1
|
Sha Z, Liu W, Jiang T, Zhang K, Yu Z. Astragaloside IV induces the protective effect of bone marrow mesenchymal stem cells derived exosomes in acute myocardial infarction by inducing angiogenesis and inhibiting apoptosis. Biotechnol Genet Eng Rev 2024; 40:1438-1455. [PMID: 36971224 DOI: 10.1080/02648725.2023.2194087] [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: 02/09/2023] [Accepted: 03/17/2023] [Indexed: 03/29/2023]
Abstract
Bone marrow mesenchymal stem cells (BMECs)-derived exosomes (MSC-Exo) can improve acute myocardial infarction (AMI). Astragaloside IV (AS-IV) has also been reported to have cardioprotective pharmacological effects. However, it is not entirely clear whether AS-IV can improve AMI by inducing MSC-Exo. BMSCs and MSC-Exo were isolated and identified, and we also established the AMI rat model and the OGD/R model with H9c2 cells. After MSC-Exo or AS-IV-mediated MSC-Exo treatment, cell angiogenesis, migration, and apoptosis were evaluated by tube formation, wound healing, and TUNEL staining. The cardiac function of the rats was measured by echocardiography. The pathological changes and collagen deposition in rats were also assessed with Masson and Sirius red staining. The levels of α-SMA, CD31 and inflammatory factors were determined by immunohistochemistry and enzyme-linked immunosorbent assay (ELISA). In vitro, AS-IV-mediated MSC-Exo can significantly enhance the angiogenesis and migration of H9c2 cells induced by OGD/R, and significantly reduce their apoptosis. In vivo, AS-IV-mediated MSC-Exo can improve the cardiac function of rats, and attenuate pathological damage and collagen deposition in AMI model rats. In addition, AS-IV-mediated MSC-Exo can also promote angiogenesis and reduce inflammatory factors in rats with AMI. AS-IV-stimulated MSC-Exo can improve myocardial contractile function, myocardial fibrosis and angiogenesis, reduce inflammatory factors and induce apoptosis in rats after AMI.
Collapse
Affiliation(s)
- Zhongxin Sha
- Department of Hypertension, The Affiliated Hospital of Guizhou Medical University, Guiyang, P.R. China
| | - Wupeng Liu
- Department of Cardiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, P. R. China
| | - Tianpeng Jiang
- Department of Interventional Radiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, P.R. China
| | - Kaiping Zhang
- Department of Cardiology, Guihang 302 Hospital, Anshun, P.R. China
| | - Zhenqiu Yu
- Department of Hypertension, The Affiliated Hospital of Guizhou Medical University, Guiyang, P.R. China
| |
Collapse
|
2
|
He H, Su H, Chen X, Chen X, Yang S. Interference with GPR4 inactivates NLRP3 inflammasome signaling by inhibiting LPAR1 expression to ameliorate oxygen-glucose deprivation/reoxygenation-induced inflammation and apoptosis of cardiomyocytes. Prostaglandins Other Lipid Mediat 2024; 174:106863. [PMID: 38936540 DOI: 10.1016/j.prostaglandins.2024.106863] [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: 04/01/2024] [Revised: 06/17/2024] [Accepted: 06/21/2024] [Indexed: 06/29/2024]
Abstract
Myocardial ischemia/reperfusion (MI/R) injury is a detrimental disease with high mortality worldwide. We aimed to explore the role of G protein-coupled receptor 4 (GPR4) and lysophosphatidic acid receptor 1 (LPAR1) in MI/R injury in vitro. H9c2 cells were exposed to oxygen-glucose deprivation/reoxygenation (OGD/R) conditions to simulate the MI/R injury and GPR4 expression was detected. Then, GPR4 was knocked down and cell viability was examined with a CCK-8 assay. The activities of LDH, CK and CK-MB were detected to evaluate the damage of OGD/R-induced H9c2 cells. ELISA kits and TUNEL staining were used to examine the inflammation and apoptosis of H9c2 cells exposed to OGD/R conditions. Western blot was employed to detect the expression of proteins related to apoptosis and NLRP3 inflammasome signaling. Additionally, Co-IP analyzed the binding between GPR4 and LPAR1. Finally, LPAR1 was overexpressed to conduct the rescue experiments. Results revealed that GPR4 was upregulated in OGD/R-treated H9c2 cells and GPR4 knockdown attenuated the damage of H9c2 cells. OGD/R induced inflammation and apoptosis were markedly inhibited by GPR4 silencing, as evidenced by the decreased TNF-α, IL-6 and IL-8 levels as well as the elevated Bcl-2 expression and reduced Bax and cleaved caspase3 expression. Moreover, GPR4 bound to LPAR1 and upregulated LPAR1 expression. Interference with GPR4 inactivated the NLRP3 inflammasome signaling. Besides, LPAR1 overexpression abrogated the effects of GPR4 silencing on the damage, inflammation and apoptosis of H9c2 cells induced by OGD/R. Particularly, LPAR1 upregulation promoted the activation of NLRP3 inflammasome signaling in GPR4-silenced H9c2 cells induced by OGD/R. To be concluded, GPR4 deficiency inactivates NLRP3 inflammasome signaling by inhibiting LPAR1 expression to ameliorate OGD/R -induced inflammation and apoptosis of cardiomyocytes.
Collapse
Affiliation(s)
- Hanlong He
- Radiology Department, Huizhou First Hospital, Huizhou, Guangdong 516001, China
| | - Huiren Su
- Radiology Department, Huizhou First Hospital, Huizhou, Guangdong 516001, China
| | - Xinjian Chen
- Radiology Department, Huizhou First Hospital, Huizhou, Guangdong 516001, China
| | - Xiaohong Chen
- Hand and Foot Microsurgery & Wound Repair Department, Huizhou First Hospital, Huizhou, Guangdong 516001, China
| | - Shaoze Yang
- Department of Clinical Medicine, School of Medicine, Etugen University, Ulaanbaatar 14191, Mongolia.
| |
Collapse
|
3
|
Suthivanich P, Boonhoh W, Sumneang N, Punsawad C, Cheng Z, Phungphong S. Aerobic Exercise Attenuates Doxorubicin-Induced Cardiomyopathy by Suppressing NLRP3 Inflammasome Activation in a Rat Model. Int J Mol Sci 2024; 25:9692. [PMID: 39273638 PMCID: PMC11395441 DOI: 10.3390/ijms25179692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 09/04/2024] [Accepted: 09/06/2024] [Indexed: 09/15/2024] Open
Abstract
Doxorubicin (DOX) is a potent chemotherapeutic agent with well-documented dose-dependent cardiotoxicity. Regular exercise is recognized for its cardioprotective effects against DOX-induced cardiac inflammation, although the precise mechanisms remain incompletely understood. The activation of inflammasomes has been implicated in the pathogenesis and treatment of DOX-induced cardiotoxicity, with the nucleotide-binding domain-like receptor protein 3 (NLRP3) inflammasome emerging as a key mediator in cardiovascular inflammation. This study aimed to investigate the role of exercise in modulating the NLRP3 inflammasome to protect against DOX-induced cardiac inflammation. Male Sprague-Dawley rats were randomly assigned to receive a 10-day course of DOX or saline injections, with or without a preceding 10-week treadmill running regimen. Cardiovascular function and histological changes were subsequently evaluated. DOX-induced cardiotoxicity was characterized by cardiac atrophy, systolic dysfunction, and hypotension, alongside activation of the NLRP3 inflammasome. Our findings revealed that regular exercise preserved cardiac mass and hypertrophic indices and prevented DOX-induced cardiac dysfunction, although it did not fully preserve blood pressure. These results underscore the significant cardioprotective effects of exercise against DOX-induced cardiotoxicity. While regular exercise did not entirely prevent DOX-induced hypotension, our findings demonstrate that it confers protection against DOX-induced cardiotoxicity by suppressing NLRP3 inflammasome activation in the heart, underscoring its anti-inflammatory role. Further research should explore the temporal dynamics and interactions among exercise, pyroptosis, and other pathways in DOX-induced cardiotoxicity to enhance translational applications in cardiovascular medicine.
Collapse
Affiliation(s)
- Phichaya Suthivanich
- Doctor of Philosophy Program in Physiology, Graduate School, Chulalongkorn University, Bangkok 10330, Thailand
| | - Worakan Boonhoh
- Akkhraratchakumari Veterinary College, Walailak University, Nakhon Si Thammarat 80161, Thailand
| | - Natticha Sumneang
- Department of Medical Sciences, School of Medicine, Walailak University, Nakhon Si Thammarat 80160, Thailand
- Research Center in Tropical Pathobiology, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Chuchard Punsawad
- Department of Medical Sciences, School of Medicine, Walailak University, Nakhon Si Thammarat 80160, Thailand
- Research Center in Tropical Pathobiology, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Zhaokang Cheng
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA
| | - Sukanya Phungphong
- Department of Medical Sciences, School of Medicine, Walailak University, Nakhon Si Thammarat 80160, Thailand
- Research Center in Tropical Pathobiology, Walailak University, Nakhon Si Thammarat 80160, Thailand
| |
Collapse
|
4
|
Huang XD, Jiang DS, Feng X, Fang ZM. The benefits of oral glucose-lowering agents: GLP-1 receptor agonists, DPP-4 and SGLT-2 inhibitors on myocardial ischaemia/reperfusion injury. Eur J Pharmacol 2024; 976:176698. [PMID: 38821168 DOI: 10.1016/j.ejphar.2024.176698] [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: 03/09/2024] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 06/02/2024]
Abstract
Myocardial infarction (MI) is a life-threatening cardiovascular disease that, on average, results in 8.5 million deaths worldwide each year. Timely revascularization of occluded vessels is a critical method of myocardial salvage. However, reperfusion paradoxically leads to the worsening of myocardial damage known as myocardial ischaemia/reperfusion injury (MI/RI). Therefore, reducing the size of myocardial infarction after reperfusion is critical and remains an important therapeutic goal. The susceptibility of the myocardium to MI/RI may be increased by diabetes. Currently, some traditional antidiabetic agents such as metformin reduce MI/RI by decreasing inflammation, inhibiting oxidative stress, and improving vascular endothelial function. This appears to be a new direction for the treatment of MI/RI. Recent cardiovascular outcome trials have shown that several oral antidiabetic agents, including glucagon-like peptide-1 receptor agonists (GLP-1RAs), dipeptidyl peptidase-4 inhibitors (DPP-4is), and sodium-glucose-linked transporter-2 inhibitors (SGLT-2is), not only have good antidiabetic effects but also have a protective effect on myocardial protection. This article aims to discuss the mechanisms and effects of oral antidiabetic agents, including GLP-1RAs, DPP-4is, and SGLT-2is, on MI/RI to facilitate their clinical application.
