1
|
Du Y, Duan C, Zhang X, Shi S, Zhu X, Lyu M, Wei Y, Hu Y. Modulation of NLRP3 Inflammasome: Advantages of Chinese Herbal Medicine in Treating Myocardial Ischemia/Reperfusion Injury. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2025; 53:737-769. [PMID: 40374375 DOI: 10.1142/s0192415x25500284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2025]
Abstract
Myocardial ischemia/reperfusion (I/R) injury is characterized by severe inflammation and oxidative stress, and involves the recruitment and activation of immune cells, the release of pro-inflammatory cytokines, and the generation of reactive oxygen species (ROS). The NOD-like receptor protein 3 (NLRP3) inflammasome, a multiprotein complex, is activated when exposed to different danger signals like excessive ROS, changes in ionic flux, and mitochondrial dysfunction. Once the NLRP3 inflammasome is activated, it promotes the maturation and release of pro-inflammatory cytokines such as interleukin-1β and interleukin-18, which contributes to the inflammatory storm in myocardial I/R injury. This inflammatory cascade not only leads to adverse cardiac remodeling but also impairs cardiac function, ultimately exacerbating the clinical outcomes of myocardial infarction. Despite the critical role of the NLRP3 inflammasome in myocardial I/R injury, there is a significant absence of effective therapeutic strategies to address it in clinical practice. In recent years, Chinese herbal medicine has emerged as a promising candidate in the therapeutic landscape of myocardial I/R injury. Chinese herbal medicine exerts its cardioprotective effects through various mechanisms of inhibiting NLRP3 inflammasomes, including enhancing mitochondrial function, reducing ROS generation, inhibiting the release of pro-inflammatory cytokines, and suppressing pyroptosis. This review emphasizes the therapeutic potential of Chinese herbal medicine and its extracts to inhibit NLRP3 inflammasomes in an effort to develop effective treatments for myocardial I/R injury. It likewise summarizes the research results of Chinese herbal medicine interventions for myocardial I/R injury by the mechanism of regulating the NLRP3 inflammasome, providing insights for the development of effective treatments for myocardial I/R injury.
Collapse
Affiliation(s)
- Yihang Du
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chenglin Duan
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaohan Zhang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shuqing Shi
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xueping Zhu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Meng Lyu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yi Wei
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuanhui Hu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
2
|
Zhang QY, Liu HX. Insights into the role of FGF21 in coronary heart disease. Int J Biol Macromol 2024; 282:136911. [PMID: 39476920 DOI: 10.1016/j.ijbiomac.2024.136911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 11/11/2024]
Abstract
Coronary heart disease (CHD) remains a leading cause of global mortality, with an alarming increase in its incidence among the younger population in recent years. This has amplified the need for early diagnostic markers and advances in therapeutic strategies to improve patient outcomes. Fibroblast growth factor 21 (FGF21), an endocrine hormone crucial for the regulation of metabolic homeostasis, has garnered significant attention over the past decade, owing to its role in cardiovascular health. FGF21 exerts cardioprotective effects through various mechanisms, including regulation of myocardial energy metabolism, prevention of cardiac cell death, suppression of inflammation, and reduction of oxidative stress in the heart. Given these properties, FGF21 shows considerable promise as a pharmacological agent for the management of CHD. Moreover, it has emerged as a promising biomarker for the diagnosis and prognostic assessment of CHD. This review aims to clarify the molecular mechanisms underlying the favorable effects of FGF21 on CHD and its related risk factors, thereby providing valuable insights for future research on the role of FGF21 in CHD management.
Collapse
Affiliation(s)
- Qin-Yao Zhang
- Health Sciences Institute, China Medical University, Shenyang, China; Institute of Life Sciences, China Medical University, Shenyang, China; Liaoning Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang, China; Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Hui-Xin Liu
- Health Sciences Institute, China Medical University, Shenyang, China; Institute of Life Sciences, China Medical University, Shenyang, China; Liaoning Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang, China.
| |
Collapse
|
3
|
Pan W, Zhang J, Zhang L, Zhang Y, Song Y, Han L, Tan M, Yin Y, Yang T, Jiang T, Li H. Comprehensive view of macrophage autophagy and its application in cardiovascular diseases. Cell Prolif 2024; 57:e13525. [PMID: 37434325 PMCID: PMC10771119 DOI: 10.1111/cpr.13525] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 06/17/2023] [Accepted: 06/22/2023] [Indexed: 07/13/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the primary drivers of the growing public health epidemic and the leading cause of premature mortality and economic burden worldwide. With decades of research, CVDs have been proven to be associated with the dysregulation of the inflammatory response, with macrophages playing imperative roles in influencing the prognosis of CVDs. Autophagy is a conserved pathway that maintains cellular functions. Emerging evidence has revealed an intrinsic connection between autophagy and macrophage functions. This review focuses on the role and underlying mechanisms of autophagy-mediated regulation of macrophage plasticity in polarization, inflammasome activation, cytokine secretion, metabolism, phagocytosis, and the number of macrophages. In addition, autophagy has been shown to connect macrophages and heart cells. It is attributed to specific substrate degradation or signalling pathway activation by autophagy-related proteins. Referring to the latest reports, applications targeting macrophage autophagy have been discussed in CVDs, such as atherosclerosis, myocardial infarction, heart failure, and myocarditis. This review describes a novel approach for future CVD therapies.
