scholarly journals Rosuvastatin protects against coronary microembolization-induced cardiac injury via inhibiting NLRP3 inflammasome activation

2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Ao Chen ◽  
Zhangwei Chen ◽  
You Zhou ◽  
Yuan Wu ◽  
Yan Xia ◽  
...  

AbstractCoronary microembolization (CME), a common reason for periprocedural myocardial infarction (PMI), bears very important prognostic implications. However, the molecular mechanisms related to CME remain largely elusive. Statins have been shown to prevent PMI, but the underlying mechanism has not been identified. Here, we examine whether the NLRP3 inflammasome contributes to CME-induced cardiac injury and investigate the effects of statin therapy on CME. In vivo study, mice with CME were treated with 40 mg/kg/d rosuvastatin (RVS) orally or a selective NLRP3 inflammasome inhibitor MCC950 intraperitoneally (20 mg/kg/d). Mice treated with MCC950 and RVS showed improved cardiac contractile function and morphological changes, diminished fibrosis and microinfarct size, and reduced serum lactate dehydrogenase (LDH) level. Mechanistically, RVS decreased the expression of NLRP3, caspase-1, interleukin-1β, and Gasdermin D N-terminal domains. Proteomics analysis revealed that RVS restored the energy metabolism and oxidative phosphorylation in CME. Furthermore, reduced reactive oxygen species (ROS) level and alleviated mitochondrial damage were observed in RVS-treated mice. In vitro study, RVS inhibited the activation of NLRP3 inflammasome induced by tumor necrosis factor α plus hypoxia in H9c2 cells. Meanwhile, the pyroptosis was also suppressed by RVS, indicated by the increased cell viability, decreased LDH and propidium iodide uptake in H9c2 cells. RVS also reduced the level of mitochondrial ROS generation in vitro. Our results indicate the NLRP3 inflammasome-dependent cardiac pyroptosis plays an important role in CME-induced cardiac injury and its inhibitor exerts cardioprotective effect following CME. We also uncover the anti-pyroptosis role of RVS in CME, which is associated with regulating mitochondrial ROS.

2020 ◽  
Vol 41 (Supplement_2) ◽  
Author(s):  
A Chen ◽  
Z.W Chen ◽  
J.Y Qian ◽  
J.B Ge

Abstract Background Coronary microembolization (CME), a common reason for periprocedural myocardial infarction (PMI) after coronary interventions, bears very important prognostic implications. However, the molecular mechanisms related to CME remain largely elusive. Statins have been shown to prevent PMI, but the underlying mechanism has not been identified. Purpose To determine whether NLRP3 inflammasome contributes to CME-induced cardiac injury and to investigate the effect of statin therapy on CME. Methods In vivo study, CME models were established in C57BL/6J mice by injecting 500,000 polyethylene microspheres (diameter 9 μm) into the left ventricle chamber with the occlusion of the ascending aorta. Mice with CME were treated with 40 mg/kg/d rosuvastatin orally or a selective NLRP3 inflammasome inhibitor MCC950 intraperitoneally (20 mg/kg/d). Cardiac function was evaluated by echocardiography. Cardiac histological analysis was performed. The NLRP3 inflammasome and pyroptosis-related genes and proteins were detected. Cardiac injury was evaluated by Heidenhain's iron hematoxylin stain and serum lactate dehydrogenase (LDH) measurement. Cardiac reactive oxygen species (ROS) production and mitochondrial morphology were also determined. In vitro study, H9c2 myoblasts, stimulated with the classic NLRP3 inflammasome activators lipopolysaccharide (LPS) plus nigericin (Nig), were used to reveal underlying mechanisms of the rosuvastatin's effects. Results The expression of NLRP3, caspase-1 and interleukin-1β (IL-1β) were all increased and peaking at 3 days after CME. Mice treated with MCC950 and rosuvastatin showed improved cardiac contractile function and morphological changes, diminished fibrosis and microinfarct size, and reduced serum LDH level. Mechanistically, rosuvastatin decreased the expression of NLRP3, caspase-1, IL-1β and the pyroptosis executor Gasdermin D. Furthermore, reduced ROS level and alleviated mitochondrial damage were observed in rosuvastatin-treated mice. In vitro study, rosuvastatin effectively inhibited the activation of NLRP3 inflammasome induced by LPS and Nig in H9c2 cells, as evidenced by decreased expression of NLRP3, caspase-1, IL-1β and Gasdermin D. Flow cytometry revealed that rosuvastatin rescued LPS and Nig-induced caspase-1 activation from 22.2±1.6% to 4.09±0.85%. Meanwhile, the pyroptosis was also suppressed by rosuvastatin, indicated by the increased cell viability, decreased LDH and propidium iodide uptake. Similarly, rosuvastatin also reduced the level of ROS generation in vitro. Conclusions NLRP3 inflammasome-dependent cardiac pyroptosis plays an important role in CME-induced cardiac injury and its inhibitor exerts cardioprotective effect following CME. We also uncover the anti-pyroptosis role of rosuvastatin in CME, which is associated with regulating mitochondrial ROS. Funding Acknowledgement Type of funding source: None