Collapse
Affiliation(s)
- Xu-Dong Huang
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Department of Cardiothoracic Surgery, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Ding-Sheng Jiang
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei, China
| | - Xin Feng
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Ze-Min Fang
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Department of Cardiothoracic Surgery, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, Hubei, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
5
|
Heger LA, Schommer N, Van Bruggen S, Sheehy CE, Chan W, Wagner DD. Neutrophil NLRP3 promotes cardiac injury following acute myocardial infarction through IL-1β production, VWF release and NET deposition in the myocardium. Sci Rep 2024; 14:14524. [PMID: 38914598 PMCID: PMC11196583 DOI: 10.1038/s41598-024-64710-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 06/12/2024] [Indexed: 06/26/2024] Open
Abstract
NLRP3 inflammasome has been implicated in neutrophil polarization and extrusion of neutrophil extracellular traps (NETs) in vitro and facilitates secretion of Il1-beta (IL-1β). Permanent ligation of the left anterior descending artery was used to induce MI in WT and NLRP3-/- mice as well as in NLRP3-/- recipient mice transfused with either WT or NLRP3-/- neutrophils. NLRP3 deficiency reduced infarct size to roughly a third of WT heart injury and preserved left ventricular (LV) function at 12 h after MI as assessed by echocardiography and triphenyltetrazolium chloride staining of live tissue. Transfusion of WT but not NLRP3-/- neutrophils after MI increased infarct size in NLRP3-/- mice and significantly reduced LV function. The key features of myocardial tissue in WT neutrophil transfused recipients were increased H3Cit-positive deposits with NET-like morphology and increased tissue levels of IL-1β and plasma levels of von Willebrand Factor (VWF). Flow cytometry analysis also revealed that neutrophil NLRP3 increased the number of labeled and transfused neutrophils in the bone marrow of recipient mice following MI. Our data suggest a key role for neutrophil NLRP3 in the production of IL-1β and deposition of NETs in cardiac tissue exacerbating injury following MI. We provide evidence for a link between neutrophil NLRP3 and VWF release likely enhancing thromboinflammation in the heart. Neutrophil NLRP3 deficiency conferred similar cardioprotective effects to general NLRP3 deletion in MI rendering anti-neutrophil NLRP3 therapy a promising target for early cardioprotective treatment.
Collapse
Affiliation(s)
- Lukas A Heger
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, 1 Blackfan Circle, Ninth Floor, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
- Departement of Cardiology and Angiology, University Hospital Freiburg Bad Krozingen, 79106, Freiburg, Germany
| | - Nicolas Schommer
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, 1 Blackfan Circle, Ninth Floor, Boston, MA, 02115, USA
- Departement of Cardiology and Angiology, University Hospital Freiburg Bad Krozingen, 79106, Freiburg, Germany
| | - Stijn Van Bruggen
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, 1 Blackfan Circle, Ninth Floor, Boston, MA, 02115, USA
- Center of Molecular and Vascular Biology, Department of Cardiovascular Science, KU Leuven, 3000, Leuven, Belgium
| | - Casey E Sheehy
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, 1 Blackfan Circle, Ninth Floor, Boston, MA, 02115, USA
| | - William Chan
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, 1 Blackfan Circle, Ninth Floor, Boston, MA, 02115, USA
| | - Denisa D Wagner
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, 1 Blackfan Circle, Ninth Floor, Boston, MA, 02115, USA.
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA.
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, 02115, USA.
| |
Collapse
|
6
|
Duan F, Li L, Liu S, Tao J, Gu Y, Li H, Yi X, Gong J, You D, Feng Z, Yu T, Tan H. Cortistatin protects against septic cardiomyopathy by inhibiting cardiomyocyte pyroptosis through the SSTR2-AMPK-NLRP3 pathway. Int Immunopharmacol 2024; 134:112186. [PMID: 38733824 DOI: 10.1016/j.intimp.2024.112186] [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: 04/02/2024] [Revised: 04/25/2024] [Accepted: 04/28/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND Although the pathophysiological mechanism of septic cardiomyopathy has been continuously discovered, it is still a lack of effective treatment method. Cortistatin (CST), a neuroendocrine polypeptide of the somatostatin family, has emerged as a novel cardiovascular-protective peptide, but the specific mechanism has not been elucidated. PURPOSE The aim of our study is to explore the role of CST in cardiomyocytes pyroptosis and myocardial injury in sepsis and whether CST inhibits cardiomyocytes pyroptosis through specific binding with somastatin receptor 2 (SSTR2) and activating AMPK/Drp1 signaling pathway. METHODS AND RESULTS In this study, plasma CST levels were significantly high and were negatively correlated with N-terminal pro-B type natriuretic peptide (NT-proBNP), a biomarker for cardiac dysfunction, in patients with sepsis. Exogenous administration of CST significantly improved survival rate and cardiac function in mouse models of sepsis by inhibiting the activation of the NLRP3 inflammasome and pyroptosis of cardiomyocytes (decreased cleavage of caspase-1, IL-1β and gasdermin D). Pharmacological inhibition and genetic ablation revealed that CST exerted anti-pyroptosis effects by specifically binding to somatostatin receptor subtype 2 (SSTR2), thus activating AMPK and inactivating Drp1 to inhibit mitochondrial fission in cardiomyocytes. CONCLUSIONS This study is the first to report that CST attenuates septic cardiomyopathy by inhibiting cardiomyocyte pyroptosis through the SSTR2-AMPK-Drp1-NLRP3 pathway. Importantly, CST specifically binds to SSTR2, which promotes AMPK phosphorylation, inhibits Drp1-mediated mitochondrial fission, and reduces ROS levels, thereby inhibiting NLRP3 inflammasome activation-mediated pyroptosis and alleviating sepsis-induced myocardial injury.
Collapse
Affiliation(s)
- Fengqi Duan
- Department of Pathophysiology, School of Medicine, Sun Yat-sen University, Shenzhen, 518107, Guangdong, China; Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Li Li
- Department of Emergency Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510012, Guangdong, China
| | - Sijun Liu
- Department of Pathophysiology, School of Medicine, Sun Yat-sen University, Shenzhen, 518107, Guangdong, China; Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Jun Tao
- Department of Pathophysiology, School of Medicine, Sun Yat-sen University, Shenzhen, 518107, Guangdong, China; Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Yang Gu
- Department of Emergency Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510012, Guangdong, China
| | - Huangjing Li
- Department of Pathophysiology, School of Medicine, Sun Yat-sen University, Shenzhen, 518107, Guangdong, China; Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Xiaoling Yi
- Department of Emergency Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510012, Guangdong, China
| | - Jianfeng Gong
- Department of Pathophysiology, School of Medicine, Sun Yat-sen University, Shenzhen, 518107, Guangdong, China; Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Daiting You
- Department of Pathophysiology, School of Medicine, Sun Yat-sen University, Shenzhen, 518107, Guangdong, China; Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Zejiang Feng
- Department of Pathophysiology, School of Medicine, Sun Yat-sen University, Shenzhen, 518107, Guangdong, China; Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Tao Yu
- Department of Emergency Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510012, Guangdong, China
| | - Hongmei Tan
- Department of Pathophysiology, School of Medicine, Sun Yat-sen University, Shenzhen, 518107, Guangdong, China; Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong, China; Laboratory Animal Center, Sun Yat-sen University, Guangzhou 510080, Guangdong, China.
| |
Collapse
|
7
|
Qin J, Yang Q, Wang Y, Shi M, Zhao X, Zhou Y. The role of pyroptosis in heart failure and related traditional chinese medicine treatments. Front Pharmacol 2024; 15:1377359. [PMID: 38868667 PMCID: PMC11168204 DOI: 10.3389/fphar.2024.1377359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 05/09/2024] [Indexed: 06/14/2024] Open
Abstract
Pyroptosis is a type of programmed cell death that is mediated by both typical and atypical pathways and ultimately leads to the lysis and rupture of cell membranes and the release of proinflammatory factors, triggering an intense inflammatory response. Heart failure (HF) is a serious and terminal stage of various heart diseases. Myocardial hypertrophy, myocardial fibrosis, ventricular remodeling, oxidative stress, the inflammatory response and cardiomyocyte ionic disorders caused by various cardiac diseases are all risk factors for and aggravate HF. Numerous studies have shown that pyroptosis can induce and exacerbate these reactions, causing progression to HF. Therefore, targeting pyroptosis is a promising strategy to treat HF. This paper summarizes the role of pyroptosis in the development of HF and the underlying mechanism involved. Recent research progress on the ability of traditional Chinese medicine (TCM) extracts and formulas to inhibit pyroptosis and treat HF was summarized, and some traditional Chinese medicine extracts and formulas can alleviate different types of HF, including heart failure with preserved ejection fraction (HFpEF), heart failure with reduced ejection fraction (HFrEF), and heart failure with midrange ejection fraction (HFmrEF), by targeting pyroptosis. These findings may provide new ideas and evidence for the treatment or adjuvant treatment of HF by targeting pyroptosis.
Collapse
Affiliation(s)
- Jie Qin
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Qianhe Yang
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Yan Wang
- Department of Cardiovascular Medicine, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Mengdi Shi
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Xin Zhao
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Yabin Zhou
- Department of Cardiovascular Medicine, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| |
Collapse
|
8
|
Cohen MV, Downey JM. Initial Despair and Current Hope of Identifying a Clinically Useful Treatment of Myocardial Reperfusion Injury: Insights Derived from Studies of Platelet P2Y 12 Antagonists and Interference with Inflammation and NLRP3 Assembly. Int J Mol Sci 2024; 25:5477. [PMID: 38791515 PMCID: PMC11122283 DOI: 10.3390/ijms25105477] [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: 03/28/2024] [Accepted: 05/11/2024] [Indexed: 05/26/2024] Open
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.
Collapse
Affiliation(s)
- Michael V. Cohen
- The Departments of Physiology and Cell Biology, Frederick P. Whiddon College of Medicine, Mobile, AL 36688, USA;
- The Departments of Medicine, Frederick P. Whiddon College of Medicine, Mobile, AL 36688, USA
| | - James M. Downey
- The Departments of Physiology and Cell Biology, Frederick P. Whiddon College of Medicine, Mobile, AL 36688, USA;
| |
Collapse
|
9
|
Hu Y, Dai S, Zhao L, Zhao L. Research progress on the improvement of cardiovascular diseases through the autonomic nervous system regulation of the NLRP3 inflammasome pathway. Front Cardiovasc Med 2024; 11:1369343. [PMID: 38650918 PMCID: PMC11034522 DOI: 10.3389/fcvm.2024.1369343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 03/28/2024] [Indexed: 04/25/2024] Open
Abstract
Cardiovascular disease stands as a leading global cause of mortality. Nucleotide-binding Oligomerization Domain-like Receptor Protein 3 (NLRP3) inflammasome is widely acknowledged as pivotal factor in specific cardiovascular disease progression, such as myocardial infarction, heart failure. Recent investigations underscore a close interconnection between autonomic nervous system (ANS) dysfunction and cardiac inflammation. It has been substantiated that sympathetic nervous system activation and vagus nerve stimulation (VNS) assumes critical roles withinNLRP3 inflammasome pathway regulation, thereby contributing to the amelioration of cardiac injury and enhancement of prognosis in heart diseases. This article reviews the nexus between NLRP3 inflammasome and cardiovascular disorders, elucidating the modulatory functions of the sympathetic and vagus nerves within the ANS with regard to NLRP3 inflammasome. Furthermore, it delves into the potential therapeutic utility of NLRP3 inflammasome to be targeted by VNS. This review serves as a valuable reference for further exploration into the potential mechanisms underlying VNS in the modulation of NLRP3 inflammasome.