Collapse
Affiliation(s)
- Wanqian Pan
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Jun Zhang
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Lei Zhang
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Yue Zhang
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Yiyi Song
- Suzhou Medical College of Soochow UniversitySuzhouChina
| | - Lianhua Han
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Mingyue Tan
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Yunfei Yin
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Tianke Yang
- Department of Ophthalmology, Eye Institute, Eye & ENT HospitalFudan UniversityShanghaiChina
- Department of OphthalmologyThe First Affiliated Hospital of USTC, University of Science and Technology of ChinaHefeiChina
| | - Tingbo Jiang
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Hongxia Li
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| |
Collapse
|
4
|
Gu X, Bao N, Zhang J, Huang G, Zhang X, Zhang Z, Du Y, Meng H, Liu J, Wu P, Wang X, Wang G. Muscone ameliorates myocardial ischemia‒reperfusion injury by promoting myocardial glycolysis. Heliyon 2023; 9:e22154. [PMID: 38045159 PMCID: PMC10692826 DOI: 10.1016/j.heliyon.2023.e22154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 11/05/2023] [Accepted: 11/05/2023] [Indexed: 12/05/2023] Open
Abstract
Objective The incidence of acute myocardial infarction (AMI) is increasing yearly. With the use of thrombolysis or percutaneous coronary intervention (PCI), the mortality rate of acute myocardial infarction has been significantly reduced. However, reperfusion can cause additional myocardial injury. There is still a lack of effective drugs to treat I/R injury, and it is urgent to find new therapeutic drugs. Methods In this study, network pharmacology was used to predict potential targets and biological processes involved in Muscone-mediated treatment of acute myocardial infarction. To model ischemia‒reperfusion injury, a hypoxia-reoxygenation model and in vivo ischemia‒reperfusion injury C57BL/6 mice model was constructed. Mice were treated with Muscone i.p. for 4 weeks. We detected the cardiac function on day 28.The expression levels of the apoptotic proteins Caspase-3 and Bax and the anti-apoptotic protein Bcl-2 were detected by immunoblotting after Muscone treatment of AC16 cells and in vivo. Additionally, the gene expression levels of the PUMA and p53 were analyzed by qRT‒PCR. Molecular docking was used to evaluate the binding energy between Muscone and NLRP3-related proteins. Immunoblotting and qRT‒PCR were used to assess the expression levels of NLRP3 signaling pathway-related proteins (NLRP3, ASC, and Caspase-1) and the NLRP3 gene, respectively. Moreover, the extracellular acidification rate of AC16 cells was measured using the Seahorse system to evaluate glycolysis levels after Muscone treatment. The expression of the key glycolytic enzyme PKM2 was analyzed by immunoblotting and qRT‒PCR. Finally, ChIP‒qPCR was performed to determine the levels of histone modifications (H3K4me3, H3K27me3, and H2AK119Ub) in the PKM2 promoter region. Results GO functional enrichment analysis revealed that muscone was involved in regulating the biological processes (BP) of AMI, which mainly included negative regulation of the apoptosis signaling pathway, the response to lipopolysaccharide, and blood pressure regulation. The cellular components (CC) involved in muscone-mediated regulation of AMI mainly included lipid rafts, membrane microdomains, and membrane regions. The molecular functions (MF) involved in muscone-mediated regulation of AMI mainly included oxidoreductase activity, nuclear receptor activity, and transcription factor activity. In vitro results indicated that muscone treatment could inhibit the expression levels of Bax and Caspase-3 in AC16 cells after ischemia‒reperfusion while increasing the expression level of the antiapoptotic protein Bcl-2. Muscone significantly suppressed the transcription levels of p53 and PUMA in AC16 cells. Molecular docking suggested that muscone could bind well with the Cryo-EM structure of NEK7(PDB ID:6NPY). Further investigation of inflammatory pathways revealed that muscone could inhibit the expression level of NLRP3 in AC16 cells and reduce the expression levels of Caspase-1 and Caspase recruitment domain. Fluorescent quantitative PCR experiments showed that muscone significantly inhibited the transcription of NLRP3. Moreover, we found that muscone could enhance the glycolytic efficiency of AC16 cells, which may be related to the increased protein expression of PKM2 in AC16 cells. Fluorescent quantitative PCR showed that muscone could increase the transcription level of PKM2. Chromatin immunoprecipitation assays showed that muscone treatment increased the expression level of H3K4me3 in the PKM2 promoter region and inhibited the levels of H3K27me3 and H2AK119Ub in the PKM2 promoter region. Conclusion Muscone promoted myocardial glycolysis and inhibited NLRP3 pathway activation to improve myocardial ischemia‒reperfusion injury.
Collapse
Affiliation(s)
- Xin Gu
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214000, China
| | - Neng Bao
- Department of Nephrology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214000, China
| | - Jing Zhang
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214000, China
| | - Guangyi Huang
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214000, China
| | - Xiaodong Zhang
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214000, China
| | - Zhixuan Zhang
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214000, China
| | - Yinqiang Du
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, 215008, China
| | - Haoyu Meng
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
| | - Jiabao Liu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
| | - Peng Wu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
| | - Xiaoyan Wang
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214000, China
| | - Guangyan Wang
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214000, China
| |
Collapse
|
5
|
Xia Z, Sun G. Blocking RIPK2 Function Alleviates Myocardial Ischemia/Reperfusion Injury by Regulating the AKT and NF-κB Pathways. Immunol Invest 2023:1-17. [PMID: 37128885 DOI: 10.1080/08820139.2023.2203715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
OBJECTIVE Inflammation and oxidation brought on by myocardial ischemia-reperfusion (MI/R) injury lead to cardiomyocyte apoptosis and necrosis. The receptor interacting serine/threonine kinase 2 (RIPK2) plays significant roles in oxidative stress and excessive inflammation. The purpose of this research is to examine the roles of RIPK2 in MI/R injury. METHODS The in vivo animal model was constructed by acute coronary I/R, and the in vitro cell model was established by oxygen and glucose deprivation/reperfusion (OGD/R)-stimulated cardiomyocyte injury. RIPK2 expression was examined using qRT-PCR and Western blot. CCK-8 was proposed as a method for detecting cell proliferation. ELISA was utilized to measure inflammatory cytokines (TNF-α, IL-6, and IL-1β) and myocardial injury indicators (CK-MB, Mb, cTnI, and LDH). The levels of MDA and ROS were determined by the kit and fluorescent probe. H&E was conducted to assess MI/R injury after silencing of RIPK2. RESULTS In MI/R rats and OGD/R-treated H9C2 cardiomyocytes, RIPK2 was overexpressed at both the mRNA and protein levels. RIPK2 inhibition promoted cell proliferation while inhibiting apoptosis, as evidenced by decreased TUNEL-positive cells and cleaved caspase-3. RIPK2 inhibition reduced MDA and ROS levels, as well as the contents of inflammatory factors. RIPK2 silencing reduced CK-MB, Mb, cTnI, and LDH levels in rat serum and alleviated MI/R injury. Furthermore, RIPK2 inhibition increased p-AKT while decreasing NF-B p-p65 expression. CONCLUSION Silencing of RIPK2 reduced apoptosis, proinflammatory factors, and oxidative stress in MI/R by activating AKT and suppressing NF-κB signals, suggesting a potential therapeutic strategy for MI/R injury.
