while groups A and B received an equivalent volume of saline. Histopathological examination using HE and safranin-fast green staining was performed, and Mankin scoring was utilized for evaluation. Immunohistochemical staining technique was employed to analyze the expressions of matrix metalloproteinase 13 (MMP-13) and collagen type Ⅱ.@*RESULTS@#The CCK-8 assay indicated a significant decrease in cell viability at VX765 concentrations exceeding 10 μmol/L ( P<0.05), so 4 μmol/L and 8 μmol/L VX765 without obvious cytotoxicity were selected for subsequent experiments. Following LPS induction, the expressions of TGF-β 1, IL-6, and TNF-α in cells significantly increased when compared with the control group ( P<0.05). However, intervention with 4 μmol/L and 8 μmol/L VX765 led to a significant decrease in expression compared to the LPS group ( P<0.05). Western blot and immunofluorescence staining demonstrated a significant upregulation of Nrf2 pathway-related molecules Nrf2 and HO-1 protein expressions by VX765 ( P<0.05), indicating Nrf2 pathway activation. Histopathological examination of rat knee joint tissues and immunohistochemical staining revealed that, compared to group B, treatment with VX765 in groups C and D improved joint structural damage in rat OA, alleviated inflammatory reactions, downregulated MMP-13 expression, and increased collagen type Ⅱ expression.@*CONCLUSION@#VX765 can improve rat OA and reduce chondrocyte inflammation, possibly through the activation of the Nrf2 pathway.


Subject(s)
Rats , Animals , Chondrocytes/metabolism , Matrix Metalloproteinase 13/metabolism , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/metabolism , Collagen Type II/metabolism , Interleukin-6 , Lipopolysaccharides/pharmacology , NF-E2-Related Factor 2/pharmacology , Inflammation/drug therapy , Osteoarthritis/metabolism , Transforming Growth Factor beta1/metabolism , Dipeptides , para-Aminobenzoates
4.
Neurotox Res ; 41(4): 338-348, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37058197

ABSTRACT

Neuroinflammation is a major contributor to bilirubin-induced neurotoxicity, which results in severe neurological deficits. Microglia are the primary immune cells in the brain, with M1 microglia promoting inflammatory injury and M2 microglia inhibiting neuroinflammation. Controlling microglial inflammation could be a promising therapeutic strategy for reducing bilirubin-induced neurotoxicity. Primary microglial cultures were prepared from 1-3-day-old rats. In the early stages of bilirubin treatment, pro-/anti-inflammatory (M1/M2) microglia mixed polarization was observed. In the late stages, bilirubin persistence induced dominant proinflammatory microglia, forming an inflammatory microenvironment and inducing iNOS expression as well as the release of tumor necrosis factor (TNF)-α, interleukin (IL)-6, and IL-1ß. Simultaneously, nuclear factor-kappa B (NF-κB) was activated and translocated into the nucleus, upregulating inflammatory target genes. As well known, neuroinflammation can have an effect on N-methyl-D-aspartate receptor (NMDAR) expression or function, which is linked to cognition. Treatment with bilirubin-treated microglia-conditioned medium did affect the expression of IL-1ß, NMDA receptor subunit 2A (NR2A), and NMDA receptor subunit 2B (NR2B) in neurons. However, VX-765 effectively reduces the levels of proinflammatory cytokines TNF-α, IL-6, and IL-1ß, as well as the expressions of CD86, and increases the expressions of anti-inflammatory related Arg-1. A timely reduction in proinflammatory microglia could protect against bilirubin-induced neurotoxicity.


Subject(s)
Microglia , Receptors, N-Methyl-D-Aspartate , Rats , Animals , Receptors, N-Methyl-D-Aspartate/metabolism , Neuroinflammatory Diseases , NF-kappa B/metabolism , Anti-Inflammatory Agents/pharmacology , Interleukin-6/metabolism , Tumor Necrosis Factor-alpha/metabolism , Lipopolysaccharides/pharmacology
5.
Ann Hepatol ; 28(3): 101082, 2023.
Article in English | MEDLINE | ID: mdl-36893888