2022 ◽  
Vol 2022 ◽  
pp. 1-15
Author(s):  
Lei Zhang ◽  
Cundong Fan ◽  
Hua-Chen Jiao ◽  
Qian Zhang ◽  
Yue-Hua Jiang ◽  
...  

Calycosin (CAL) is the main active component present in Astragalus and reportedly possesses diverse pharmacological properties. However, the cardioprotective effect and underlying mechanism of CAL against doxorubicin- (DOX-) induced cardiotoxicity need to be comprehensively examined. Herein, we aimed to investigate whether the cardioprotective effects of CAL are related to its antipyroptotic effect. A cardiatoxicity model was established by stimulating H9c2 cells and C57BL/6J mice using DOX. In vitro, CAL increased H9c2 cell viability and decreased DOX-induced pyroptosis via NLRP3, caspase-1, and gasdermin D signaling pathways in a dose-dependent manner. In vivo, CAL-DOX cotreatment effectively suppressed DOX-induced cytotoxicity as well as inflammatory and cardiomyocyte pyroptosis via the same molecular mechanism. Next, we used nigericin (Nig) and NLRP3 forced overexpression to determine whether CAL imparts antipyroptotic effects by inhibiting the NLRP3 inflammasome in vitro. Furthermore, CAL suppressed DOX-induced mitochondrial oxidative stress injury in H9c2 cells by decreasing the generation of reactive oxygen species and increasing mitochondrial membrane potential and adenosine triphosphate. Likewise, CAL attenuated the DOX-induced increase in malondialdehyde content and decreased superoxide dismutase and glutathione peroxidase activities in H9c2 cells. In vivo, CAL afforded a protective effect against DOX-induced cardiac injury by improving myocardial function, inhibiting brain natriuretic peptide, and improving the changes of the histological morphology of DOX-treated mice. Collectively, our findings confirmed that CAL alleviates DOX-induced cardiotoxicity and pyroptosis by inhibiting NLRP3 inflammasome activation in vivo and in vitro.


2021 ◽  
Author(s):  
Sahabuddin Ahmed ◽  
Samir Ranjan Panda ◽  
Mohit Kwatra ◽  
Bidya Dhar Sahu ◽  
VGM Naidu

Abstract Several activators of NLRP3 inflammasome have been described; however, the central mechanisms of NLRP3 inflammasome activation in brain microglia, especially at the activating step through free radical generation, still require further clarification. Hence the present study aimed to investigate the role of free radicals in activating NLRP3 inflammasome driven neurodegeneration and elucidated the neuroprotective role of perillyl alcohol (PA) in vitro and in vivo models of Parkinson’s disease. Initial priming of microglial cells with lipopolysaccharide (LPS) following treatment with hydrogen peroxide (H2O2) induces NF-κB translocation to nucleus with robust generation of free radicals that act as Signal 2 in augmenting NLRP3 inflammasome assembly and its downstream targets. PA treatment suppresses nuclear translocation of NF-κB and maintains cellular redox homeostasis in microglia that limits NLRP3 inflammasome activation along with processing active caspase-1, IL-1β and IL-18. To further correlates the in vitro study with in vivo MPTP model, treatment with PA also inhibits the nuclear translocation of NF-κB and downregulates the NLRP3 inflammasome activation. PA administration upregulates various antioxidant enzymes levels and restored the level of dopamine and other neurotransmitters in the striatum of the mice brain with improved behavioural activities. Additionally, treatment with Mito-TEMPO (a mitochondrial ROS inhibitor) was also seen to inhibit NLRP3 inflammasome and rescue dopaminergic neuron loss in the mice brain. Therefore, we conclude that NLRP3 inflammasome activation requires a signal from damaged mitochondria for its activation. Further pharmacological scavenging of free radicals restricts microglia activation and simultaneously supports neuronal survival via targeting NLRP3 inflammasome pathway in Parkinson’s disease.