Collapse
Affiliation(s)
- Zhen Xia
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. china
| | - Guofang Sun
- Department of Electrocardiogram Diagnosis, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. china
| |
Collapse
|
6
|
Mokhtari B, Badalzadeh R. Protective and deleterious effects of autophagy in the setting of myocardial ischemia/reperfusion injury: an overview. Mol Biol Rep 2022; 49:11081-11099. [DOI: 10.1007/s11033-022-07837-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 08/01/2022] [Indexed: 11/30/2022]
|
7
|
NLRP3 Inflammasome/Pyroptosis: A Key Driving Force in Diabetic Cardiomyopathy. Int J Mol Sci 2022; 23:ijms231810632. [PMID: 36142531 PMCID: PMC9501057 DOI: 10.3390/ijms231810632] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 09/04/2022] [Accepted: 09/09/2022] [Indexed: 11/17/2022] Open
Abstract
Diabetic cardiomyopathy (DCM), a serious diabetic complication, is a kind of low-grade inflammatory cardiovascular disorder. Due to the high risk of morbidity and mortality, DCM has demanded the attention of medical researchers worldwide. The pathophysiological nature of DCM is intricate, and the genesis and development of which are a consequence of the coaction of many factors. However, the exact pathogenesis mechanism of DCM remains unclear. Pyroptosis is a newly identified programmed cell death (PCD) that is directly related to gasdermin D(GSDMD). It is characterized by pore formation on the cell plasma membrane, the release of inflammatory mediators, and cell lysis. The initiation of pyroptosis is closely correlated with NOD-like receptor 3 (NLRP3) activation, which activates caspase-1 and promotes the cleaving of GSDMD. In addition to adjusting the host’s immune defense, NLRP3 inflammasome/pyroptosis plays a critical role in controlling the systemic inflammatory response. Recent evidence has indicated that NLRP3 inflammasome/pyroptosis has a strong link with DCM. Targeting the activation of NLRP3 inflammasome or pyroptosis may be a hopeful therapeutic strategy for DCM. The focus of this review is to summarize the relevant mechanisms of pyroptosis and the relative contributions in DCM, highlighting the potential therapeutic targets in this field.
Collapse
|
8
|
Toldo S, Mezzaroma E, Buckley LF, Potere N, Di Nisio M, Biondi-Zoccai G, Van Tassell BW, Abbate A. Targeting the NLRP3 inflammasome in cardiovascular diseases. Pharmacol Ther 2022; 236:108053. [PMID: 34906598 PMCID: PMC9187780 DOI: 10.1016/j.pharmthera.2021.108053] [Citation(s) in RCA: 155] [Impact Index Per Article: 51.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 11/21/2021] [Accepted: 12/06/2021] [Indexed: 02/05/2023]
Abstract
The NACHT, leucine-rich repeat (LRR), and pyrin domain (PYD)-containing protein 3 (NLRP3) inflammasome is an intracellular sensing protein complex that plays a major role in innate immunity. Following tissue injury, activation of the NLRP3 inflammasome results in cytokine production, primarily interleukin(IL)-1β and IL-18, and, eventually, inflammatory cell death - pyroptosis. While a balanced inflammatory response favors damage resolution and tissue healing, excessive NLRP3 activation causes detrimental effects. A key involvement of the NLRP3 inflammasome has been reported across a wide range of cardiovascular diseases (CVDs). Several pharmacological agents selectively targeting the NLRP3 inflammasome system have been developed and tested in animals and early phase human studies with overall promising results. While the NLRP3 inhibitors are in clinical development, multiple randomized trials have demonstrated the safety and efficacy of IL-1 blockade in atherothrombosis, heart failure and recurrent pericarditis. Furthermore, the non-selective NLRP3 inhibitor colchicine has been recently shown to significantly reduce cardiovascular events in patients with chronic coronary disease. In this review, we will outline the mechanisms driving NLRP3 assembly and activation, and discuss the pathogenetic role of the NLRP3 inflammasome in CVDs, providing an overview of the current and future therapeutic approaches targeting the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Stefano Toldo
- VCU Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Eleonora Mezzaroma
- VCU Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA; Department of Pharmacotherapy and Outcome Studies, Virginia Commonwealth University, Richmond, VA, USA
| | - Leo F Buckley
- Department of Pharmacy, Brigham and Women's Hospital, Boston, MA, USA
| | - Nicola Potere
- Department of Innovative Technologies in Medicine and Dentistry, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Marcello Di Nisio
- Department of Medicine and Ageing Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Giuseppe Biondi-Zoccai
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy; Mediterranea Cardiocentro, Napoli, Italy
| | - Benjamin W Van Tassell
- VCU Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA; Department of Pharmacotherapy and Outcome Studies, Virginia Commonwealth University, Richmond, VA, USA
| | - Antonio Abbate
- VCU Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA; Wright Center for Clinical and Translational Research, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
9
|
Shen S, Wang Z, Sun H, Ma L. Role of NLRP3 Inflammasome in Myocardial Ischemia-Reperfusion Injury and Ventricular Remodeling. Med Sci Monit 2022; 28:e934255. [PMID: 35042840 PMCID: PMC8790935 DOI: 10.12659/msm.934255] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Reperfusion therapy is the optimal therapy for acute myocardial infarction (AMI), but acute inflammatory injury and chronic heart failure (HF) after myocardial ischemia and reperfusion (MI/R) remain the leading cause of death after AMI. Pyroptosis, a newly discovered form of cell death, has been proven to play a significant role in the acute reperfusion process and the subsequent chronic process of ventricular remodeling. Current research shows that multiple stimuli activate the pyroptotic signaling pathway and contribute to cell death and nonbacterial inflammation after MI/R. These stimuli promote the assembly of the nucleotide-binding and oligomerization-like receptor pyrin domain-containing protein 3 (NLRP3) inflammasome by activating NLRP3. The mature NLRP3 inflammasome cleaves procaspase-1 to active caspase-1, which leads to mature processing of interleukin (IL)-18, IL-1β, and gasdermin D (GSDMD) protein. That eventually results in cell lysis and generation of nonbacterial inflammation. The present review summarizes the mechanism of NLRP3 inflammasome activation after MI/R and discusses the role that NLRP3-mediated pyroptosis plays in the pathophysiology of MI/R injury and ventricular remodeling. We also discuss potential mechanisms and targeted therapy for which there is evidence supporting treatment of ischemic heart disease.