ABSTRACT

INTRODUCTION AND OBJECTIVES: As a fatal clinical syndrome, acute liver failure (ALF) is characterized by overwhelming liver inflammation and hepatic cell death. Finding new therapeutic methods has been a challenge in ALF research. VX-765 is a known pyroptosis inhibitor and has been reported to prevent damage in a variety of diseases by reducing inflammation. However, the role of VX-765 in ALF is still unclear. MATERIALS AND METHODS: ALF model mice were treated with D-galactosamine (D-GalN) and lipopolysaccharide (LPS). LO2 cells were stimulated with LPS. Thirty subjects were enrolled in clinical experiments. The levels of inflammatory cytokines, pyroptosis-associated proteins and peroxisome proliferator-activated receptor α (PPARα) were detected using quantitative reverse transcription-polymerase chain reaction (qRT‒PCR), western blotting and immunohistochemistry. An automatic biochemical analyzer was used to determine the serum aminotransferase enzyme levels. Hematoxylin and eosin (HE) staining was used to observe the pathological features of the liver. RESULTS: With the progression of ALF, the expression levels of interleukin (IL) -1ß, IL-18, caspase-1, and serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) were increased. VX-765 could reduce the mortality rate of ALF mice, relieve liver pathological damage, and reduce inflammatory responses to protect against ALF. Further experiments showed that VX-765 could protect against ALF through PPARα, and this protective effect against ALF was reduced in the context of PPARα inhibition. CONCLUSIONS: As ALF progresses, inflammatory responses and pyroptosis deteriorate gradually. VX-765 can inhibit pyroptosis and reduce inflammatory responses to protect against ALF by upregulating PPARα expression, thus providing a possible therapeutic strategy for ALF.


Subject(s)
Liver Failure, Acute , PPAR alpha , Mice , Animals , PPAR alpha/genetics , PPAR alpha/metabolism , Pyroptosis , Lipopolysaccharides/metabolism , Lipopolysaccharides/pharmacology , Liver Failure, Acute/chemically induced , Liver Failure, Acute/prevention & control , Liver/pathology , Inflammation/prevention & control , Inflammation/metabolism , Tumor Necrosis Factor-alpha/metabolism , Mice, Inbred C57BL
6.
Curr Cancer Drug Targets ; 23(4): 307-318, 2023.
Article in English | MEDLINE | ID: mdl-36200259

ABSTRACT

BACKGROUND: Malignant tumors continue to remain a main global public health issue. In the past 40 years, due to strides made in multi-disciplinary comprehensive treatment schemes for patients suffering from malignant tumors, especially chemotherapy schemes, the survival rate has been greatly improved in such patients. This group can be expected to maintain their fertility or have restored endocrine function following successful malignant tumor treatment. Therefore, focusing on the ovarian damage caused by chemotherapy in women of childbearing age is vital in order to protect their fertility and improve their quality of life. OBJECTIVE: This study attempted to evaluate whether VX-765 possesses an ovarian protective effect in ovarian injury induced by chemotherapy in the mice model. METHODS: Female C57BL/6J mice were administered with VX-765 gavage once a day for 21 consecutive days. Use of cyclophosphamide (Cy) began one week after the last gavage administration of VX- 765. Detailed classification of follicles at various levels was then quantified in each group. Immunohistochemistry and Western blot analysis were then used in order to analyze the expression of key proteins (FOXO3a, mTOR, RPS6 and AKT) as well as their phosphorylation of the PI3K / PTEN / AKT pathways in the ovary. The concentrations of AMH were measured by ELISA. RESULTS: The follicles at all levels of Cy treated mice were less than those of the normal group (P < 0.05). Meanwhile, mice treated with VX-765 prior to receiving Cy treatment had more primordial follicles (PMF) than mice treated with Cy alone (P < 0.05). In early growing follicles (EGF) and antral follicles (AF), no difference was observed among the experimental groups (P > 0.05), however, they were lower than those in the normal group (P < 0.05). In mice treated with continuous Cy, the total follicle number (TF) of mice combined with VX-765 (C-Cy-Vx765) was higher than that of mice without VX-765, and the TF of the two groups was lower than that of the normal group (P < 0.05). The value of PMF/TF in C-Cy-Vx765 group was significantly higher than that in the other three groups, while that of EGF/TF was significantly lower (P < 0.05). Immunohistochemical results showed that the phosphorylated forms of the main proteins of the PI3K / PTEN / AKT pathway were found to be more positive in Cy treated mice. The Western blot analysis showed that when Cy and VX-765 were cotreated, the increased levels of these phosphorylated proteins decreased compared with those treated with Cy alone. The AMH level of infancy Cy and VX-765 co-treated mice was higher than that of infancy normal mice (P < 0.05). After the mice grew to sexual maturity, the AMH level of Cy and VX- 765 co-treated mice was still higher than that of Cy treated mice (P < 0.05), and there was no significant difference with normal mice (P > 0.05). CONCLUSION: VX-765 can maintain the level of AMH and inhibit the recruitment of PMF, thus protecting mice from Cy induced gonadotropic toxicity. Accordingly, VX-765 may play a protective role in mice with ovarian injury caused by chemotherapy.