Circulation ◽  
2014 ◽  
Vol 130 (suppl_2) ◽  
Author(s):  
Zufeng Ding ◽  
Sadip Pant ◽  
Abhishek Deshmukh ◽  
Jawahar L Mehta

Objective: This study tested the hypothesis that mitochondrial DNA damage could trigger NLRP3 inflammasome activation during inflammation, and LOX-1 may play a critical role in this process. Methods and Results: We performed studies in cultured human THP1 macrophages exposed to ox-LDL or LPS,which are often used as inflammation stimuli in vitro . We examined and confirmed the increase in LOX-1 expression when cells were treated with ox-LDL or LPS. Parallel groups of cells were treated with LOX-1 Ab to bind LOX-1. In accordance with our previous studies in endothelial cells and smooth muscle cells, LOX-1 Ab markedly reduced ox-LDL- as well as LPS-stimulated LOX-1 expression. To assess mitochondrial ROS generation, MitoSOX™ Red mitochondrial superoxide indicator was used. Both fluorescence staining and flow cytometry analysis showed that LPS induced (more than ox-LDL) mitochondrial ROS generation. Pretreatment with LOX-1 Ab significantly attenuated mitochondrial ROS generation in response to ox-LDL or LPS. Then we observed mtDNA damage in THP1 cells exposed to ox-LDL or LPS. Importantly, pretreatment with LOX-1 Ab protected mtDNA from damage in response to both stimuli. This was also confirmed by q-PCR (mtDNA/nDNA ratio) analysis. Further, ox-LDL or LPS induced the expression of phos-NF-kB p65, caspase-1 p10 and p20, and cleaved proteins IL-1β and IL-18. Of note, NLRP3 inflammasome was activated in response to ox-LDL or LPS in a similar manner. Pretreatment of cells with LOX-1 Ab treatment blocked or significantly attenuated these inflammatory responses. Conclusions: These observations based on in vitro observations indicate that LOX-1 via ROS generation plays a key role in mtDNA damage which then leads to NLRP3 inflammasome activation during inflammation.


Inflammasome ◽  
2016 ◽  
Vol 2 (1) ◽  
Author(s):  
Xuesong Sang ◽  
Hongbin Wang ◽  
Yihui Chen ◽  
Qiuhong Guo ◽  
Ailing Lu ◽  
...  

AbstractInflammasomes are intracellular protein complexes that mediate maturation and secretion of the pro-inflammatory cytokines IL-1β and IL-18. Inflammasomes have been connected with various diseases, therefore the regulation of inflammasome activation is important for the development of novel therapies for many inflammatory syndromes. Vitamin C is an essential nutrient and has regulatory effects on immune cells. Here we report that vitamin C has an inhibitory effect on the activation of the NLRP3 inflammasome in vitro and in vivo. Mechanistically, this inhibition is through scavenging mitochondrial ROS but not through NF-κB inhibition. Moreover, specificity tests show that the AIM2 inflammasome and the NLRC4 inflammasome can also be inhibited by vitamin C. Our results have thus identified a new inflammasome regulator and provide therapeutic potential for inflammasome-associated diseases.


Bone Research ◽  
2022 ◽  
Vol 10 (1) ◽  
Author(s):  
Sheng Chen ◽  
Xiaohao Wu ◽  
Yumei Lai ◽  
Di Chen ◽  
Xiaochun Bai ◽  
...  

AbstractIntervertebral disc (IVD) degeneration (IVDD) is the main cause of low back pain with major social and economic burdens; however, its underlying molecular mechanisms remain poorly defined. Here we show that the focal adhesion protein Kindlin-2 is highly expressed in the nucleus pulposus (NP), but not in the anulus fibrosus and the cartilaginous endplates, in the IVD tissues. Expression of Kindlin-2 is drastically decreased in NP cells in aged mice and severe IVDD patients. Inducible deletion of Kindlin-2 in NP cells in adult mice causes spontaneous and striking IVDD-like phenotypes in lumbar IVDs and largely accelerates progression of coccygeal IVDD in the presence of abnormal mechanical stress. Kindlin-2 loss activates Nlrp3 inflammasome and stimulates expression of IL-1β in NP cells, which in turn downregulates Kindlin-2. This vicious cycle promotes extracellular matrix (ECM) catabolism and NP cell apoptosis. Furthermore, abnormal mechanical stress reduces expression of Kindlin-2, which exacerbates Nlrp3 inflammasome activation, cell apoptosis, and ECM catabolism in NP cells caused by Kindlin-2 deficiency. In vivo blocking Nlrp3 inflammasome activation prevents IVDD progression induced by Kindlin-2 loss and abnormal mechanical stress. Of translational significance, adeno-associated virus-mediated overexpression of Kindlin-2 inhibits ECM catabolism and cell apoptosis in primary human NP cells in vitro and alleviates coccygeal IVDD progression caused by mechanical stress in rat. Collectively, we establish critical roles of Kindlin-2 in inhibiting Nlrp3 inflammasome activation and maintaining integrity of the IVD homeostasis and define a novel target for the prevention and treatment of IVDD.