Collapse
Affiliation(s)
- Shichun Shen
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China (mainland)
| | - Zhen Wang
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China (mainland)
| | - Haozhong Sun
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China (mainland)
| | - Likun Ma
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China (mainland)
| |
Collapse
|
10
|
Zhang KZ, Shen XY, Wang M, Wang L, Sun HX, Li XZ, Huang JJ, Li XQ, Wu C, Zhao C, Liu JL, Lu X, Gao W. Retinol-Binding Protein 4 Promotes Cardiac Injury After Myocardial Infarction Via Inducing Cardiomyocyte Pyroptosis Through an Interaction With NLRP3. J Am Heart Assoc 2021; 10:e022011. [PMID: 34726071 PMCID: PMC8751920 DOI: 10.1161/jaha.121.022011] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Background Acute myocardial infarction (AMI) is one of the leading causes of cardiovascular morbidity and mortality worldwide. Pyroptosis is a form of inflammatory cell death that plays a major role in the development and progression of cardiac injury in AMI. However, the underlying mechanisms for the activation of pyroptosis during AMI are not fully elucidated. Methods and Results Here we show that RBP4 (retinol‐binding protein 4), a previous identified proinflammatory adipokine, was increased both in the myocardium of left anterior descending artery ligation‐induced AMI mouse model and in ischemia‐hypoxia‒induced cardiomyocyte injury model. The upregulated RBP4 may contribute to the activation of cardiomyocyte pyroptosis in AMI because overexpression of RBP4 activated NLRP3 (nucleotide‐binding oligomerization domain‐like receptor family pyrin domain‐containing 3) inflammasome, promoted the precursor cleavage of Caspase‐1, and subsequently induced GSDMD (gasdermin‐D)‐dependent pyroptosis. In contrast, knockdown of RBP4 alleviated ischemia‐hypoxia‒induced activation of NLRP3 inflammasome signaling and pyroptosis in cardiomyocytes. Mechanistically, coimmunoprecipitation assay showed that RBP4 interacted directly with NLRP3 in cardiomyocyte, while genetic knockdown or pharmacological inhibition of NLRP3 attenuated RBP4‐induced pyroptosis in cardiomyocytes. Finally, knockdown of RBP4 in heart decreased infarct size and protected against AMI‐induced pyroptosis and cardiac dysfunction in mice. Conclusions Taken together, these findings reveal RBP4 as a novel modulator promoting cardiomyocyte pyroptosis via interaction with NLRP3 in AMI. Therefore, targeting cardiac RBP4 might represent a viable strategy for the prevention of cardiac injury in patients with AMI.
Collapse
Affiliation(s)
- Kang-Zhen Zhang
- Department of Geriatrics Sir Run Run HospitalNanjing Medical University Nanjing China.,Key Laboratory for Aging and Disease Nanjing Medical University Nanjing China.,Department of Geriatrics The Second Affiliated Hospital of Nanjing Medical University Nanjing China
| | - Xi-Yu Shen
- Department of Geriatrics Sir Run Run HospitalNanjing Medical University Nanjing China.,Key Laboratory for Aging and Disease Nanjing Medical University Nanjing China
| | - Man Wang
- Department of Geriatrics Sir Run Run HospitalNanjing Medical University Nanjing China.,Key Laboratory for Aging and Disease Nanjing Medical University Nanjing China
| | - Li Wang
- Department of Geriatrics Sir Run Run HospitalNanjing Medical University Nanjing China.,Key Laboratory for Aging and Disease Nanjing Medical University Nanjing China
| | - Hui-Xian Sun
- Department of Geriatrics Sir Run Run HospitalNanjing Medical University Nanjing China.,Key Laboratory for Aging and Disease Nanjing Medical University Nanjing China
| | - Xiu-Zhen Li
- Key Laboratory for Aging and Disease Nanjing Medical University Nanjing China.,Department of Geriatrics The Second Affiliated Hospital of Nanjing Medical University Nanjing China
| | - Jing-Jing Huang
- Key Laboratory for Aging and Disease Nanjing Medical University Nanjing China.,Department of Geriatrics The Second Affiliated Hospital of Nanjing Medical University Nanjing China
| | - Xiao-Qing Li
- Key Laboratory for Aging and Disease Nanjing Medical University Nanjing China.,Department of Geriatrics The Second Affiliated Hospital of Nanjing Medical University Nanjing China
| | - Cheng Wu
- Key Laboratory for Aging and Disease Nanjing Medical University Nanjing China.,Department of Geriatrics The Second Affiliated Hospital of Nanjing Medical University Nanjing China
| | - Can Zhao
- Department of Geriatrics Sir Run Run HospitalNanjing Medical University Nanjing China.,Key Laboratory for Aging and Disease Nanjing Medical University Nanjing China
| | - Jia-Li Liu
- Department of Geriatrics Sir Run Run HospitalNanjing Medical University Nanjing China.,Key Laboratory for Aging and Disease Nanjing Medical University Nanjing China
| | - Xiang Lu
- Department of Geriatrics Sir Run Run HospitalNanjing Medical University Nanjing China.,Key Laboratory for Aging and Disease Nanjing Medical University Nanjing China.,Department of Geriatrics The Second Affiliated Hospital of Nanjing Medical University Nanjing China
| | - Wei Gao
- Department of Geriatrics Sir Run Run HospitalNanjing Medical University Nanjing China.,Key Laboratory for Aging and Disease Nanjing Medical University Nanjing China
| |
Collapse
|
11
|