Subject(s)
Epidermal Growth Factor , Proto-Oncogene Proteins c-akt , Mice , Female , Animals , Proto-Oncogene Proteins c-akt/metabolism , Quality of Life , Mice, Inbred C57BL , Cyclophosphamide , Phosphatidylinositol 3-Kinases/metabolism
7.
Ecotoxicol Environ Saf ; 249: 114359, 2023 Jan 01.
Article in English | MEDLINE | ID: mdl-36508797

ABSTRACT

Silicosis is a diffuse fibrotic lung disease in which excessive inflammatory responses are triggered by silica exposure. Pyroptosis, a pro-inflammatory mode of programmed cell death, is mediated by gasdermin and may play a pivotal role in the development of silicosis. The caspase-1 inhibitor, VX-765, was used in vivo and in vitro to investigate the effects of silica-induced early inflammatory injury and later lung fibrosis. Our findings show that VX-765 reduces inflammatory lung injury by inhibiting silica-induced pyroptosis of alveolar macrophages in a silicosis mouse model. VX-765 limits the infiltration of inflammatory M1 alveolar macrophages, decreasing expression of inflammatory cytokines, including IL-1ß, TNF-α, IL-6, CCL2, and CCL3, and down-regulating endogenous DAMPs and inflammatory immune-related cell pattern recognition receptors TLR4 and NLRP3. Furthermore, VX-765 alleviates fibrosis by down-regulating α-smooth muscle actin (α-SMA), collagen, and fibronectin. In this study, we illustrate that Alveolar macrophages pyroptosis occur in the early stages of silicosis, and VX-765 can alleviate the development of silicosis by inhibiting the pyroptosis signaling pathway. These results may provide new insight into the prevention and treatment of early-stage silicosis.


Subject(s)
Caspase Inhibitors , Lung Injury , Pulmonary Fibrosis , Pyroptosis , Silicosis , Animals , Mice , Lung Injury/chemically induced , Lung Injury/drug therapy , Lung Injury/pathology , Macrophages, Alveolar/drug effects , Pyroptosis/drug effects , Silicon Dioxide/toxicity , Silicosis/drug therapy , Caspase Inhibitors/pharmacology , Caspase Inhibitors/therapeutic use , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/drug therapy
8.
J Mol Neurosci ; 72(12): 2389-2397, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36441377

ABSTRACT

Focal cortical infarction leads to secondary degeneration of the ipsilateral hippocampus, which is associated with poststroke cognitive impairment. VX-765 is a potent small-molecule caspase-1 inhibitor that protects against central nervous system diseases. The present study aimed to determine the protective effects of VX-765 on ß-amyloid (Aß) deposition and secondary degeneration in the hippocampus as well as cognitive decline after cortical infarction. Sprague-Dawley rats were used to establish a distal middle cerebral artery occlusion (dMCAO) model and randomly divided into the vehicle and VX-765 groups. Rats in the vehicle and VX-765 groups, respectively, were subcutaneously injected with VX-765 (50 mg/kg/d) and an isopycnic vehicle once a day for 28 days, starting 1 h after dMCAO. At the end of this 28-day period, cognitive impairment was evaluated with the Morris water maze, and secondary hippocampal damage was evaluated with Nissl staining and immunostaining methods. Neuronal damage and pyroptosis were detected by TUNEL and immunoblotting. The results revealed that VX-765 treatment ameliorated poststroke cognitive dysfunction after ischemia. VX-765 reduced Aß deposition, neuronal loss, and glial activation compared with the vehicle control. In addition, VX-765 treatment increased BDNF levels and normalized synaptophysin protein levels in the hippocampus after cortical infarction. Notably, VX-765 treatment significantly reduced the expression of the pyroptosis-related molecules caspase-1, NLRP3, apoptosis-associated speck-like protein (ASC), gasdermin D, IL-1ß, and IL-18. Additionally, VX-765 significantly decreased the numbers of TUNEL-positive cells and the levels of Bax and cleaved caspase-3 (cC3) and enhanced the levels of Bcl-2 and Bcl-xl after ischemia. Inflammatory pathways, such as the NF-κB and mitogen-activated protein kinase (MAPK) pathways, were inhibited by VX-765 treatment after ischemia. These findings revealed that VX-765 reduced Aß deposition, pyroptosis, and apoptosis in the ipsilateral hippocampus, which may be associated with reduced secondary degeneration and cognitive decline following focal cortical infarction.