2021 ◽  
Vol 4 (Supplement_1) ◽  
pp. 206-207
Author(s):  
M Bording-Jorgensen ◽  
H Armstrong ◽  
E Wine

Abstract Background The etiology of Inflammatory Bowel Diseases is unknown; however, a dysfunctional immune response has been well characterized. The role of the NLRP3 inflammasome in IBD is controversial with both beneficial and detrimental results. This pathway is required for the secretion of the proinflammatory cytokine IL-1β. Extracellular ATP is a well characterized inflammasome activator, which we have previously shown can increase the ability of J774A.1 macrophages to clear the mouse pathogen Citrobacter rodentium in an in vitro environment through the generation of reactive oxygen species (ROS). Aims Our objectives were to determine: 1) if extracellular ATP was inducing mitochondrial stress, causing the production of ROS, leading to microbial death; and 2) what effects these activated macropahges have on naïve macrophages. Methods Murine macrophage J774A.1 cells were infected with C. rodentium; extracellular ATP was added as an inflammasome activator and YVAD as an inhibitor. Lysotracker red and MitoSOX were used to determine cellular location of bacteria and quantify mitochondrial ROS, respectively. Secreted cytokines were measured using ELISA and a proteome profiler, ROS was measured using DCFDA, Gasdermin D and Caspase 11 activities were determined by Western Blot. Supernatants taken from infected macrophages were filtered and then added to naïve macrophages during infection with C. rodentium. Results Activation of mitochondrial ROS by ATP was found to be independent of infection. Secreted cytokines sICAM-1, MIP-1α, and MCP-2 were all increased by ATP but not inhibited by YVAD. Cleavage of Gasdermin D was increased with the addition of ATP but not inhibited by YVAD whereas Caspase 11 was unchanged between treatments. Supernatants from ATP-induced macrophages were able to induce IL-1β secretion in naïve macrophages and increase bacterial killing. Conclusions Mitochondrial ROS production in response to extracellular ATP may be involved in the decrease of bacterial survival. ATP induces the secretion of cytokines, chemokines, and other factors that affect newly infected macrophages. Gasdermin D cleavage, independent of caspase 11, suggests that a noncannonical pathway is activated; this may explain the lack of pyroptotic cells in our study. In addition, we have shown that these macrophages are able to illicit the same behavior in naive macrophages, suggesting that a corrected dysfunctional pathway in macrophages can have beneficial effects downstream. Understanding how the NLRP3 inflammasome is activated and what the downstream pathways are may lead to potential therapies for inflammatory conditions, including IBD. Funding Agencies CCC, CIHR


2017 ◽  
Vol 37 (suppl_1) ◽  
Author(s):  
Sai Ma ◽  
Jiangwei Chen ◽  
Zhenli Luo ◽  
Yabin Wang ◽  
Feng Cao