Collapse
Affiliation(s)
| | | | - Lulu Zhao
- Department of Cardiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Ling Zhao
- Department of Cardiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
10
|
Jin L, Piao Z. Irisin protects against cardiac injury by inhibiting NLRP3 inflammasome-mediated pyroptosis during remodeling after infarction. Int Immunopharmacol 2024; 130:111714. [PMID: 38412677 DOI: 10.1016/j.intimp.2024.111714] [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: 12/13/2023] [Revised: 02/04/2024] [Accepted: 02/14/2024] [Indexed: 02/29/2024]
Abstract
This study aimed to explore the cardioprotective mechanism of irisin in the context of cardiac injury. Utilizing a myocardial infarction (MI) mouse model, we investigated the therapeutic potential of recombinant human irisin (rhIrisin) administered for 28 days post-infarction. The efficacy of irisin treatment was evaluated through echocardiographic assessment of cardiac function and serum analysis of myocardial injury markers. Our research provided novel insights into the impacts of irisin on the NLR Family Pyrin Domain Containing 3 (NLRP3) inflammasome activation and pyroptosis, assessed both in vivo in MI mice and in vitro in hypoxia/reoxygenation-treated H9C2 cells. Remarkably, irisin treatment significantly reduced levels of lactate dehydrogenase (LDH), creatine kinase-MB (CK-MB), and troponin I, indicating reduced myocardial injury. Echocardiography highlighted substantial improvements in left ventricular ejection fraction (LVEF), left ventricular fractional shortening (LVFS), and dimensions (LVIDd and LVIDs) in irisin-treated mice, underscoring enhanced cardiac function. Moreover, irisin was shown to significantly suppress the mRNA and protein expressions of key components involved in NLRP3 inflammasome pathway (NLRP3, ASC, caspase-1 (p20), and interleukin-18 (IL-18)) both in MI-induced mice and hypoxia/reoxygenation-treated cells. This study firstly reveals that the cardioprotective effect of irisin is mediated through the attenuation of NLRP3 inflammasome activation and pyroptosis, positioning irisin as a promising therapeutic agent for cardiac injury.
Collapse
Affiliation(s)
- Li Jin
- Department of Cardiology, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang, Wenzhou, Zhejiang, China
| | - Zhehao Piao
- Department of Cardiology, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China.
| |
Collapse
|
11
|
Liu CX, Guo XY, Zhou YB, Wang H. Therapeutic Role of Chinese Medicine Targeting Nrf2/HO-1 Signaling Pathway in Myocardial Ischemia/Reperfusion Injury. Chin J Integr Med 2024:10.1007/s11655-024-3657-0. [PMID: 38329655 DOI: 10.1007/s11655-024-3657-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/01/2023] [Indexed: 02/09/2024]
Abstract
Acute myocardial infarction (AMI), characterized by high incidence and mortality rates, poses a significant public health threat. Reperfusion therapy, though the preferred treatment for AMI, often exacerbates cardiac damage, leading to myocardial ischemia/reperfusion injury (MI/RI). Consequently, the development of strategies to reduce MI/RI is an urgent priority in cardiovascular therapy. Chinese medicine, recognized for its multi-component, multi-pathway, and multi-target capabilities, provides a novel approach for alleviating MI/RI. A key area of interest is the nuclear factor E2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) pathway. This pathway is instrumental in regulating inflammatory responses, oxidative stress, apoptosis, endoplasmic reticulum stress, and ferroptosis in MI/RI. This paper presents a comprehensive overview of the Nrf2/HO-1 signaling pathway's structure and its influence on MI/RI. Additionally, it reviews the latest research on leveraging Chinese medicine to modulate the Nrf2/HO-1 pathway in MI/RI treatment.
Collapse
Affiliation(s)
- Chang-Xing Liu
- First Clinical Medical School, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Xin-Yi Guo
- Clinical Medical School, Chengdu University of Traditional Chinese Medicine, Chengdu, 610036, China
| | - Ya-Bin Zhou
- Department of Cardiology, the First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - He Wang
- Department of Cardiology, the First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, 150040, China.
| |
Collapse
|
12
|
Hu C. Marine natural products and human immunity: novel biomedical resources for anti-infection of SARS-CoV-2 and related cardiovascular disease. NATURAL PRODUCTS AND BIOPROSPECTING 2024; 14:12. [PMID: 38282092 PMCID: PMC10822835 DOI: 10.1007/s13659-024-00432-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 01/17/2024] [Indexed: 01/30/2024]
Abstract
Marine natural products (MNPs) and marine organisms include sea urchin, sea squirts or ascidians, sea cucumbers, sea snake, sponge, soft coral, marine algae, and microalgae. As vital biomedical resources for the discovery of marine drugs, bioactive molecules, and agents, these MNPs have bioactive potentials of antioxidant, anti-infection, anti-inflammatory, anticoagulant, anti-diabetic effects, cancer treatment, and improvement of human immunity. This article reviews the role of MNPs on anti-infection of coronavirus, SARS-CoV-2 and its major variants (such as Delta and Omicron) as well as tuberculosis, H. Pylori, and HIV infection, and as promising biomedical resources for infection related cardiovascular disease (irCVD), diabetes, and cancer. The anti-inflammatory mechanisms of current MNPs against SARS-CoV-2 infection are also discussed. Since the use of other chemical agents for COVID-19 treatment are associated with some adverse effects in cardiovascular system, MNPs have more therapeutic advantages. Herein, it's time to protect this ecosystem for better sustainable development in the new era of ocean economy. As huge, novel and promising biomedical resources for anti-infection of SARS-CoV-2 and irCVD, the novel potential mechanisms of MNPs may be through multiple targets and pathways regulating human immunity and inhibiting inflammation. In conclusion, MNPs are worthy of translational research for further clinical application.
Collapse
Affiliation(s)
- Chunsong Hu
- Department of Cardiovascular Medicine, Jiangxi Academy of Medical Science, Nanchang University, Hospital of Nanchang University, No. 461 Bayi Ave, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
13
|
Zhang H, Hu H, Zhai C, Jing L, Tian H. Cardioprotective Strategies After Ischemia-Reperfusion Injury. Am J Cardiovasc Drugs 2024; 24:5-18. [PMID: 37815758 PMCID: PMC10806044 DOI: 10.1007/s40256-023-00614-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/28/2023] [Indexed: 10/11/2023]
Abstract
Acute myocardial infarction (AMI) is associated with high morbidity and mortality worldwide. Although early reperfusion is the most effective strategy to salvage ischemic myocardium, reperfusion injury can develop with the restoration of blood flow. Therefore, it is important to identify protection mechanisms and strategies for the heart after myocardial infarction. Recent studies have shown that multiple intracellular molecules and signaling pathways are involved in cardioprotection. Meanwhile, device-based cardioprotective modalities such as cardiac left ventricular unloading, hypothermia, coronary sinus intervention, supersaturated oxygen (SSO2), and remote ischemic conditioning (RIC) have become important areas of research. Herein, we review the molecular mechanisms of cardioprotection and cardioprotective modalities after ischemia-reperfusion injury (IRI) to identify potential approaches to reduce mortality and improve prognosis in patients with AMI.
Collapse
Affiliation(s)
- Honghong Zhang
- Department of Cardiology, Affiliated Hospital of Jiaxing University: First Hospital of Jiaxing, No. 1882 Zhonghuan South Road, Jiaxing, 314000, Zhejiang, People's Republic of China
| | - Huilin Hu
- Department of Cardiology, Affiliated Hospital of Jiaxing University: First Hospital of Jiaxing, No. 1882 Zhonghuan South Road, Jiaxing, 314000, Zhejiang, People's Republic of China.
| | - Changlin Zhai
- Department of Cardiology, Affiliated Hospital of Jiaxing University: First Hospital of Jiaxing, No. 1882 Zhonghuan South Road, Jiaxing, 314000, Zhejiang, People's Republic of China
| | - Lele Jing
- Department of Cardiology, Affiliated Hospital of Jiaxing University: First Hospital of Jiaxing, No. 1882 Zhonghuan South Road, Jiaxing, 314000, Zhejiang, People's Republic of China
| | - Hongen Tian
- Department of Cardiology, Affiliated Hospital of Jiaxing University: First Hospital of Jiaxing, No. 1882 Zhonghuan South Road, Jiaxing, 314000, Zhejiang, People's Republic of China
| |
Collapse
|
14
|
Rabinovich-Nikitin I, Kirshenbaum LA. Circadian regulated control of myocardial ischemia-reperfusion injury. Trends Cardiovasc Med 2024; 34:1-7. [PMID: 36150629 DOI: 10.1016/j.tcm.2022.09.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 09/15/2022] [Accepted: 09/16/2022] [Indexed: 01/14/2023]
Abstract
Circadian mechanisms have been associated with the pathogenesis of a variety of cardiovascular diseases, including myocardial ischemia-reperfusion injury (I-R). Myocardial ischemia resulting from impaired oxygen delivery to cardiac muscle sets into motion a cascade of cellular events that paradoxically triggers greater cardiac dysfunction upon reinstitution of coronary blood supply, a phenomenon known as I-R. I-R injury has been attributed to a number of cellular defects including increased reactive oxygen species (ROS), increased intracellular calcium and impaired mitochondrial bioenergetics that ultimately lead to cardiac cell death, ventricular remodeling and heart failure. Emerging evidence has identified a strong correlation between cellular defects that underlie I-R and the disrupted circadian. In fact, recent studies have shown that circadian dysfunction exacerbates cardiac injury following MI from impaired cellular quality control mechanisms such as autophagy, which are vital in the clearance of damaged cellular proteins and organelles such as mitochondria from the cell. The accumulation of cellular debris is posited as the central underlying cause of excessive cardiac cell death and ventricular dysfunction following MI. The complexities that govern the interplay between circadian biology and I-R injury following MI is at its infancy and understanding how circadian misalignment, such as in shift workers impacts I-R injury is of great scientific and clinical importance toward development of new therapeutic strategies using chronotherapy and circadian regulation to mitigate cardiac injury and improve cardiac outcomes after injury. In this review, we highlight recent advances in circadian biology and adaptive cellular quality control mechanisms that influence cardiac injury in response to MI injury with a specific focus on how circadian biology can be utilized to further cardiovascular medicine and patient care.
Collapse
Affiliation(s)
- Inna Rabinovich-Nikitin
- Department of Physiology and Pathophysiology, The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Canada
| | - Lorrie A Kirshenbaum
- Department of Physiology and Pathophysiology, The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Canada; Department of Pharmacology and Therapeutics Rady College of Medicine, Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba R2H 2H6, Canada.
| |
Collapse
|
15
|
Wei X, Lv Y, Yang C, Gao R, Zou S, Xu Y. Bufalin reduces myocardial infarction-induced myocardial fibrosis and improves cardiac function by inhibiting the NLRP3/IL-1β signalling pathway. Clin Exp Pharmacol Physiol 2023. [PMID: 37243403 DOI: 10.1111/1440-1681.13783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 03/23/2023] [Accepted: 05/01/2023] [Indexed: 05/28/2023]
Abstract
Early inflammatory responses post myocardial infarction (MI) is associated with increased myocardial fibrosis and cardiac remodelling. The NLRP3 inflammasome, a key factor in this response, regulates the expression of interleukins (IL)-1β and IL-18. Inhibiting the inflammatory process may be beneficial for post-MI recovery. Bufalin effectively inhibits inflammation and fibrosis. The aim of this study was to evaluate the effects of bufalin and MCC950, an NLRP3 inflammasome inhibitor, as potential treatment agents for MI using an experimental mouse model. Male C57BL/6 mice were subjected to left coronary artery ligation to induce MI and subsequently treated with bufalin (0.5 mg/kg), MCC950 (10 mg/kg) or saline thrice a week for 2 weeks. After 4 weeks, cardiac function and myocardial fibrosis were evaluated. Myocardial levels of fibrotic markers and inflammatory factors were analysed using western blotting, enzyme-linked immunosorbent assay, real-time quantitative polymerase chain reaction and immunofluorescence. In mice with MI, cardiac ultrasonography showed decreased cardiac function and myocardial fibrosis. Bufalin treatment restored left ventricular ejection fraction and fractional shortening and decreased the myocardial infarct size. Moreover, both bufalin and MCC950 preserved cardiac function and relieved myocardial fibrosis, with no significant difference. Hence, the present study findings suggest that bufalin can alleviate fibrosis and improve cardiac function in a mouse model by suppressing NLRP3/IL-1β signalling post-MI.