Subject(s)
Cognitive Dysfunction , Hippocampus , Animals , Rats , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/etiology , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/drug therapy , Rats, Sprague-Dawley
9.
Metab Brain Dis ; 37(4): 1197-1205, 2022 04.
Article in English | MEDLINE | ID: mdl-35143023

ABSTRACT

The inflammasome assembles leading to increased cleavage and activity of caspase-1 and downstream IL-1ß release, which plays a significant role in the pathogenesis of Alzheimer's disease (AD). Previous studies have shown that caspase-1-mediated neuroinflammation occurs early in AD process. However, the detailed role of caspase-1 in aging-related AD-like neuropathology is still unclear so far. In this study, by using SAMP8 mice, an animal model of accelerated aging, we detected the levels of caspase-1 in brains of 3-, 7-, and 11-month-old mice and observed that caspase-1 was activated during aging process. More importantly, we provided the evidence that VX-765, a selective inhibitor of caspase-1, significantly rescued spatial learning and memory impairments and reduced tau hyperphosphorylation in brains of SAMP8 mice at early stages of the disease. This amelioration might be attributed to IL-1ß-induced hypoactivation of tau kinases. Our results imply that caspase-1 may represent as a potential therapeutic target for neurodegenerative tauopathies.


Subject(s)
Alzheimer Disease , Caspase 1/metabolism , Cognitive Dysfunction , Tauopathies , Animals , Cognitive Dysfunction/drug therapy , Memory Disorders , Mice , Tauopathies/drug therapy
10.
Tissue Cell ; 75: 101718, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35131633

ABSTRACT

BACKGROUND: Intestinal ischemia-reperfusion injury (IIRI) is a common clinical event that can cause serious consequences. The study aimed to investigated the effect of VX-765 in IIRI and its mechanism. METHODS: The hypoxia-reoxygenation (H/R) cell model and IIRI mouse model were generated to examine the in vitro and in vivo effects of VX-765 on IIRI. IIRI was evaluated by histological assessment. ELISA was performed to determine the levels of IL-6, TNF-α, IL-1ß, caspase-1, and GSDMD in intestinal tissues as well as the levels of MDA, SOD, CAT, caspase-1, and GSDMD in Caco-2 cells. Relative protein levels of NLRP3, ASC, IL-18, IL-1ß, cleaved Caspase1, and GSDMD-N were analyzed by Western blotting. CCK-8 Assay was conducted to determine the optimal concentration of VX-765 for the in vitro studies. Flow cytometry, fluorescence microscopy and real-time PCR (RT-PCR) were used to assess ROS levels and the mRNA levels of IL-18 and IL-1ß, respectively. Immunofluorescence staining was performed to examine the subcellular localization of P65 and NLRP3. RESULTS: VX-765 reduced IIRI-induced oxidative stress and inflammatory response both in vivo and in vitro, while it decreased the levels of TNF-α, IL-6, IL-1ß as well as the modified Park/Chiu scores. The optimal concentration of VX-765 for the in vitro studies was 10 µM. Moreover, VX-765 inhibited the nuclear translocation of P65, reduced oxidative stress and down-regulated the activation of NLRP3 inflammasome. CONCLUSION: VX-765 prevents IIRI presumably by inhibiting the activation of NLRP3 inflammasome.


Subject(s)
Inflammasomes , Reperfusion Injury , Animals , Caco-2 Cells , Dipeptides , Humans , Inflammasomes/metabolism , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Reperfusion Injury/drug therapy , Reperfusion Injury/prevention & control , para-Aminobenzoates
11.
Acta Pharmacol Sin ; 43(9): 2289-2301, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35132192

ABSTRACT

Connexin 43 (Cx43) is the most important protein in the gap junction channel between cardiomyocytes. Abnormalities of Cx43 change the conduction velocity and direction of cardiomyocytes, leading to reentry and conduction block of the myocardium, thereby causing arrhythmia. It has been shown that IL-1ß reduces the expression of Cx43 in astrocytes and cardiomyocytes in vitro. However, whether caspase-1 and IL-1ß affect connexin 43 after myocardial infarction (MI) is uncertain. In this study we investigated the effects of VX765, a caspase-1 inhibitor, on the expression of Cx43 and cell-to-cell communication after MI. Rats were treated with VX765 (16 mg/kg, i.v.) 1 h before the left anterior descending artery (LAD) ligation, and then once daily for 7 days. The ischemic heart was collected for histochemical analysis and Western blot analysis. We showed that VX765 treatment significantly decreased the infarct area, and alleviated cardiac dysfunction and remodeling by suppressing the NLRP3 inflammasome/caspase-1/IL-1ß expression in the heart after MI. In addition, VX765 treatment markedly raised Cx43 levels in the heart after MI. In vitro experiments were conducted in rat cardiac myocytes (RCMs) stimulated with the supernatant from LPS/ATP-treated rat cardiac fibroblasts (RCFs). Pretreatment of the RCFs with VX765 (25 µM) reversed the downregulation of Cx43 expression in RCMs and significantly improved intercellular communication detected using a scrape-loading/dye transfer assay. We revealed that VX765 suppressed the activation of p38 MAPK signaling in the heart tissue after MI as well as in RCMs stimulated with the supernatant from LPS/ATP-treated RCFs. Taken together, these data show that the caspase-1 inhibitor VX765 upregulates Cx43 expression and improves cell-to-cell communication in rat heart after MI via suppressing the IL-1ß/p38 MAPK pathway.