Sun W, Lu H, Dong S, Li R, Chu Y, Wang N, Zhao Y, Zhang Y, Wang L, Sun L, Lu D. Beclin1 controls caspase-4 inflammsome activation and pyroptosis in mouse myocardial reperfusion-induced microvascular injury. Cell Commun Signal 2021; 19:107. [PMID: 34732218 PMCID: PMC8565084 DOI: 10.1186/s12964-021-00786-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 09/06/2021] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Myocardial reperfusion injury is often accompanied by cell death and inflammatory reactions. Recently, pyroptosis is gradually recognized as pivotal role in cardiovascular disease. However, little is known about the regulatory role of beclin1 in the control of caspase-4 activation and pyroptosis. The present study confirmed whether beclin1 regulates caspase-4 mediated pyroptosis and thereby protects Human Cardiac microvascular endothelial cells (HCMECs) against injury. METHODS TTC and Evan's blue dye, western blot, immunofluorescence and immunohistochemistry staining were performed in wild mice and transgenic mice with overexpression of beclin 1(BECN1-Tg). CMECs were transfected with a beclin1 lentivirus. The cell cytotoxicity was analyzed by LDH-Cytotoxicity Assay Kit. The protein levels of autophagy protein (Beclin1, p62 and LC3II/LC3I) and caspase-4/GSDMD pathway were determined by western blot. Autophagic vacuoles in cells were monitored with RFP-GFP-LC3 using fluorescence microscope. RESULTS I/R caused caspase-4 activity and gasdermin D expression increase in vivo and in vitro. Overexpression of beclin1 in heart tissue and CMECs suppressed the caspase-4 activity and decreased the levels of gasdermin D; meanwhile beclin1 overexpression also reduced IL-1β levels, promoted autophagy (p62 expression was inhibited while LC3II expression was increased) in the heart and CMECs. Interestingly, beclin1 overexpression increased animal survival and attenuated myocardial infarct size (45 ± 6.13 vs 22 ± 4.37), no-reflow area (39 ± 5.22 vs 16 ± 2.54) post-myocardial ischemia reperfusion. CONCLUSIONS Induction of beclin-1 signaling can be a potential therapeutic target in myocardial reperfusion-induced microvascular injury. Video Abstract.
Collapse
Affiliation(s)
- Wenjing Sun
- Department of Cardiology, Henan Provincial People’s Hospital, Zhengzhou, 450000 China
| | - Hongquan Lu
- Department of Nuclear Medicine, Third People’s Hospital of Honghe State, Honghe, 661000 China
| | - Shujuan Dong
- Department of Cardiology, Henan Provincial People’s Hospital, Zhengzhou, 450000 China
| | - Rui Li
- Department of Cardiology, Yunnan Geriatric Hospital, Kunming, 650501 China
| | - Yingjie Chu
- Department of Cardiology, Henan Provincial People’s Hospital, Zhengzhou, 450000 China
| | - Nan Wang
- Science and Technology Achievement Incubation Center, Kunming Medical University, Chenggong District, 1168 West Chunrong Road, Yuhua Avenue, Kunming, 650500 Yunnan China
| | - Yu Zhao
- Science and Technology Achievement Incubation Center, Kunming Medical University, Chenggong District, 1168 West Chunrong Road, Yuhua Avenue, Kunming, 650500 Yunnan China
| | - Yabin Zhang
- Department of Nuclear Medicine, Third People’s Hospital of Honghe State, Honghe, 661000 China
| | - Limeiting Wang
- Science and Technology Achievement Incubation Center, Kunming Medical University, Chenggong District, 1168 West Chunrong Road, Yuhua Avenue, Kunming, 650500 Yunnan China
| | - Lin Sun
- Department of Cardiology, The Second Affiliated Hospital, Kunming Medical University, 374 Dianmian Road, Wuhua District, Kunming, 650101 China
| | - Di Lu
- Science and Technology Achievement Incubation Center, Kunming Medical University, Chenggong District, 1168 West Chunrong Road, Yuhua Avenue, Kunming, 650500 Yunnan China
| |
Collapse
|
12
|
Zhao G, Zhang H, Zhu S, Wang S, Zhu K, Zhao Y, Xu L, Zhang P, Xie J, Sun A, Zou Y, Ge J. Interleukin-18 accelerates cardiac inflammation and dysfunction during ischemia/reperfusion injury by transcriptional activation of CXCL16. Cell Signal 2021; 87:110141. [PMID: 34487815 DOI: 10.1016/j.cellsig.2021.110141] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 08/30/2021] [Accepted: 08/31/2021] [Indexed: 01/03/2023]
Abstract
Myocardial ischemia/reperfusion(I/R) injury elicits an inflammatory response that drives tissue damage and cardiac remodeling. The trafficking and recruitment of inflammatory cells are controlled by C-X-C motif chemokine ligands and their receptors. CXCL16, a hallmark of acute coronary syndromes, is responsible for the recruitment of macrophages, monocytes and T lymphocytes. However, its role in cardiac I/R injury remains poorly characterized. Here we reported that CXCL16-mediated cardiac infiltration of CD11b+Ly6C+ cells played a crucial role in IL-18-induced myocardial inflammation, apoptosis and left ventricular(LV) dysfunction during I/R. Treatment with CXCL16 shRNA attenuated I/R-induced cardiac injury, LV remodeling and cardiac inflammation by reducing the recruitment of inflammatory cells and the release of TNFα, IL-17 and IFN-γ in the heart. We found that I/R-mediated NLRP3/IL-18 signaling pathway triggered CXCL16 transcription in cardiac vascular endothelial cells(VECs). Two binding sites of FOXO3 were found at the promoter region of CXCL16. By luciferase report assay and ChIP analysis, we confirmed that FOXO3 was responsible for endothelial CXCL16 transcription. A pronounced reduction of CXCL16 was observed in FOXO3 siRNA pretreated-VECs. Further experiments revealed that IL-18 activated FOXO3 by promoting the phosphorylation of STAT3 but not STAT4. An interaction between FOXO3 and STAT3 enhanced the transcription of CXCL16 induced by FOXO3. Treatment with Anakinra or Stattic either effectively inhibited IL-18-mediated nuclear import of FOXO3 and CXCL16 transcription. Our findings suggested that IL-18 accelerated I/R-induced cardiac damage and dysfunction through activating CXCL-16 and CXCL16-mediated cardiac infiltration of the CD11b+Ly6C+ cells. CXCL16 might be a novel therapeutic target for the treatment of I/R-related ischemic heart diseases.