Introduction: NLRP3 inflammasome mediated inflammatory factors secretion is critically involved in atherosclerosis (AS). Melatonin has anti-inflammatory properties. However, it is unknown whether melatonin is beneficial in AS. Hypothesis: Melatonin plays a beneficial role in AS by decreasing NLRP3 inflammasome activation in macrophages. Methods: AS model was induced with high fat diet in apoE -/- mice. Plaque stability was examined with historical staining. In vitro study was performed in ox-LDL treated RAW264.7 cells. NLRP3 inflammasome activation, inflammatory factors secretion, mitochondrial ROS generation, autophagy, mitophagy indexes and potential signaling pathways were investigated. Results: Historical staining results showed that melatonin treatment markedly alleviated AS plaque progression. Despite of unchanged protein expression, Sirt 3 activity was elevated in plaque tissue in melatonin treated mice. Melatonin attenuated NLRP3 inflammasome activation and inflammatory factors secretion in ox-LDL treated macrophages, while this protective effect was abolished by Sirt3-siRNA. Mitochondrial ROS (mitoROS), which was an inducer for NLRP3 inflammasome, was reduced by melatonin through the elimination of damaged mitochondria (mitophagy). Similar with Sirt3-siRNA, autophagy inhibitor 3-MA also abolished the effects of melatonin on mitoROS clearance, indicating the crucial role of autophagy and mitophagy in melatonin caused NLRP3 inactivation. Furthermore, melatonin protected against AS via Sirt3/FoxO3/Parkin signaling pathway. Conclusions: Melatonin prevented atherosclerotic progression. Melatonin reduced mitochondrial ROS through the activation of autophagy and mitophagy, thereby attenuating NLRP3 inflammasome activation in macrophages. Moreover, the protective effect of melatonin was mediated by Sirt3/FoxO3/Parkin signaling pathway. Our study provides insight into a new therapeutic target for AS.


Author(s):  
Zhiyong Zhou ◽  
Menghan He ◽  
Qingqing Zhao ◽  
Dongfan Wang ◽  
Changcheng Zhang ◽  
...  

Introduction:: Microglia-mediated inflammatory responses play a crucial role in aging-related neurodegenerative diseases. The TXNIP/NLRP3 pathway is a key pathway leading to microglial activation. Panax notoginseng saponins (PNS) have been widely used for the treatment of stroke in China. Objective:: This study evaluates the anti-neuroinflammatory effect of PNS and investigates the mechanism via TXNIPmediated NLRP3 inflammasome activation in aging rats. Materials and Methods:: Eighteen-month-old Sprague-Dawley rats were randomly divided into the aging control group and PNS treated groups (n=15 each group). For PNS-treated groups, rats were administrated food with PNS at the doses of 10 mg/kg and 30 mg/kg for consecutive 6 months until they were 24-month old. Rats from the aging control group were given the same food without PNS. Two-month-old rats were purchased and given the same food until 6-month old as the adult control group (n = 15). Then, the cortex and hippocampus were rapidly harvested and deposited. H&E staining was used to assess histo-morphological changes. Western blotting was carried out to detect the protein expression. Immunofluorescence was employed to measure the co-localization of NLRP3, TXNIP and Iba-1. In vitro model was established by LPS+ATP coincubation in the BV2 microglia cell line. Results:: Aging rats exhibited increased activation of microglia, accompanied by a high level of IL-1β expression. Meanwhile, aging rats showed enhanced protein expression of TXNIP and NLRP3 related molecules, which co-localized with microglia. PNS treatment effectively reduced the number of degenerated neurons and reversed the activation of the TXNIP/NLRP3 inflammatory pathway. In vitro results showed that PNS up to 100 μg / ml had no significant toxicity on BV2 microglia. Discussion:: PNS (25, 50 μg/ml) effectively reduced the inflammatory response induced by LPS and ATP co-stimulation, thus inhibiting the expression of TXNIP/NLRP3 pathway-related proteins. Conclusion:: PNS treatment improved aging-related neuronal damage through inhibiting TXNIP mediated NLRP3 inflammasome activation, which provided a potential target for the treatment of inflammatory-related neurodegenerative diseases.


Antioxidants ◽  
2021 ◽  
Vol 10 (7) ◽  
pp. 1020
Author(s):  
Burak Ibrahim Arioz ◽  
Emre Tarakcioglu ◽  
Melis Olcum ◽  
Sermin Genc

NLRP3 inflammasome is a part of the innate immune system and responsible for the rapid identification and eradication of pathogenic microbes, metabolic stress products, reactive oxygen species, and other exogenous agents. NLRP3 inflammasome is overactivated in several neurodegenerative, cardiac, pulmonary, and metabolic diseases. Therefore, suppression of inflammasome activation is of utmost clinical importance. Melatonin is a ubiquitous hormone mainly produced in the pineal gland with circadian rhythm regulatory, antioxidant, and immunomodulatory functions. Melatonin is a natural product and safer than most chemicals to use for medicinal purposes. Many in vitro and in vivo studies have proved that melatonin alleviates NLRP3 inflammasome activity via various intracellular signaling pathways. In this review, the effect of melatonin on the NLRP3 inflammasome in the context of diseases will be discussed.


Sign in / Sign up

Export Citation Format

Share Document