Subject(s)
Caspase 1 , Connexin 43 , Myocardial Infarction , Animals , Rats , Adenosine Triphosphate/pharmacology , Arrhythmias, Cardiac , Caspase 1/metabolism , Caspase 1/pharmacology , Caspase Inhibitors/pharmacology , Caspases , Cell Communication/drug effects , Connexin 43/genetics , Connexin 43/metabolism , Interleukin-1beta/metabolism , Lipopolysaccharides/pharmacology , Myocardial Infarction/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Serpins , Viral Proteins , Gene Expression/drug effects
12.
Int Immunopharmacol ; 102: 108405, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34865993

ABSTRACT

Inflammatory bowel disease (IBD) is an autoimmune disease involving intestinal tissue. IBD activates a series of cell death pathways. Pyroptosis is recently identified as a critical cell death pathway in IBD associated with the activation of caspase-1. VX765 is a caspase-1 inhibitor that can be converted to VRT-043198 in vivo. This study was designed to explore the therapeutic effect of VX765 on colitis using a dextran sulfate sodium (DSS)-induced colitis model in mice. In this research, the caspase-1 inhibitor on inflammatory, pyroptosis, apoptosis, macrophage activation, and intestinal barrier were investigated. We found that administration of VX765 attenuated body weight loss, colonic shortening, and colonic pathological injury in mice. Our study also revealed a therapeutic effect of VX765 on colitis in a dose-dependent manner. VX765 inhibited pyroptosis by curbing the Caspase-1/GSDMD pathway and its downstream key inflammatory cytokines--IL-1ß and IL-18. These results indicated that VX765 might have a dose-dependent therapeutic effect on DSS-induced colitis in mice.


Subject(s)
Caspase 1/metabolism , Caspase Inhibitors/therapeutic use , Colitis/drug therapy , Dipeptides/therapeutic use , Pyroptosis/drug effects , para-Aminobenzoates/therapeutic use , Animals , Blotting, Western , Caspase 1/drug effects , Colitis/chemically induced , Dextran Sulfate/pharmacology , Male , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction
13.
J Pharm Pharmacol ; 74(3): 377-386, 2022 Mar 03.
Article in English | MEDLINE | ID: mdl-34383065

ABSTRACT

OBJECTIVE: This study explores the potential role of a highly selective caspase-1 inhibitor, VX-765, on extracellular matrix (ECM) accumulation and inflammation in diabetic nephropathy (DN) and the underlying mechanisms. METHODS: DN rats, induced via high-fat diet/streptozotocin, were used to assess the effects of VX-765. Parallel experiments were carried out on rat mesangial cell line HBZY-1 exposed to high glucose (HG) to reveal the molecular mechanism of VX-765 in preventing DN. Survival analysis, biochemical parameters and renal oxidative stress of rats were observed, and Western blotting and immunofluorescence were evaluated. In vitro, Nicotinamide adenine dinucleotide phosphate (NADPH) oxidases (NOX)1 silencing by RNA interference and quantitative real-time PCR (qPCR) assays were conducted in HBZY-1 cells exposed to HG levels. KEY FINDINGS: In vivo, VX-765 significantly reduced the increase in urine albumin excretion and ECM accumulation. The phosphorylation of nuclear factor kappa-B (NF-κB) and the expression of pro-inflammatory cytokines IL-1ß, IL-6 and tumor necrosis factor (TNF)-α were significantly down-regulated. Furthermore, the generation of reactive oxygen species (ROS), phosphorylation of NF-κB and the expression of the NOX1 gene or protein were significantly decreased in HBZY-1 with VX-765 (5 µM) treatment in vitro. CONCLUSIONS: Our results demonstrated that VX-765 exerts favourable effects on DN via the simultaneous alleviation of systemic metabolic syndrome and down-regulating the renal NOX1/ROS/NF-κB pathway, suggesting that it has therapeutic potential for DN.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Diabetic Nephropathies/prevention & control , Dipeptides/pharmacology , Inflammation/drug therapy , para-Aminobenzoates/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cell Line , Diabetes Mellitus, Experimental/complications , Down-Regulation/drug effects , Extracellular Matrix/drug effects , Male , NADPH Oxidase 1/metabolism , NF-kappa B/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Streptozocin
14.
J Diabetes Investig ; 13(1): 22-33, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34494385