Collapse
Affiliation(s)
- Gang Zhao
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China; NHC Key Laboratory of Viral Heart Diseases, Shanghai, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China; Department of Cardiology, Kashgar Prefecture Second People's Hospital, Kashi, China
| | - Hongqiang Zhang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shijie Zhu
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shijun Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; NHC Key Laboratory of Viral Heart Diseases, Shanghai, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Kai Zhu
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yun Zhao
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lei Xu
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China; NHC Key Laboratory of Viral Heart Diseases, Shanghai, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Ping Zhang
- Department of Cardiology, Kashgar Prefecture Second People's Hospital, Kashi, China
| | - Jing Xie
- Department of Cardiology, Kashgar Prefecture Second People's Hospital, Kashi, China
| | - Aijun Sun
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; NHC Key Laboratory of Viral Heart Diseases, Shanghai, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Yunzeng Zou
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China; NHC Key Laboratory of Viral Heart Diseases, Shanghai, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Junbo Ge
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China; NHC Key Laboratory of Viral Heart Diseases, Shanghai, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China.
| |
Collapse
|
13
|
The Interplay between Autophagy and NLRP3 Inflammasome in Ischemia/Reperfusion Injury. Int J Mol Sci 2021; 22:ijms22168773. [PMID: 34445481 PMCID: PMC8395601 DOI: 10.3390/ijms22168773] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 07/29/2021] [Accepted: 08/12/2021] [Indexed: 12/13/2022] Open
Abstract
Ischemia/reperfusion (I/R) injury is characterized by a limited blood supply to organs, followed by the restoration of blood flow and reoxygenation. In addition to ischemia, blood flow recovery can also lead to very harmful injury, especially inflammatory injury. Autophagy refers to the transport of cellular materials to the lysosomes for degradation, leading to the conversion of cellular components and offering energy and macromolecular precursors. It can maintain the balance of synthesis, decomposition and reuse of the intracellular components, and participate in many physiological processes and diseases. Inflammasomes are a kind of protein complex. Under physiological and pathological conditions, as the cellular innate immune signal receptors, inflammasomes sense pathogens to trigger an inflammatory response. TheNLRP3 inflammasome is the most deeply studied inflammasome and is composed of NLRP3, the adaptor apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) and pro-caspase-1. Its activation triggers the cleavage of pro-interleukin (IL)-1β and pro-IL-18 mediated by caspase-1 and promotes a further inflammatory process. Studies have shown that autophagy and the NLRP3 inflammasome play an important role in the process of I/R injury, but the relevant mechanisms have not been fully explained, especially how the interaction between autophagy and the NLRP3 inflammasome participates in I/R injury, which remains to be further studied. Therefore, we reviewed the recent studies about the interplay between autophagy and the NLRP3 inflammasome in I/R injury and analyzed the mechanisms to provide the theoretical references for further research in the future.
Collapse
|
14
|
Qu D, Guo H, Xu Y. Effects of Tranilast on Inflammasome and Macrophage Phenotype in a Mouse Model of Myocardial Infarction. J Interferon Cytokine Res 2021; 41:102-110. [PMID: 33750216 DOI: 10.1089/jir.2020.0208] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Acute myocardial infarction (AMI) has been a devastating actuality and accounts for half of cardiovascular emergency department visits. Nucleotide oligomerization domain, leucine-rich repeat, and pyrin domain-containing protein 3 (NLRP3) inflammasome participates in the mediation of myocardial inflammation during AMI. Therefore, this study aimed to reveal the therapeutic function of tranilast, an agent targeting NLRP3, for AMI. AMI mouse model was first established by transient myocardial ischemia. Western blot and quantitative reverse transcription polymerase chain reaction assay were performed to estimate the expression levels of related genes. Flow cytometry was used to analyze the macrophage types, and the therapeutic effects of tranilast were estimated by echocardiographic analysis and Masson's trichrome stain. We demonstrated that AMI induced the activation of NLRP3 inflammasome in the heart tissues of mice with AMI. Tranilast decreased the expression of interleukin-1β and cleaved caspase-1 in bone marrow-derived macrophages and thus re-educated M1-macrophages toward the M2-phenotype both in vitro and in vivo. Tranilast inhibited the activation in the heart tissues of AMI mice and thus improved cardiac functional recovery in the AMI mouse model. In conclusion, we revealed that tranilast ameliorated myocardial infarction by inhibiting NLRP3 inflammasome and re-educating macrophage phenotype in this study.