Collapse
Affiliation(s)
- Xiang Wei
- Fifth People's Hospital of Shanghai Fudan University, Shanghai, China
| | - Yang Lv
- Fifth People's Hospital of Shanghai Fudan University, Shanghai, China
| | - Chenxi Yang
- Fifth People's Hospital of Shanghai Fudan University, Shanghai, China
| | - Rifeng Gao
- Fifth People's Hospital of Shanghai Fudan University, Shanghai, China
| | - Su Zou
- Fifth People's Hospital of Shanghai Fudan University, Shanghai, China
| | - Yingjia Xu
- Fifth People's Hospital of Shanghai Fudan University, Shanghai, China
| |
Collapse
|
16
|
Leancă SA, Afrăsânie I, Crișu D, Matei IT, Duca ȘT, Costache AD, Onofrei V, Tudorancea I, Mitu O, Bădescu MC, Șerban LI, Costache II. Cardiac Reverse Remodeling in Ischemic Heart Disease with Novel Therapies for Heart Failure with Reduced Ejection Fraction. Life (Basel) 2023; 13:1000. [PMID: 37109529 PMCID: PMC10143569 DOI: 10.3390/life13041000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/03/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Despite the improvements in the treatment of coronary artery disease (CAD) and acute myocardial infarction (MI) over the past 20 years, ischemic heart disease (IHD) continues to be the most common cause of heart failure (HF). In clinical trials, over 70% of patients diagnosed with HF had IHD as the underlying cause. Furthermore, IHD predicts a worse outcome for patients with HF, leading to a substantial increase in late morbidity, mortality, and healthcare costs. In recent years, new pharmacological therapies have emerged for the treatment of HF, such as sodium-glucose cotransporter-2 inhibitors, angiotensin receptor-neprilysin inhibitors, selective cardiac myosin activators, and oral soluble guanylate cyclase stimulators, demonstrating clear or potential benefits in patients with HF with reduced ejection fraction. Interventional strategies such as cardiac resynchronization therapy, cardiac contractility modulation, or baroreflex activation therapy might provide additional therapeutic benefits by improving symptoms and promoting reverse remodeling. Furthermore, cardiac regenerative therapies such as stem cell transplantation could become a new therapeutic resource in the management of HF. By analyzing the existing data from the literature, this review aims to evaluate the impact of new HF therapies in patients with IHD in order to gain further insight into the best form of therapeutic management for this large proportion of HF patients.
Collapse
Affiliation(s)
- Sabina Andreea Leancă
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Irina Afrăsânie
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Daniela Crișu
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
| | - Iulian Theodor Matei
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Ștefania Teodora Duca
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Alexandru Dan Costache
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
- Department of Cardiovascular Rehabilitation, Clinical Rehabilitation Hospital, 700661 Iași, Romania
| | - Viviana Onofrei
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Ionuţ Tudorancea
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
- Department of Physiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Ovidiu Mitu
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Minerva Codruța Bădescu
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
- Internal Medicine Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
| | - Lăcrămioara Ionela Șerban
- Department of Physiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Irina Iuliana Costache
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| |
Collapse
|
17
|
Alsaidan AA, Al‐Kuraishy HM, Al‐Gareeb AI, Alexiou A, Papadakis M, Alsayed KA, Saad HM, Batiha GE. The potential role of SARS-CoV-2 infection in acute coronary syndrome and type 2 myocardial infarction (T2MI): Intertwining spread. Immun Inflamm Dis 2023; 11:e798. [PMID: 36988260 PMCID: PMC10022425 DOI: 10.1002/iid3.798] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 02/17/2023] [Accepted: 02/18/2023] [Indexed: 03/19/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a novel pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). It has been shown that SARS-CoV-2 infection-induced inflammatory and oxidative stress and associated endothelial dysfunction may lead to the development of acute coronary syndrome (ACS). Therefore, this review aimed to ascertain the link between severe SARS-CoV-2 infection and ACS. ACS is a spectrum of acute myocardial ischemia due to a sudden decrease in coronary blood flow, ranging from unstable angina to myocardial infarction (MI). Primary or type 1 MI (T1MI) is mainly caused by coronary plaque rupture and/or erosion with subsequent occlusive thrombosis. Secondary or type 2 MI (T2MI) is due to cardiac and systemic disorders without acute coronary atherothrombotic disruption. Acute SARS-CoV-2 infection is linked with the development of nonobstructive coronary disorders such as coronary vasospasm, dilated cardiomyopathy, myocardial fibrosis, and myocarditis. Furthermore, SARS-CoV-2 infection is associated with systemic inflammation that might affect coronary atherosclerotic plaque stability through augmentation of cardiac preload and afterload. Nevertheless, major coronary vessels with atherosclerotic plaques develop minor inflammation during COVID-19 since coronary arteries are not initially and primarily targeted by SARS-CoV-2 due to low expression of angiotensin-converting enzyme 2 in coronary vessels. In conclusion, SARS-CoV-2 infection through hypercytokinemia, direct cardiomyocyte injury, and dysregulation of the renin-angiotensin system may aggravate underlying ACS or cause new-onset T2MI. As well, arrhythmias induced by anti-COVID-19 medications could worsen underlying ACS.
Collapse
Affiliation(s)
- Aseel Awad Alsaidan
- Department of Family and Community Medicine, College of MedicineJouf UniversitySakakaSaudi Arabia
| | - Hayder M. Al‐Kuraishy
- Department of Clinical Pharmacology and Medicine, College of MedicineALmustansiriyia UniversityBaghdadIraq
| | - Ali I. Al‐Gareeb
- Department of Clinical Pharmacology and Medicine, College of MedicineALmustansiriyia UniversityBaghdadIraq
| | - Athanasios Alexiou
- Department of Science and EngineeringNovel Global Community Educational FoundationHebershamNew South WalesAustralia
- Department of Research and DevelopmentAFNP MedWienAustria
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten‐Herdecke, Heusnerstrasse 40University of Witten‐HerdeckeWuppertalGermany
| | - Khalid Adel Alsayed
- Department of Family and Community MedicineSecurity Forces Hospital ProgramRiyadhSaudi Arabia
| | - Hebatallah M. Saad
- Department of Pathology, Faculty of Veterinary MedicineMatrouh UniversityMatrouhEgypt
| | - Gaber El‐Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary MedicineDamanhour UniversityAlBeheiraEgypt
| |
Collapse
|
18
|
Mauro AG, Mezzaroma E, Toldo S, Melendez GC, Franco RL, Lesnefsky EJ, Abbate A, Hundley WG, Salloum FN. NLRP3-mediated inflammation in cardio-oncology: sterile yet harmful. Transl Res 2023; 252:9-20. [PMID: 35948198 PMCID: PMC9839540 DOI: 10.1016/j.trsl.2022.08.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/28/2022] [Accepted: 08/03/2022] [Indexed: 01/17/2023]
Abstract
Despite significant advances and the continuous development of novel, effective therapies to treat a variety of malignancies, cancer therapy-induced cardiotoxicity has been identified as a prominent cause of morbidity and mortality, closely competing with secondary malignancies. This unfortunate limitation has prompted the inception of the field of cardio-oncology with its purpose to provide the necessary knowledge and key information on mechanisms that support the use of the most efficacious cancer therapy with minimal or no interruption while paying close attention to preventing cardiovascular related morbidity and mortality. Several mechanisms that contribute to cancer therapy-induced cardiotoxicity have been proposed and studied. These mainly involve mitochondrial dysfunction and reactive oxygen species-induced oxidative stress, lysosomal damage, impaired autophagy, cell senescence, DNA damage, and sterile inflammation with the formation and activation of the NLRP3 inflammasome. In this review, we focus on describing the principal mechanisms for different classes of cancer therapies that lead to cardiotoxicity involving the NLRP3 inflammasome. We also summarize current evidence of cardio-protection with inflammasome inhibitors in the context of heart disease in general, and further highlight the potential application of this evidence for clinical translation in at risk patients for the purpose of preventing cancer therapy associated cardiovascular morbidity and mortality.
Collapse
Affiliation(s)
- Adolfo G Mauro
- Pauley Heart Center, Department of Internal Medicine, Cardiology, Virginia Commonwealth University, Richmond, VA
| | - Eleonora Mezzaroma
- Pauley Heart Center, Department of Internal Medicine, Cardiology, Virginia Commonwealth University, Richmond, VA
| | - Stefano Toldo
- Pauley Heart Center, Department of Internal Medicine, Cardiology, Virginia Commonwealth University, Richmond, VA
| | - Giselle C Melendez
- Department of Internal Medicine, Sections on Cardiovascular Medicine, Department of Pathology, Section on Comparative Medicine, Wake Forest, School of Medicine, Winston-Salem, NC
| | - R Lee Franco
- College of Humanities and Sciences, Department of Kinesiology and Health Sciences, Virginia Commonwealth University, Richmond, VA
| | - Edward J Lesnefsky
- Pauley Heart Center, Department of Internal Medicine, Cardiology, Virginia Commonwealth University, Richmond, VA; Department of the Medical Service of the McGuire Veterans Affairs Medical Center, Richmond, VA
| | - Antonio Abbate
- Pauley Heart Center, Department of Internal Medicine, Cardiology, Virginia Commonwealth University, Richmond, VA
| | - W Gregory Hundley
- Pauley Heart Center, Department of Internal Medicine, Cardiology, Virginia Commonwealth University, Richmond, VA
| | - Fadi N Salloum
- Pauley Heart Center, Department of Internal Medicine, Cardiology, Virginia Commonwealth University, Richmond, VA.
| |
Collapse
|
19
|
Yanpiset P, Maneechote C, Sriwichaiin S, Siri-Angkul N, Chattipakorn SC, Chattipakorn N. Gasdermin D-mediated pyroptosis in myocardial ischemia and reperfusion injury: Cumulative evidence for future cardioprotective strategies. Acta Pharm Sin B 2023; 13:29-53. [PMID: 36815034 PMCID: PMC9939317 DOI: 10.1016/j.apsb.2022.08.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 06/21/2022] [Accepted: 07/28/2022] [Indexed: 11/01/2022] Open
Abstract
Cardiomyocyte death is one of the major mechanisms contributing to the development of myocardial infarction (MI) and myocardial ischemia/reperfusion (MI/R) injury. Due to the limited regenerative ability of cardiomyocytes, understanding the mechanisms of cardiomyocyte death is necessary. Pyroptosis, one of the regulated programmed cell death pathways, has recently been shown to play important roles in MI and MI/R injury. Pyroptosis is activated by damage-associated molecular patterns (DAMPs) that are released from damaged myocardial cells and activate the formation of an apoptosis-associated speck-like protein containing a CARD (ASC) interacting with NACHT, LRR, and PYD domains-containing protein 3 (NLRP3), resulting in caspase-1 cleavage which promotes the activation of Gasdermin D (GSDMD). This pathway is known as the canonical pathway. GSDMD has also been shown to be activated in a non-canonical pathway during MI and MI/R injury via caspase-4/5/11. Suppression of GSDMD has been shown to provide cardioprotection against MI and MI/R injury. Although the effects of MI or MI/R injury on pyroptosis have previously been discussed, knowledge concerning the roles of GSDMD in these settings remains limited. In this review, the evidence from in vitro, in vivo, and clinical studies focusing on cardiac GSDMD activation during MI and MI/R injury is comprehensively summarized and discussed. Implications from this review will help pave the way for a new therapeutic target in ischemic heart disease.