ABSTRACT

INTRODUCTION: As a lytic inflammatory cell death, pyroptosis has been recently described but has not been unequivocally elucidated in diabetic nephropathy (DN). VX-765 is a safe and effective inhibitor of caspase-1, that was well tolerated in a phase II clinical trial in patients with epilepsy, but its application in DN is still undefined. MATERIALS AND METHODS: Immunoblot, co-immunoprecipitation, confocal microscope and flow cytometry were used to analyze the effects of glucose on pyroptosis in renal tubular epithelia (HK-2). In vitro, selective caspase-1 inhibitors VX-765 and Z-YVAD-FMK were administered. Pyroptosis and fibrogenesis were determined by immunoblot, ELISA, cytotoxicity assay and flow cytometry. In vivo, diabetic mice were administered with 100 mg/kg VX-765. Renal function, pathological changes, and the expressions of NLRC4, GSDMD, IL-1ß, collagen I, fibronectin and CD45 in renal cortex were evaluated. RESULTS: We identified NLRC4 as a sensor for caspase-1 activation. Moreover, we provided morphological and molecular evidence for pyroptosis in glucose-stressed tubular cells, including ballooned cell membrane, caspase-1 immunoreactivity, GSDMD cleavage, and the release of inflammatory cytokine and cellular contents. All these effects were prevented by treatment with VX-765 or Z-YVAD-FMK, confirming that caspase-1 effectively regulates the occurrence of pyroptosis in HK-2 cells. In vivo, treatment of diabetic animals with VX-765 ameliorated renal function, suppressed inflammatory cell infiltration and pyroptosis-associated protein expression, and mitigated tubulointerstitial fibrosis. CONCLUSIONS: This work revealed that caspase-1-mediated pyroptosis drives renal inflammation and fibrosis in diabetes. Our results are the first demonstration of VX-765 representing a promising therapeutic opportunity for alleviating the progression of DN.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Diabetic Nephropathies/drug therapy , Dipeptides/pharmacology , Kidney/pathology , Pyroptosis/drug effects , para-Aminobenzoates/pharmacology , Animals , Caspase 1/metabolism , Cell Culture Techniques , Diabetes Mellitus, Experimental/pathology , Diabetic Nephropathies/pathology , Fibrosis , Glucose/pharmacology , Humans , Inflammation , Kidney Tubules/drug effects , Kidney Tubules/pathology , Male , Mice
15.
Pharmacol Res ; 175: 106033, 2022 01.
Article in English | MEDLINE | ID: mdl-34915124

ABSTRACT

Acute kidney injury (AKI) is a worldwide problem, and there is no effective drug to eliminate AKI. The death of renal cells is an important pathological basis of intrinsic AKI. At present, targeted therapy for TEC death is a research hotspot in AKI therapy. There are many ways of cell death involved in the occurrence and development of AKI, such as apoptosis, necrosis, ferroptosis, and pyroptosis. This article mainly focuses on the role of pyroptosis in AKI. The assembly and activation of NLRP3 inflammasome is a key event in the occurrence of pyroptosis, which is affected by many factors, such as the activation of the NF-κB signaling pathway, mitochondrial instability and excessive endoplasmic reticulum (ER) stress. The activation of NLRP3 inflammasome can trigger its downstream inflammatory cytokines, which will lead to pyroptosis and eventually induce AKI. In this paper, we reviewed the possible mechanism of pyroptosis in AKI and the potential effective inhibitors of various key targets in this process. It may provide potential therapeutic targets for novel intrinsic AKI therapies based on pyroptosis, so as to develop better therapeutic strategies.