Collapse
Affiliation(s)
- Di Qu
- Department of Internal Medicine-Cardiovascular, Shengzhou People's Hospital (the First Affiliated Hospital of Zhejiang University Shengzhou Branch), Shengzhou, Zhejiang, China
| | - Huihui Guo
- Department of Internal Medicine-Cardiovascular, Shengzhou People's Hospital (the First Affiliated Hospital of Zhejiang University Shengzhou Branch), Shengzhou, Zhejiang, China
| | - Yanan Xu
- Rehabilitation Center, Beijing Xiaotangshan Hospital, Beijing, China
| |
Collapse
|
15
|
Deng J, Guo M, Li G, Xiao J. Gene therapy for cardiovascular diseases in China: basic research. Gene Ther 2020; 27:360-369. [PMID: 32341485 DOI: 10.1038/s41434-020-0148-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/26/2020] [Accepted: 04/02/2020] [Indexed: 12/14/2022]
Abstract
Cardiovascular disease has become a major disease affecting health in the whole world. Gene therapy, delivering foreign normal genes into target cells to repair damages caused by defects and abnormal genes, shows broad prospects in treating different kinds of cardiovascular diseases. China has achieved great progress of basic gene therapy researches and pathogenesis of cardiovascular diseases in recent years. This review will summarize the latest research about gene therapy of proteins, epigenetics, including noncoding RNAs and genome-editing technology in myocardial infarction, cardiac ischemia-reperfusion injury, atherosclerosis, muscle atrophy, and so on in China. We wish to highlight some important findings about the essential roles of basic gene therapy in this field, which might be helpful for searching potential therapeutic targets for cardiovascular disease.
Collapse
Affiliation(s)
- Jiali Deng
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, 200444, China
| | - Mengying Guo
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, 200444, China.,School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Guoping Li
- Cardiovascular Division of the Massachusetts, General Hospital and Harvard Medical School, Boston, MA, 02215, USA
| | - Junjie Xiao
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, 200444, China. .,School of Medicine, Shanghai University, Shanghai, 200444, China.
| |
Collapse
|
16
|
Ye JX, Wang M, Wang RY, Liu HT, Qi YD, Fu JH, Zhang Q, Zhang BG, Sun XB. Hydroxysafflor yellow A inhibits hypoxia/reoxygenation-induced cardiomyocyte injury via regulating the AMPK/NLRP3 inflammasome pathway. Int Immunopharmacol 2020; 82:106316. [PMID: 32088642 DOI: 10.1016/j.intimp.2020.106316] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 01/23/2020] [Accepted: 02/12/2020] [Indexed: 02/06/2023]
Abstract
Hydroxysafflor yellow A (HSYA) is an effective therapeutic agent that alleviates myocardial ischaemia/reperfusion injury (MIRI), but the exact mechanisms remain elusive. The aim of this study was to investigate the potential protective effect of HSYA against MIRI through mechanisms related to NLRP3 inflammasome regulation. In this study, hypoxia/reoxygenation (H/R)-induced H9c2 cardiomyocytes were treated with HSYA or the AMPK inhibitor, compound C (CC). Our results showed that HSYA pretreatment improved cardiomyocyte viability, maintained mitochondrial membrane potential, reduced apoptotic cardiomyocytes, decreased caspase-3 activity, and inhibited NOD-like receptor 3 (NLRP3) inflammasome activation during H/R injury. Moreover, the inhibition of AMPK activation by the CC inhibitor partially abolished the effects of HSYA treatment, including suppressing the upregulation of NLRP3 inflammasome components (NLRP3, caspase-1 and interleukin-1β) and promoting autophagy (LC3-II/LC3-I and p62). In conclusion, the protective mechanism of HSYA in H/R-induced cardiomyocyte injury is associated with inhibiting NLRP3 inflammasome activation through the AMPK signalling pathway.
Collapse
Affiliation(s)
- Jing-Xue Ye
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, PR China
| | - Min Wang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, PR China
| | - Rui-Ying Wang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, PR China
| | - Hai-Tao Liu
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, PR China
| | - Yao-Dong Qi
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, PR China
| | - Jian-Hua Fu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, PR China
| | - Qiong Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, PR China.
| | - Ben-Gang Zhang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, PR China.
| | - Xiao-Bo Sun
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, PR China.
| |
Collapse
|
17
|
Shi H, Dong Z, Gao H. LncRNA TUG1 protects against cardiomyocyte ischaemia reperfusion injury by inhibiting HMGB1. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2020; 47:3511-3516. [PMID: 31432688 DOI: 10.1080/21691401.2018.1556214] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The aim of this study was to investigate whether lncRNA TUG1 could mediate the progression of ischemia-reperfusion injury following acute myocardial infraction. Mouse cardiomyocytes HL-1 cells were subjected to oxygen glucose deprivation followed by reperfusion (OGD/R) to induce myocardial I/R injury. The expression of TUG1 was detected by real-time PCR. Overexpression or down expression of TUG1 was performed in mouse HL-1 cardiomyocytes. The myocardial cell viability and apoptosis were respectively detected. In addition, the expression levels of inflammatory factors, apoptosis-related proteins and HMGB1 proteins were detected. Besides, an inhibitor of HMGB1 was used to treat cells to verify the relationship between TUG1 and HMGB1 protein. The expression of TUG1 was significantly up-regulated in OGD/R-induced myocardial HL-1 cells. The overexpression of TUG1-induced inflammation and apoptosis in OGD-R-induced myocardial HL-1 cells. Knock down of TUG1 protected OGD/R-induced myocardial I/R injury by inhibiting HMGB1 expression. Suppression of lncRNA TUG1 may prevent myocardial I/R injury following acute myocardial infarction via inhibiting HMGB1 expression.