Collapse
Affiliation(s)
- Panat Yanpiset
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Chayodom Maneechote
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sirawit Sriwichaiin
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Natthaphat Siri-Angkul
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Siriporn C. Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand,Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand,Corresponding author. Tel.: +66 53 935329; fax: +66 53 935368.
| |
Collapse
|
20
|
Peng Y, Zhou G, Guo M, Cheng Z, Luo S, Guo Y. Inhibition of Stimulator of Interferon Genes Protects Against Myocardial Ischemia-Reperfusion Injury in Diabetic Mice. CARDIOVASCULAR INNOVATIONS AND APPLICATIONS 2023; 8. [DOI: 10.15212/cvia.2023.0020] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2024] Open
Abstract
Background: Although the past decade has witnessed substantial scientific progress with the advent of cardioprotective pharmacological agents, most have failed to protect against myocardial ischemia/reperfusion (I/R) injury in diabetic hearts. This study was aimed at investigating the role of stimulator of interferon genes (STING) in I/R injury in diabetic mice and further exploring the underlying mechanisms.
Methods: Type 2 diabetic mice were subjected to I/R or sham operation to investigate the role of STING. STING knockout mice were subjected to 30 minutes of ischemia followed by reperfusion for 24 hours. Finally, myocardial injury, cardiac function, and inflammation levels were assessed.
Results: STING pathway activation was observed in diabetic I/R hearts, as evidenced by increased p-TBK and p-IRF3 expression. STING knockout significantly decreased the ischemic area and improved cardiac function after I/R in diabetic mice. STING knockout also elicited cardio-protective effects by decreasing serum cardiac troponin T and lactate dehydrogenase levels, thus diminishing the inflammatory response in the heart after I/R in diabetic mice. In vitro, STING inhibition decreased the expression of hypoxia-re-oxygenation-induced inflammatory cytokines.
Conclusions: Targeting STING inhibits inflammation and prevents I/R injury in diabetic mice. Thus, STING may be a potential novel therapeutic target against myocardial I/R injury in diabetes.
Collapse
|
21
|
Bhagat A, Shrestha P, Kleinerman ES. The Innate Immune System in Cardiovascular Diseases and Its Role in Doxorubicin-Induced Cardiotoxicity. Int J Mol Sci 2022; 23:ijms232314649. [PMID: 36498974 PMCID: PMC9739741 DOI: 10.3390/ijms232314649] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/15/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022] Open
Abstract
Innate immune cells are the early responders to infection and tissue damage. They play a critical role in the initiation and resolution of inflammation in response to insult as well as tissue repair. Following ischemic or non-ischemic cardiac injury, a strong inflammatory response plays a critical role in the removal of cell debris and tissue remodeling. However, persistent inflammation could be detrimental to the heart. Studies suggest that cardiac inflammation and tissue repair needs to be tightly regulated such that the timely resolution of the inflammation may prevent adverse cardiac damage. This involves the recognition of damage; activation and release of soluble mediators such as cytokines, chemokines, and proteases; and immune cells such as monocytes, macrophages, and neutrophils. This is important in the context of doxorubicin-induced cardiotoxicity as well. Doxorubicin (Dox) is an effective chemotherapy against multiple cancers but at the cost of cardiotoxicity. The innate immune system has emerged as a contributor to exacerbate the disease. In this review, we discuss the current understanding of the role of innate immunity in the pathogenesis of cardiovascular disease and dox-induced cardiotoxicity and provide potential therapeutic targets to alleviate the damage.
Collapse
|
22
|
Chiorescu RM, Mocan M, Inceu AI, Buda AP, Blendea D, Vlaicu SI. Vulnerable Atherosclerotic Plaque: Is There a Molecular Signature? Int J Mol Sci 2022; 23:13638. [PMID: 36362423 PMCID: PMC9656166 DOI: 10.3390/ijms232113638] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 10/30/2022] [Accepted: 10/31/2022] [Indexed: 08/18/2023] Open
Abstract
Atherosclerosis and its clinical manifestations, coronary and cerebral artery diseases, are the most common cause of death worldwide. The main pathophysiological mechanism for these complications is the rupture of vulnerable atherosclerotic plaques and subsequent thrombosis. Pathological studies of the vulnerable lesions showed that more frequently, plaques rich in lipids and with a high level of inflammation, responsible for mild or moderate stenosis, are more prone to rupture, leading to acute events. Identifying the vulnerable plaques helps to stratify patients at risk of developing acute vascular events. Traditional imaging methods based on plaque appearance and size are not reliable in prediction the risk of rupture. Intravascular imaging is a novel technique able to identify vulnerable lesions, but it is invasive and an operator-dependent technique. This review aims to summarize the current data from literature regarding the main biomarkers involved in the attempt to diagnose vulnerable atherosclerotic lesions. These biomarkers could be the base for risk stratification and development of the new therapeutic drugs in the treatment of patients with vulnerable atherosclerotic plaques.
Collapse
Affiliation(s)
- Roxana Mihaela Chiorescu
- Internal Medicine Department, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- Department of Internal Medicine, Emergency Clinical County Hospital, 400006 Cluj-Napoca, Romania
| | - Mihaela Mocan
- Internal Medicine Department, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- Department of Internal Medicine, Emergency Clinical County Hospital, 400006 Cluj-Napoca, Romania
| | - Andreea Ioana Inceu
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine, 400349 Cluj-Napoca, Romania
- Department of Cardiology, Nicolae Stăncioiu Heart Institute, 400001 Cluj-Napoca, Romania
| | - Andreea Paula Buda
- Department of Cardiology, Nicolae Stăncioiu Heart Institute, 400001 Cluj-Napoca, Romania
| | - Dan Blendea
- Department of Cardiology, Nicolae Stăncioiu Heart Institute, 400001 Cluj-Napoca, Romania
- Department of Cardiology, Iuliu Hațieganu University of Medicine and Pharmacy, 400437 Cluj-Napoca, Romania
| | - Sonia Irina Vlaicu
- Internal Medicine Department, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- Department of Internal Medicine, Emergency Clinical County Hospital, 400006 Cluj-Napoca, Romania
| |
Collapse
|
23
|
Potere N, Del Buono MG, Caricchio R, Cremer PC, Vecchié A, Porreca E, Dalla Gasperina D, Dentali F, Abbate A, Bonaventura A. Interleukin-1 and the NLRP3 inflammasome in COVID-19: Pathogenetic and therapeutic implications. EBioMedicine 2022; 85:104299. [PMID: 36209522 PMCID: PMC9536001 DOI: 10.1016/j.ebiom.2022.104299] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 09/01/2022] [Accepted: 09/16/2022] [Indexed: 11/11/2022] Open
Abstract
A hyperinflammatory response during severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection crucially worsens clinical evolution of coronavirus disease 2019 (COVID-19). The interaction between SARS-CoV-2 and angiotensin-converting enzyme 2 (ACE2) triggers the activation of the NACHT, leucine-rich repeat, and pyrin domain-containing protein 3 (NLRP3) inflammasome. Enhanced inflammasome activity has been associated with increased disease severity and poor prognosis. Evidence suggests that inflammasome activation and interleukin-1β (IL-1β) release aggravate pulmonary injury and induce hypercoagulability, favoring progression to respiratory failure and widespread thrombosis eventually leading to multiorgan failure and death. Observational studies with the IL-1 blockers anakinra and canakinumab provided promising results. In the SAVE-MORE trial, early treatment with anakinra significantly shortened hospital stay and improved survival in patients with moderate-to-severe COVID-19. In this review, we summarize current evidence supporting the pathogenetic role of the NLRP3 inflammasome and IL-1β in COVID-19, and discuss clinical trials testing IL-1 inhibition in COVID-19.
Collapse
Affiliation(s)
- Nicola Potere
- Department of Medicine and Ageing Sciences and Department of Innovative Technologies in Medicine and Dentistry, G. D'Annunzio University, Chieti, Italy
| | - Marco Giuseppe Del Buono
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University of the Sacred Heart, Rome, Italy
| | | | - Paul C. Cremer
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Alessandra Vecchié
- Medicina Generale 1, Medical Center, Ospedale di Circolo e Fondazione Macchi, Department of Internal Medicine, ASST Sette Laghi, Varese, Italy
| | - Ettore Porreca
- Department of Medicine and Ageing Sciences and Department of Innovative Technologies in Medicine and Dentistry, G. D'Annunzio University, Chieti, Italy
| | | | - Francesco Dentali
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Antonio Abbate
- Robert M. Berne Cardiovascular Research Center and Division of Cardiovascular Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Aldo Bonaventura
- Medicina Generale 1, Medical Center, Ospedale di Circolo e Fondazione Macchi, Department of Internal Medicine, ASST Sette Laghi, Varese, Italy,Corresponding author.
| |
Collapse
|
24
|
Alhajri N, Rustom M, Adegbile A, Ahmed W, Kilidar S, Afify N. Deciphering the Basis of Molecular Biology of Selected Cardiovascular Diseases: A View on Network Medicine. Int J Mol Sci 2022; 23:ijms231911421. [PMID: 36232723 PMCID: PMC9569471 DOI: 10.3390/ijms231911421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 09/23/2022] [Indexed: 11/16/2022] Open
Abstract
Cardiovascular diseases are the leading cause of death across the world. For decades, researchers have been studying the causes of cardiovascular disease, yet many of them remain undiscovered or poorly understood. Network medicine is a recently expanding, integrative field that attempts to elucidate this issue by conceiving of disease as the result of disruptive links between multiple interconnected biological components. Still in its nascent stages, this revolutionary application of network science facilitated a number of important discoveries in complex disease mechanisms. As methodologies become more advanced, network medicine harbors the potential to expound on the molecular and genetic complexities of disease to differentiate how these intricacies govern disease manifestations, prognosis, and therapy. This is of paramount importance for confronting the incredible challenges of current and future cardiovascular disease research. In this review, we summarize the principal molecular and genetic mechanisms of common cardiac pathophysiologies as well as discuss the existing knowledge on therapeutic strategies to prevent, halt, or reverse these pathologies.