Subject(s)
Acute Kidney Injury/drug therapy , Pyroptosis , Acute Kidney Injury/metabolism , Animals , Humans , Signal Transduction
16.
ACS Chem Neurosci ; 12(15): 2929-2939, 2021 08 04.
Article in English | MEDLINE | ID: mdl-34296848

ABSTRACT

Kernicterus is a severe complication of extreme neonatal hyperbilirubinemia. Prolonged exposure to high-level unconjugated bilirubin (UCB) directly damages brain tissue. Neuroinflammation is believed to contribute to UCB-induced neurotoxicity. Pyroptosis has been as a highly inflammatory form of programmed cell death. Therefore, this study aimed to explore whether pyroptosis was involved in the pathogenesis of UCB neurotoxicity in kernicterus model rats. VX-765, a specific inhibitor of caspase-1, was intraperitoneally administered to the model rats to observe its effects on the short-term and long-term outcomes of the model animals at the molecular, cellular, morphological, and behavioral levels. The results indicated that UCB significantly induced the activation of caspase-1 and gasdermin D(GSDMD), and VX-765 inhibited caspase-1-GSDMD pathway. Compared with those of the UCB group and the vehicle+UCB group, VX-765-treated rats released lower levels of IL-1ß and IL-18. Furthermore, H&E and TUNEL staining showed that nerve cells in the VX-765-treated group were better preserved and had less DNA fragmentation. Most importantly, VX-765 improved both the short-term and long-term neurological functions of kernicterus model rats. This study demonstrated that pyroptosis was involved in the pathogenesis of kernicterus through caspase-1 activation, which could be inhibited by VX-765, exerting a neuroprotective effect in kernicterus model rats.


Subject(s)
Kernicterus , Animals , Bilirubin , Caspase 1 , Kernicterus/drug therapy , Neurons , Pyroptosis , Rats
17.
Pharmacol Res ; 165: 105447, 2021 03.
Article in English | MEDLINE | ID: mdl-33516832

ABSTRACT

Pyroptosis is a form of programmed cell death activated by various stimuli and is characterized by inflammasome assembly, membrane pore formation, and the secretion of inflammatory cytokines (IL-1ß and IL-18). Atherosclerosis-related risk factors, including oxidized low-density lipoprotein (ox-LDL) and cholesterol crystals, have been shown to promote pyroptosis through several mechanisms that involve ion flux, ROS, endoplasmic reticulum stress, mitochondrial dysfunction, lysosomal rupture, Golgi function, autophagy, noncoding RNAs, post-translational modifications, and the expression of related molecules. Pyroptosis of endothelial cells, macrophages, and smooth muscle cells in the vascular wall can induce plaque instability and accelerate atherosclerosis progression. In this review, we focus on the pathogenesis, influence, and therapy of pyroptosis in atherosclerosis and provide novel ideas for suppressing pyroptosis and the progression of atherosclerosis.


Subject(s)
Atherosclerosis/immunology , Endothelial Cells/immunology , Immunity, Cellular/immunology , Inflammation Mediators/immunology , Pyroptosis/immunology , Animals , Atherosclerosis/metabolism , Endothelial Cells/metabolism , Humans , Inflammation Mediators/metabolism , Lipoproteins, LDL/immunology , Lipoproteins, LDL/metabolism , Macrophages/immunology , Macrophages/metabolism , Myocytes, Smooth Muscle/immunology , Myocytes, Smooth Muscle/metabolism
18.
Glia ; 69(1): 216-229, 2021 01.
Article in English | MEDLINE | ID: mdl-32882086

ABSTRACT

Inflammatory demyelination and axonal injury in the central nervous system (CNS) are cardinal features of progressive multiple sclerosis (MS), and linked to activated brain macrophage-like cells (BMCs) including resident microglia and trafficking macrophages. Caspase-1 is a pivotal mediator of inflammation and cell death in the CNS. We investigated the effects of caspase-1 activation and its regulation in models of MS. Brains from progressive MS and non-MS patients, as well as cultured human oligodendrocytes were examined by transcriptomic and morphological methods. Next generation transcriptional sequencing of progressive MS compared to non-MS patients' normal appearing white matter (NAWM) showed induction of caspase-1 as well as other inflammasome-associated genes with concurrent suppression of neuron-specific genes. Oligodendrocytes exposed to TNFα exhibited upregulation of caspase-1 with myelin gene suppression in a cell differentiation state-dependent manner. Brains from cuprizone-exposed mice treated by intranasal delivery of the caspase-1 inhibitor, VX-765 or its vehicle, were investigated in morphological and molecular studies, as well as by fluorodeoxyglucose-positron emission tomography (FDG-PET) imaging. Cuprizone exposure resulted in BMC and caspase-1 activation accompanied by demyelination and axonal injury, which was abrogated by intranasal VX-765 treatment. FDG-PET imaging revealed suppressed glucose metabolism in the thalamus, hippocampus and cortex of cuprizone-exposed mice that was restored with VX-765 treatment. These studies highlight the caspase-1 dependent interactions between inflammation, demyelination, and glucose metabolism in progressive MS and associated models. Intranasal delivery of an anti-caspase-1 therapy represents a promising therapeutic approach for progressive MS and other neuro-inflammatory diseases.