Collapse
Affiliation(s)
- Hanyu Shi
- Cadre Health Section, Qilu Hospital of Shandong University , Ji'nan , China
| | - Zhenhua Dong
- Department of Endocrinology, Jinan Central Hospital , Ji'nan , China
| | - Haiqing Gao
- Cadre Health Section, Qilu Hospital of Shandong University , Ji'nan , China
| |
Collapse
|
18
|
Pu Q, Lin P, Wang Z, Gao P, Qin S, Cui L, Wu M. Interaction among inflammasome, autophagy and non-coding RNAs: new horizons for drug. PRECISION CLINICAL MEDICINE 2019; 2:166-182. [PMID: 31598387 PMCID: PMC6770284 DOI: 10.1093/pcmedi/pbz019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 08/22/2019] [Accepted: 08/25/2019] [Indexed: 02/07/2023] Open
Abstract
Autophagy and inflammasomes are shown to interact in various situations including
infectious disease, cancer, diabetes and neurodegeneration. Since multiple layers of
molecular regulators contribute to the interplay between autophagy and inflammasome
activation, the detail of such interplay remains largely unknown. Non-coding RNAs
(ncRNAs), which have been implicated in regulating an expanding list of cellular processes
including immune defense against pathogens and inflammatory response in cancer and
metabolic diseases, may join in the crosstalk between inflammasomes and autophagy in
physiological or disease conditions. In this review, we summarize the latest research on
the interlink among ncRNAs, inflammasomes and autophagy and discuss the emerging role of
these three in multiple signaling transduction pathways involved in clinical conditions.
By analyzing these intriguing interconnections, we hope to unveil the mechanism
inter-regulating these multiple processes and ultimately discover potential drug targets
for some refractory diseases.
Collapse
Affiliation(s)
- Qinqin Pu
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203, USA.,State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ping Lin
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203, USA
| | - Zhihan Wang
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Pan Gao
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shugang Qin
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Luqing Cui
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203, USA
| | - Min Wu
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203, USA
| |
Collapse
|
19
|
|
20
|
Aghaei M, Motallebnezhad M, Ghorghanlu S, Jabbari A, Enayati A, Rajaei M, Pourabouk M, Moradi A, Alizadeh AM, Khori V. Targeting autophagy in cardiac ischemia/reperfusion injury: A novel therapeutic strategy. J Cell Physiol 2019; 234:16768-16778. [PMID: 30807647 DOI: 10.1002/jcp.28345] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 01/17/2019] [Accepted: 01/22/2019] [Indexed: 12/31/2022]
Abstract
Acute myocardial infarction (AMI) is one of the leading causes of morbidity worldwide. Myocardial reperfusion is known as an effective therapeutic choice against AMI. However, reperfusion of blood flow induces ischemia/reperfusion (I/R) injury through different complex processes including ion accumulation, disruption of mitochondrial membrane potential, the formation of reactive oxygen species, and so forth. One of the processes that gets activated in response to I/R injury is autophagy. Indeed, autophagy acts as a "double-edged sword" in the pathology of myocardial I/R injury and there is a controversy about autophagy being beneficial or detrimental. On the basis of the autophagy effect and regulation on myocardial I/R injury, many studies targeted it as a therapeutic strategy. In this review, we discuss the role of autophagy in I/R injury and its targeting as a therapeutic strategy.
Collapse
Affiliation(s)
- Mehrdad Aghaei
- Rheumatology Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Morteza Motallebnezhad
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Student Research Committee, Iran University of Medical Sciences, Tehran, Iran
| | - Sajjad Ghorghanlu
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Ali Jabbari
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Ayesheh Enayati
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran.,Department of Pharmacognosy, Faculty of Pharmacy and Medicinal Plants Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Rajaei
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mona Pourabouk
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Alireza Moradi
- Department of Physiology, Medical School, Ardabil University of Medical Sciences, Ardabil, Iran
| | | | - Vahid Khori
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| |
Collapse
|
21
|
Liu Q, Zhang D, Hu D, Zhou X, Zhou Y. The role of mitochondria in NLRP3 inflammasome activation. Mol Immunol 2018; 103:115-124. [PMID: 30248487 DOI: 10.1016/j.molimm.2018.09.010] [Citation(s) in RCA: 331] [Impact Index Per Article: 47.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Revised: 09/10/2018] [Accepted: 09/13/2018] [Indexed: 12/19/2022]
Abstract
The NLRP3 inflammasome is a multiprotein platform which is activated upon cellular infection or stress. Its activation leads to caspase-1-dependent secretion of proinflammatory cytokines like interleukin-1β (IL-1β) and IL-18, and an inflammatory form of cell death termed as pyroptosis. Recent studies have unveiled the pivotal roles of mitochondria in initiation and regulation of the NLRP3 (nucleotide-binding domain, leucine-rich-repeat containing family, pyrin domain-containing 3) inflammasome. NLRP3 activators induce mitochondrial destabilization, NLRP3 deubiquitination, linear ubiquitination of ASC, and externalization or release of mitochondria-derived molecules such as cardiolipin and mitochondrial DNA. These molecules bind to NLRP3 that is translocated on mitochondria and activate the NLRP3 inflammasome. Here we review recently described mechanisms by which mitochondria regulate NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Qiuyun Liu
- College of Animal Science, Southwest University, Chongqing 402460, China
| | - Danyan Zhang
- College of Animal Science, Southwest University, Chongqing 402460, China
| | - Diyu Hu
- College of Animal Science, Southwest University, Chongqing 402460, China
| | - Xiangmei Zhou
- State Key Laboratories for Agrobiotechnology, Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Yang Zhou
- College of Animal Science, Southwest University, Chongqing 402460, China.
| |
Collapse
|