Collapse
Affiliation(s)
- Noora Alhajri
- Department of Internal Medicine, Cleveland Clinic Abu Dhabi (CCAD), Abu Dhabi P.O. Box 112412, United Arab Emirates
- Correspondence:
| | - Mohammad Rustom
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
| | - Adedayo Adegbile
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
| | - Weshah Ahmed
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
| | - Salsabeel Kilidar
- Department of Emergency Medicine, Sheikh Shakhbout Medical City SSMC, Abu Dhabi P.O. Box 11001, United Arab Emirates
| | - Nariman Afify
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
| |
Collapse
|
25
|
Montelukast and Acute Coronary Syndrome: The Endowed Drug. Pharmaceuticals (Basel) 2022; 15:ph15091147. [PMID: 36145367 PMCID: PMC9500901 DOI: 10.3390/ph15091147] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/06/2022] [Accepted: 09/09/2022] [Indexed: 12/16/2022] Open
Abstract
Acute coronary syndrome (ACS) is a set of signs and symptoms caused by a reduction of coronary blood flow with subsequent myocardial ischemia. ACS is associated with activation of the leukotriene (LT) pathway with subsequent releases of various LTs, including LTB4, LTC4, and LTD4, which cause inflammatory changes and induction of immunothrombosis. LTs through cysteine leukotriene (CysLT) induce activation of platelets and clotting factors with succeeding coronary thrombosis. CysLT receptor (CysLTR) antagonists such as montelukast (MK) may reduce the risk of the development of ACS and associated complications through suppression of the activation of platelet and clotting factors. Thus, this critical review aimed to elucidate the possible protective role of MK in the management of ACS. The LT pathway is implicated in the pathogenesis of atherosclerosis, cardiac hypertrophy, and heart failure. Inhibition of the LT pathway and CysL1TR by MK might be effective in preventing cardiovascular complications. MK could be an effective novel therapy in the management of ACS through inhibition of pro-inflammatory CysLT1R and modulation of inflammatory signaling pathways. MK can attenuate thrombotic events by inhibiting platelet activation and clotting factors that are activated during the development of ACS. In conclusion, MK could be an effective agent in reducing the severity of ACS and associated complications. Experimental, preclinical, and clinical studies are recommended to confirm the potential therapeutic of MK in the management of ACS.
Collapse
|
26
|
Leancă SA, Crișu D, Petriș AO, Afrăsânie I, Genes A, Costache AD, Tesloianu DN, Costache II. Left Ventricular Remodeling after Myocardial Infarction: From Physiopathology to Treatment. Life (Basel) 2022; 12:1111. [PMID: 35892913 PMCID: PMC9332014 DOI: 10.3390/life12081111] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/19/2022] [Accepted: 07/21/2022] [Indexed: 12/11/2022] Open
Abstract
Myocardial infarction (MI) is the leading cause of death and morbidity worldwide, with an incidence relatively high in developed countries and rapidly growing in developing countries. The most common cause of MI is the rupture of an atherosclerotic plaque with subsequent thrombotic occlusion in the coronary circulation. This causes cardiomyocyte death and myocardial necrosis, with subsequent inflammation and fibrosis. Current therapies aim to restore coronary flow by thrombus dissolution with pharmaceutical treatment and/or intravascular stent implantation and to counteract neurohormonal activation. Despite these therapies, the injury caused by myocardial ischemia leads to left ventricular remodeling; this process involves changes in cardiac geometry, dimension and function and eventually progression to heart failure (HF). This review describes the pathophysiological mechanism that leads to cardiac remodeling and the therapeutic strategies with a role in slowing the progression of remodeling and improving cardiac structure and function.
Collapse
Affiliation(s)
- Sabina Andreea Leancă
- Department of Cardiology, Emergency Clinical Hospital “Sf. Spiridon”, Bd. Independentei nr. 1, 700111 Iasi, Romania; (S.A.L.); (A.O.P.); (I.A.); (A.G.); (D.N.T.); (I.I.C.)
| | - Daniela Crișu
- Department of Cardiology, Emergency Clinical Hospital “Sf. Spiridon”, Bd. Independentei nr. 1, 700111 Iasi, Romania; (S.A.L.); (A.O.P.); (I.A.); (A.G.); (D.N.T.); (I.I.C.)
| | - Antoniu Octavian Petriș
- Department of Cardiology, Emergency Clinical Hospital “Sf. Spiridon”, Bd. Independentei nr. 1, 700111 Iasi, Romania; (S.A.L.); (A.O.P.); (I.A.); (A.G.); (D.N.T.); (I.I.C.)
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Str. University nr. 16, 700083 Iasi, Romania;
| | - Irina Afrăsânie
- Department of Cardiology, Emergency Clinical Hospital “Sf. Spiridon”, Bd. Independentei nr. 1, 700111 Iasi, Romania; (S.A.L.); (A.O.P.); (I.A.); (A.G.); (D.N.T.); (I.I.C.)
| | - Antonia Genes
- Department of Cardiology, Emergency Clinical Hospital “Sf. Spiridon”, Bd. Independentei nr. 1, 700111 Iasi, Romania; (S.A.L.); (A.O.P.); (I.A.); (A.G.); (D.N.T.); (I.I.C.)
| | - Alexandru Dan Costache
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Str. University nr. 16, 700083 Iasi, Romania;
- Department of Cardiovascular Rehabilitation, Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Dan Nicolae Tesloianu
- Department of Cardiology, Emergency Clinical Hospital “Sf. Spiridon”, Bd. Independentei nr. 1, 700111 Iasi, Romania; (S.A.L.); (A.O.P.); (I.A.); (A.G.); (D.N.T.); (I.I.C.)
| | - Irina Iuliana Costache
- Department of Cardiology, Emergency Clinical Hospital “Sf. Spiridon”, Bd. Independentei nr. 1, 700111 Iasi, Romania; (S.A.L.); (A.O.P.); (I.A.); (A.G.); (D.N.T.); (I.I.C.)
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Str. University nr. 16, 700083 Iasi, Romania;
| |
Collapse
|
27
|
Cheng X, Zhang R, Wei S, Huang J, Zhai K, Li Y, Gao B. Dexamethasone Alleviates Myocardial Injury in a Rat Model of Acute Myocardial Infarction Supported by Venoarterial Extracorporeal Membrane Oxygenation. Front Public Health 2022; 10:900751. [PMID: 35928492 PMCID: PMC9343845 DOI: 10.3389/fpubh.2022.900751] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
Myocardial ischemia causes myocardial inflammation. Research indicates that the venoarterial extracorporeal membrane oxygenation (VA ECMO) provides cardiac support; however, the inflammatory response caused by myocardial ischemia remains unresolved. Dexamethasone (Dex), a broad anti-inflammatory agent, exhibits a cardioprotective effect. This study aims to investigate the effect of Dex on a rat model of acute myocardial infarction (AMI) supported by VA ECMO. Male Sprague-Dawley rats (300–350 g) were randomly divided into three groups: Sham group (n = 5), ECMO group (n = 6), and ECMO + Dex group (n = 6). AMI was induced by ligating the left anterior descending (LAD) coronary artery. Sham group only thoracotomy was performed but LAD was not ligated. The ECMO and ECMO + Dex groups were subjected to 1 h of AMI and 2 h of VA ECMO. In the ECMO + Dex group, Dex (0.2 mg/kg) was intravenously injected into the rats after 1 h of AMI. Lastly, myocardial tissue and blood samples were harvested for further evaluation. The ECMO + Dex group significantly reduced infarct size and levels of cTnI, cTnT, and CK-MB. Apoptotic cells and the expression levels of Bax, Caspase3, and Cle-Caspase3 proteins were markedly lower in the ECMO + Dex group than that in the ECMO group. Neutrophil and macrophage infiltration was lower in the ECMO + Dex group than in the ECMO group. A significant reduction was noted in ICAM-1, C5a, MMP-9, IL-1β, IL-6, and TNF-α. In summary, our findings revealed that Dex alleviates myocardial injury in a rat model of AMI supported by VA ECMO.
Collapse
Affiliation(s)
- Xingdong Cheng
- Department of Cardiac Surgery, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Rongzhi Zhang
- Department of Anesthesiology, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Shilin Wei
- Department of Cardiac Surgery, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Jian Huang
- Department of Cardiac Surgery, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Kerong Zhai
- Department of Cardiac Surgery, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Yongnan Li
- Department of Cardiac Surgery, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
- *Correspondence: Yongnan Li
| | - Bingren Gao
- Department of Cardiac Surgery, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
- Bingren Gao
| |
Collapse
|
28
|
Cheng W, Cui C, Liu G, Ye C, Shao F, Bagchi AK, Mehta JL, Wang X. NF-κB, A Potential Therapeutic Target in Cardiovascular Diseases. Cardiovasc Drugs Ther 2022; 37:571-584. [PMID: 35796905 DOI: 10.1007/s10557-022-07362-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/17/2022] [Indexed: 11/03/2022]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death globally. Atherosclerosis is the basis of major CVDs - myocardial ischemia, heart failure, and stroke. Among numerous functional molecules, the transcription factor nuclear factor κB (NF-κB) has been linked to downstream target genes involved in atherosclerosis. The activation of the NF-κB family and its downstream target genes in response to environmental and cellular stress, hypoxia, and ischemia initiate different pathological events such as innate and adaptive immunity, and cell survival, differentiation, and proliferation. Thus, NF-κB is a potential therapeutic target in the treatment of atherosclerosis and related CVDs. Several biologics and small molecules as well as peptide/proteins have been shown to regulate NF-κB dependent signaling pathways. In this review, we will focus on the function of NF-κB in CVDs and the role of NF-κB inhibitors in the treatment of CVDs.
Collapse
Affiliation(s)
- Weijia Cheng
- Department of Cardiology, The First Affiliated Hospital, Xinxiang Medical University, Weihui, China.,Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Can Cui
- Department of Cardiology, The First Affiliated Hospital, Xinxiang Medical University, Weihui, China.,Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Gang Liu
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Chenji Ye
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Fang Shao
- Department of Cardiology, Fuwai Central China Cardiovascular Hospital, Zhengzhou, 450046, China
| | - Ashim K Bagchi
- Division of Cardiology, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, 72205, USA
| | - Jawahar L Mehta
- Division of Cardiology, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, 72205, USA.