Subject(s)
Multiple Sclerosis, Chronic Progressive , Animals , Caspase 1 , Cuprizone/toxicity , Disease Models, Animal , Fluorodeoxyglucose F18 , Glucose , Humans , Inflammation , Mice , Mice, Inbred C57BL , Myelin Sheath
19.
Front Cell Dev Biol ; 9: 801422, 2021.
Article in English | MEDLINE | ID: mdl-35127716

ABSTRACT

Background: Alzheimer's disease (AD) is the most common form of dementia worldwide. Previous studies have reported that sevoflurane, a frequently used anesthetic, can induce cognitive impairment in preclinical and clinical settings. However, the mechanism underlying the development of this neurotoxicity is currently unclear. Methods: Seven-month-old APP/PS1 mice were placed in an anesthesia induction box containing 3% sevoflurane in 100% O2 for 6 h, while BV2 cells were cultured with 4% sevoflurane for 6 h. Pyroptosis and tau protein expression in excised hippocampus tissues and cells were measured using Western blotting and immunofluorescence assay. Caspase-1 and NLRP3 were knocked out in BV2 microglia using CRISPR/Cas9 technology to determine whether they mediate the effects induced by sevoflurane. Results: Sevoflurane directly activated caspase-1 to induce pyroptosis in the mouse model of AD via NLRP3 and AIM2 activation. In addition, sevoflurane mediated cleavage of gasdermin D (GSDMD) but not gasdermin E (GSDME), promoted the biosynthesis of downstream interleukin-1ß and interleukin-18, and increased ß-amyloid (Aß) deposition and tau phosphorylation. The nontoxic caspase-1 small-molecule inhibitor VX-765 significantly inhibited this activation process in microglia, while NLRP3 deletion suppressed sevoflurane-induced caspase-1 cleavage and subsequently pyroptosis, as well as tau pathology. Furthermore, silencing caspase-1 alleviated the sevoflurane-induced release of IL-1ß and IL-18 and inhibited tau-related enzymes in microglia. Conclusion: This study is the first to report that clinical doses of sevoflurane aggravate the progression of AD via the NLRP3/caspase-1/GSDMD axis. Collectively, our findings elucidate the crucial mechanisms of NLRP3/caspase-1 in pyroptosis and tau pathogenesis induced by sevoflurane and suggest that VX-765 could represent a novel therapeutic intervention for treating AD.

20.
Mol Med Rep ; 22(1): 33-42, 2020 07.
Article in English | MEDLINE | ID: mdl-32377730

ABSTRACT

Previous studies have shown that caspase-1 plays an important role in the acute inflammatory response of spinal cord injury (SCI). VX­765, a novel and irreversible caspase­1 inhibitor, has been reported to effectively intervene in inflammation. However, the effect of VX­765 on genome­wide transcription in acutely injured spinal cords remains unknown. Therefore, in the present study, RNA­sequencing (RNA­Seq) was used to analyze the effect of VX­765 on the local expression of gene transcription 8 h following injury. The differentially expressed genes (DEGs) underwent enrichment analysis of functions and pathways by Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses, respectively. Parallel analysis of western blot confirmed that VX­765 can effectively inhibit the expression and activation of caspase­1. RNA­Seq showed that VX­765 treatment resulted in 1,137 upregulated and 1,762 downregulated DEGs. These downregulated DEGs and their associated signaling pathways, such as focal adhesion, cytokine­cytokine receptor interaction, leukocyte transendothelial migration, extracellular matrix­receptor interaction, phosphatidylinositol 3­kinase­protein kinase B, Rap1 and hypoxia inducible factor­1 signaling pathway, are mainly associated with inflammatory response, local hypoxia, macrophage differentiation, adhesion migration and apoptosis of local cells. This suggests that the application of VX­765 in the acute phase can improve the local microenvironment of SCI by inhibiting caspase­1. However, whether VX­765 can be used as a therapeutic drug for SCI requires further exploration. The sequence data have been deposited into the Sequence Read Archive (https://www.ncbi.nlm.nih.gov/sra/PRJNA548970).


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Caspase Inhibitors/pharmacology , Dipeptides/pharmacology , Spinal Cord Injuries/drug therapy , Transcriptome/drug effects , para-Aminobenzoates/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Caspase 1/metabolism , Caspase Inhibitors/therapeutic use , Dipeptides/therapeutic use , Female , Gene Expression Profiling , Mice , Mice, Inbred C57BL , Spinal Cord Injuries/genetics , Spinal Cord Injuries/metabolism , para-Aminobenzoates/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...