| | - Xianwei Wang
- Department of Cardiology, The First Affiliated Hospital, Xinxiang Medical University, Weihui, China. .,Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
29
|
Hua F, Li JY, Zhang M, Zhou P, Wang L, Ling TJ, Bao GH. Kaempferol-3-O-rutinoside exerts cardioprotective effects through NF-κB/NLRP3/Caspase-1 pathway in ventricular remodeling after acute myocardial infarction. J Food Biochem 2022; 46:e14305. [PMID: 35758877 DOI: 10.1111/jfbc.14305] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/21/2022] [Accepted: 05/30/2022] [Indexed: 11/29/2022]
Abstract
Ventricular remodeling (VR) after acute myocardial infarction (AMI) is the main pathogenesis of chronic heart failure (CHF). Kaempferol-3-O-rutinoside (KR) is the flavonoid glycoside with the highest content in Lu'an GuaPian tea, which has good pharmacological activities. However, the mechanism of KR against VR after AMI remains unclear. Molecular docking was used to predict the targets of KR on the NLRP3/Caspase-1 signaling pathway. Histological changes in the myocardium were visualized using HE staining, Masson staining. Cardiomyocyte apoptosis was detected using TUNEL. Immunohistochemistry was used to examine NLRP3, Caspase-1 p20, and GSDMD. IL-1β level in serum was detected using ELISA. Finally, the expressions of NF-κB p65, NLRP3, ASC, Caspase-1 p20, GSDMD, and IL-1β were measured using RT-PCR and Western blotting. Our results showed that KR had a good binding activity with NLRP3, Caspase-1, and GSDMD, significantly improved cardiac function, alleviated cardiac pathological changes, reduced the excessive deposition of myocardial interstitial collagen, and inhibited cardiomyocyte apoptosis in AMI rats. Furthermore, KR could decrease the IL-1β level and inhibit the expressions of NF-κB p65, NLRP3, ASC, Caspase-1 p20, GSDMD, and IL-1β. Our study suggests that KR can prevent and treat VR after AMI, and the protective effect is related to its regulatory NF-κB/NLRP3/Caspase-1 signaling pathway. PRACTICAL APPLICATIONS: Kaempferol-3-O-rutinoside is present in Carthamus tinctorius L., Nymphaea candida, Afgekia mahidoliae and green tea, which has good pharmacological activities against liver injury, cerebral ischemia/reperfusion injury, dementia, hyperglycemia, and myocardial infarction. Our previous study found that kaempferol-3-O-rutinoside had an obvious anti-inflammatory effect, and could significantly improve the cell survival rate of H9c2 myocardium inflammatory injury induced by LPS. In this study, kaempferol-3-O-rutinoside significantly improved cardiac function, alleviated cardiac pathological changes, reduced the excessive deposition of myocardial interstitial collagen, and inhibited cardiomyocyte apoptosis in AMI rats. Furthermore, kaempferol-3-O-rutinoside could decrease the IL-1β level and inhibit the expressions of NF-κB p65, NLRP3, ASC, Caspase-1, GSDMD and IL-1β, suggesting that kaempferol-3-O-rutinoside could regulate NF-κB/NLRP3/Caspase-1 signaling pathway.
Collapse
Affiliation(s)
- Fang Hua
- School of Pharmacy, Anhui Xinhua University, Hefei, Anhui, People's Republic of China.,State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, Hefei, Anhui, People's Republic of China
| | - Jing Ya Li
- Department of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, People's Republic of China
| | - Meng Zhang
- Department of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, People's Republic of China
| | - Peng Zhou
- Department of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, People's Republic of China
| | - Liang Wang
- Department of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, People's Republic of China
| | - Tie Jun Ling
- State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, Hefei, Anhui, People's Republic of China
| | - Guan Hu Bao
- State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, Hefei, Anhui, People's Republic of China
| |
Collapse
|
30
|
Inhibition of GSDMD Activates Poly(ADP-ribosyl)ation and Promotes Myocardial Ischemia-Reperfusion Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1115749. [PMID: 35783187 PMCID: PMC9249530 DOI: 10.1155/2022/1115749] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 04/03/2022] [Accepted: 04/15/2022] [Indexed: 12/21/2022]
Abstract
The precise control of cardiomyocyte viability is imperative to combat myocardial ischemia-reperfusion injury (I/R), in which apoptosis and pyroptosis putatively contribute to the process. Recent researches indicated that GSDMD is involved in I/R as an executive protein of pyroptosis. However, its effect on other forms of cell death is unclear. We identified that GSDMD and GSDMD-N levels were significantly upregulated in the I/R myocardium of mice. Knockout of GSDMD conferred the resistance of the hearts to reperfusion injury in the acute phase of I/R but aggravated reperfusion injury in the chronic phase of I/R. Mechanistically, GSDMD deficiency induced the activation of PARylation and the consumption of NAD+ and ATP, leading to cardiomyocyte apoptosis. Moreover, PJ34, a putative PARP-1 inhibitor, reduced the myocardial injury caused by GSDMD deficiency. Our results reveal a novel action modality of GSDMD in the regulation of cardiomyocyte death; inhibition of GSDMD activates PARylation, suggesting the multidirectional role of GSDMD in I/R and providing a new theory for clinical treatment.
Collapse
|
31
|
Lu Y, Xiang M, Xin L, Zhang Y, Wang Y, Shen Z, Li L, Cui X. Qiliqiangxin Modulates the Gut Microbiota and NLRP3 Inflammasome to Protect Against Ventricular Remodeling in Heart Failure. Front Pharmacol 2022; 13:905424. [PMID: 35721118 PMCID: PMC9201726 DOI: 10.3389/fphar.2022.905424] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 04/13/2022] [Indexed: 02/03/2023] Open
Abstract
Aims: Pathological left ventricular (LV) remodeling induced by multiple causes often triggers fatal cardiac dysfunction, heart failure (HF), and even cardiac death. This study is aimed to investigate whether qiliqiangxin (QL) could improve LV remodeling and protect against HF via modulating gut microbiota and inhibiting nod-like receptor pyrin domain 3 (NLRP3) inflammasome activation. Methods: Rats were respectively treated with QL (100 mg/kg/day) or valsartan (1.6 mg/kg/day) by oral gavage after transverse aortic constriction or sham surgery for 13 weeks. Cardiac functions and myocardial fibrosis were assessed. In addition, gut microbial composition was assessed by 16S rDNA sequencing. Furthermore, rats’ hearts were harvested for histopathological and molecular analyses including immunohistochemistry, immunofluorescence, terminal-deoxynucleotidyl transferase-mediated 2’-deoxyuridine 5’-triphosphated nick end labeling, and Western blot. Key findings: QL treatment preserved cardiac functions including LV ejection fractions and fractional shortening and markedly improved the LV remodeling. Moreover, HF was related to the gut microbial community reorganization like a reduction in Lactobacillus, while QL reversed it. Additionally, the protein expression levels like IL-1β, TNF-α, NF-κB, and NLRP3 were decreased in the QL treatment group compared to the model one. Conclusion: QL ameliorates ventricular remodeling to some extent in rats with HF by modulating the gut microbiota and NLRP3 inflammasome, which indicates the potential therapeutic effects of QL on those who suffer from HF.
Collapse
Affiliation(s)
- Yingdong Lu
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Mi Xiang
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Laiyun Xin
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.,First Clinical Medical School, Shandong University of Chinese Medicine, Jinan, China
| | - Yang Zhang
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.,First Clinical Medical School, Shandong University of Chinese Medicine, Jinan, China
| | - Yuling Wang
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zihuan Shen
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Li Li
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiangning Cui
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
32
|
Hu S, Gao Y, Gao R, Wang Y, Qu Y, Yang J, Wei X, Zhang F, Ge J. The selective STING inhibitor H-151 preserves myocardial function and ameliorates cardiac fibrosis in murine myocardial infarction. Int Immunopharmacol 2022; 107:108658. [DOI: 10.1016/j.intimp.2022.108658] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 02/15/2022] [Accepted: 02/23/2022] [Indexed: 12/12/2022]
|
33
|
Hagerty T, Kluge MA, LeWinter MM. Recurrent Pericarditis: a Stubborn Opponent Meets New Treatments in 2022. Curr Cardiol Rep 2022; 24:915-923. [PMID: 35612721 PMCID: PMC9130990 DOI: 10.1007/s11886-022-01719-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/11/2022] [Indexed: 11/29/2022]
Abstract
Purpose of Review Our goal in writing this review was to provide a comprehensive appraisal of current therapies for idiopathic recurrent pericarditis with a particular focus on the newest therapeutic agents. We sought to understand the role of the inflammasome in the pathophysiology of pericarditis and how it informs the use of interleukin-1 (IL-1)-directed therapies. Recent Findings The latest research on this topic has focused on the critical role of the NLRP3 (NACHT, leucine-rich repeat, and pyrin domain-containing protein) inflammasome. Very recently, components of the NLRP3 inflammasome were detected by immune staining in pericardial tissue from patients with recurrent idiopathic pericarditis. In a mouse model of pericarditis, anti-IL-1 agents anakinra and rilonacept reduced NLRP3 immunostaining. Subsequent study of these drugs in human subjects with idiopathic recurrent pericarditis demonstrated their efficacy. Summary Recurrent idiopathic pericarditis, while relatively rare, poses a continued treatment challenge and contributes to a diminished quality of life for those patients who are afflicted. Recent developments, including an animal model of the disease and the use of IL-1-directed therapies, represent an exciting leap forward in our understanding of treatment targets. These advances offer not only new tools in our fight against this disease, but also the promise of earlier intervention and attenuation of disease morbidity. As our experience with these new agents expands, we can address questions about the ideal timing of introduction of anti-IL-1 therapy and duration of therapy and better understand the potential side effect profile.
Supplementary Information The online version contains supplementary material available at 10.1007/s11886-022-01719-z.
Collapse
Affiliation(s)
- Tracy Hagerty
- Cardiology Unit, University of Vermont Medical Center, 111 Colchester Avenue, Burlington, VT 05401 USA
- Larner College of Medicine, University of Vermont, Burlington, VT USA
| | - Matthew A. Kluge
- Cardiology Unit, University of Vermont Medical Center, 111 Colchester Avenue, Burlington, VT 05401 USA
| | - Martin M. LeWinter
- Cardiology Unit, University of Vermont Medical Center, 111 Colchester Avenue, Burlington, VT 05401 USA
- Larner College of Medicine, University of Vermont, Burlington, VT USA
| |
Collapse
|
34
|
Puspitasari YM, Ministrini S, Schwarz L, Karch C, Liberale L, Camici GG. Modern Concepts in Cardiovascular Disease: Inflamm-Aging. Front Cell Dev Biol 2022; 10:882211. [PMID: 35663390 PMCID: PMC9158480 DOI: 10.3389/fcell.2022.882211] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 04/13/2022] [Indexed: 11/18/2022] Open
Abstract
The improvements in healthcare services and quality of life result in a longer life expectancy and a higher number of aged individuals, who are inevitably affected by age-associated cardiovascular (CV) diseases. This challenging demographic shift calls for a greater effort to unravel the molecular mechanisms underlying age-related CV diseases to identify new therapeutic targets to cope with the ongoing aging "pandemic". Essential for protection against external pathogens and intrinsic degenerative processes, the inflammatory response becomes dysregulated with aging, leading to a persistent state of low-grade inflammation known as inflamm-aging. Of interest, inflammation has been recently recognized as a key factor in the pathogenesis of CV diseases, suggesting inflamm-aging as a possible driver of age-related CV afflictions and a plausible therapeutic target in this context. This review discusses the molecular pathways underlying inflamm-aging and their involvement in CV disease. Moreover, the potential of several anti-inflammatory approaches in this context is also reviewed.
Collapse
Affiliation(s)
| | - Stefano Ministrini
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
- Internal Medicine, Angiology and Atherosclerosis, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Lena Schwarz
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Caroline Karch
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Luca Liberale
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa—Italian Cardiovascular Network, Genoa, Italy
| | - Giovanni G. Camici
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
- Department of Cardiology, University Heart Center, University Hospital Zurich, Zurich, Switzerland
- Department of Research and Education, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
35
|
Chai R, Xue W, Shi S, Zhou Y, Du Y, Li Y, Song Q, Wu H, Hu Y. Cardiac Remodeling in Heart Failure: Role of Pyroptosis and Its Therapeutic Implications. Front Cardiovasc Med 2022; 9:870924. [PMID: 35509275 PMCID: PMC9058112 DOI: 10.3389/fcvm.2022.870924] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 03/31/2022] [Indexed: 12/17/2022] Open
Abstract
Pyroptosis is a kind of programmed cell death closely related to inflammation. The pathways that mediate pyroptosis can be divided into the Caspase-1-dependent canonical pathway and the Caspase4/5/11-dependent non-canonical pathway. The most significant difference from other cell death is that pyroptosis rapidly causes rupture of the plasma membrane, cell expansion, dissolution and rupture of the cell membrane, the release of cell contents and a large number of inflammatory factors, and send pro-inflammatory signals to adjacent cells, recruit inflammatory cells and induce inflammatory responses. Cardiac remodeling is the basic mechanism of heart failure (HF) and the core of pathophysiological research on the underlying mechanism. A large number of studies have shown that pyroptosis can cause cardiac fibrosis, cardiac hypertrophy, cardiomyocytes death, myocardial dysfunction, excessive inflammation, and cardiac remodeling. Therefore, targeting pyroptosis has a good prospect in improving cardiac remodeling in HF. In this review, the basic molecular mechanism of pyroptosis is summarized, the relationship between pyroptosis and cardiac remodeling in HF is analyzed in-depth, and the potential therapy of targeting pyroptosis to improve adverse cardiac remodeling in HF is discussed, providing some ideas for improving the study of adverse cardiac remodeling in HF.
Collapse
Affiliation(s)
- Ruoning Chai
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Wenjing Xue
- Department of Clinical Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Shuqing Shi
- Department of Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yu Zhou
- Department of Clinical Medicine, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Yihang Du
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuan Li
- Department of Clinical Medicine, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Qingqiao Song
- Department of Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Huaqin Wu
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Huaqin Wu
| | - Yuanhui Hu
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Yuanhui Hu
| |
Collapse
|
36
|
Bajic Z, Sobot T, Skrbic R, Stojiljkovic MP, Ponorac N, Matavulj A, Djuric DM. Homocysteine, Vitamins B6 and Folic Acid in Experimental Models of Myocardial Infarction and Heart Failure—How Strong Is That Link? Biomolecules 2022; 12:biom12040536. [PMID: 35454125 PMCID: PMC9027107 DOI: 10.3390/biom12040536] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/24/2022] [Accepted: 03/28/2022] [Indexed: 12/29/2022] Open
Abstract
Cardiovascular diseases are the leading cause of death and the main cause of disability. In the last decade, homocysteine has been found to be a risk factor or a marker for cardiovascular diseases, including myocardial infarction (MI) and heart failure (HF). There are indications that vitamin B6 plays a significant role in the process of transsulfuration in homocysteine metabolism, specifically, in a part of the reaction in which homocysteine transfers a sulfhydryl group to serine to form α-ketobutyrate and cysteine. Therefore, an elevated homocysteine concentration (hyperhomocysteinemia) could be a consequence of vitamin B6 and/or folate deficiency. Hyperhomocysteinemia in turn could damage the endothelium and the blood vessel wall and induce worsening of atherosclerotic process, having a negative impact on the mechanisms underlying MI and HF, such as oxidative stress, inflammation, and altered function of gasotransmitters. Given the importance of the vitamin B6 in homocysteine metabolism, in this paper, we review its role in reducing oxidative stress and inflammation, influencing the functions of gasotransmitters, and improving vasodilatation and coronary flow in animal models of MI and HF.
Collapse
Affiliation(s)
- Zorislava Bajic
- Department of Physiology, Faculty of Medicine, University of Banja Luka, 78000 Banja Luka, Bosnia and Herzegovina; (Z.B.); (T.S.); (N.P.); (A.M.)
| | - Tanja Sobot
- Department of Physiology, Faculty of Medicine, University of Banja Luka, 78000 Banja Luka, Bosnia and Herzegovina; (Z.B.); (T.S.); (N.P.); (A.M.)
| | - Ranko Skrbic
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Banja Luka, 78000 Banja Luka, Bosnia and Herzegovina; (R.S.); (M.P.S.)
| | - Milos P. Stojiljkovic
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Banja Luka, 78000 Banja Luka, Bosnia and Herzegovina; (R.S.); (M.P.S.)
| | - Nenad Ponorac
- Department of Physiology, Faculty of Medicine, University of Banja Luka, 78000 Banja Luka, Bosnia and Herzegovina; (Z.B.); (T.S.); (N.P.); (A.M.)
| | - Amela Matavulj
- Department of Physiology, Faculty of Medicine, University of Banja Luka, 78000 Banja Luka, Bosnia and Herzegovina; (Z.B.); (T.S.); (N.P.); (A.M.)
| | - Dragan M. Djuric
- Faculty of Medicine, Institute of Medical Physiology “Richard Burian”, University of Belgrade, 11000 Belgrade, Serbia
- Correspondence:
| |
Collapse
|
37
|
Vecchié A, Del Buono MG, Mauro AG, Cremer PC, Imazio M, Klein AL, Abbate A, Dentali F, Bonaventura A. Advances in pharmacotherapy for acute and recurrent pericarditis. Expert Opin Pharmacother 2022; 23:681-691. [DOI: 10.1080/14656566.2022.2054327] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
| | - Marco Giuseppe Del Buono
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University of the Sacred Heart, Rome, Italy
| | | | - Paul C. Cremer
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Massimo Imazio
- Cardiology, Cardiothoracic Department, University Hospital “Santa Maria della Misericordia,” ASUFC, Udine, Italy
| | - Allan L. Klein
- Center for the Diagnosis and Treatment of Pericardial Diseases, Section of Cardiovascular Imaging, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Antonio Abbate
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Francesco Dentali
- Department of Medicine and Surgery, Insubria University, Varese, Italy
| | - Aldo Bonaventura
- Medicina Generale 1, Medical Center, Ospedale di Circolo e Fondazione Macchi, ASST Sette Laghi, Varese, Italy
| |
Collapse
|
38
|
Autoantibodies Present in Hidradenitis Suppurativa Correlate with Disease Severity and Promote the Release of Proinflammatory Cytokines in Macrophages. J Invest Dermatol 2022; 142:924-935. [PMID: 34606886 PMCID: PMC8860851 DOI: 10.1016/j.jid.2021.07.187] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/09/2021] [Accepted: 07/19/2021] [Indexed: 12/11/2022]
Abstract
Hidradenitis suppurativa (HS), also known as acne inversa, is a debilitating inflammatory skin disorder that is characterized by nodules that lead to the development of connected tunnels and scars as it progresses from Hurley stages I to III. HS has been associated with several autoimmune diseases, including inflammatory bowel disease and spondyloarthritis. We previously reported dysregulation of humoral immune responses in HS, characterized by elevated serum total IgG, B-cell activation, and antibodies recognizing citrullinated proteins. In this study, we characterized IgG autoreactivity in HS sera and lesional skin compared with those in normal healthy controls using an array-based high-throughput autoantibody screening. The Cy3-labeled anti-human assay showed the presence of autoantibodies against nuclear antigens, cytokines, cytoplasmic proteins, extracellular matrix proteins, neutrophil proteins, and citrullinated antigens. Most of these autoantibodies were significantly elevated in stages II‒III in HS sera and stage III in HS skin lesions compared with those of healthy controls. Furthermore, immune complexes containing both native and citrullinated versions of antigens can activate M1 and M2 macrophages to release proinflammatory cytokines such as TNF-α, IL-8, IL-6, and IL-12. Taken together, the identification of specific IgG autoantibodies that recognize circulating and tissue antigens in HS suggests an autoimmune mechanism and uncovers putative therapeutic targets.
Collapse
|
39
|
Bonaventura A, Vecchié A, Dagna L, Tangianu F, Abbate A, Dentali F. Colchicine for COVID-19: targeting NLRP3 inflammasome to blunt hyperinflammation. Inflamm Res 2022; 71:293-307. [PMID: 35113170 PMCID: PMC8811745 DOI: 10.1007/s00011-022-01540-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 11/14/2021] [Accepted: 01/13/2022] [Indexed: 01/08/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is capable of inducing the activation of NACHT, leucine-rich repeat, and pyrin domain-containing protein 3 (NLRP3) inflammasome, a macromolecular structure sensing the danger and amplifying the inflammatory response. The main product processed by NLRP3 inflammasome is interleukin (IL)-1β, responsible for the downstream production of IL-6, which has been recognized as an important mediator in coronavirus disease 2019 (COVID-19). Since colchicine is an anti-inflammatory drug with the ability to block NLRP3 inflammasome oligomerization, this may prevent the release of active IL-1β and block the detrimental effects of downstream cytokines, i.e. IL-6. To date, few randomized clinical trials and many observational studies with colchicine have been conducted, showing interesting signals. As colchicine is a nonspecific inhibitor of the NLRP3 inflammasome, compounds specifically blocking this molecule might provide increased advantages in reducing the inflammatory burden and its related clinical manifestations. This may occur through a selective blockade of different steps preceding NLRP3 inflammasome oligomerization as well as through a reduced release of the main cytokines (IL-1β and IL-18). Since most evidence is based on observational studies, definitive conclusion cannot be drawn and additional studies are needed to confirm preliminary results and further dissect how colchicine and other NLRP3 inhibitors reduce the inflammatory burden and evaluate the timing and duration of treatment.
Collapse
Affiliation(s)
- Aldo Bonaventura
- Medicina Generale 1, Medical Center, Ospedale di Circolo e Fondazione Macchi, ASST Sette Laghi, Varese, Italy.
| | - Alessandra Vecchié
- Medicina Generale 1, Medical Center, Ospedale di Circolo e Fondazione Macchi, ASST Sette Laghi, Varese, Italy
| | - Lorenzo Dagna
- Università Vita-Salute San Raffaele, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS Ospedale San Raffaele, Milan, Italy
| | - Flavio Tangianu
- Medicina Generale 1, Medical Center, Ospedale di Circolo e Fondazione Macchi, ASST Sette Laghi, Varese, Italy
| | - Antonio Abbate
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Francesco Dentali
- Department of Medicine and Surgery, Insubria University, Varese, Italy
| |
Collapse
|
40
|
Wang S, Zhang J, Wang Y, Jiang X, Guo M, Yang Z. NLRP3 inflammasome as a novel therapeutic target for heart failure. Anatol J Cardiol 2022; 26:15-22. [PMID: 35191381 PMCID: PMC8878950 DOI: 10.5152/anatoljcardiol.2021.580] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2021] [Indexed: 06/30/2024] Open
Abstract
Heart failure (HF) is a leading cause of mortality worldwide. The pathogenesis of HF is complex and has not yet been fully elucidated, which has slowed drug development and long-term treatments. Inflammasome-mediated responses occur during the progression of HF. It has been reported that energy metabolism and metabolites of intestinal flora are also involved in the process of HF, and they interact with each other to promote the progression of HF. NLR family pyrin domain containing 3 (NLRP3) inflammasome may be a key target in the relationship between inflammation-mediated energy metabolism and metabolites of intestinal flora. Elucidating the relationship among the above three factors may help to identify new molecular targets for the prevention and treatment of HF and ultimately affect the course of HF. In this study, we systematically summarize evidence regarding the relationship among NLRP3 inflammasome, energy metabolism, intestinal microflora metabolites, and inflammation, as well as highlight advantages of NLRP3 inflammasome in treating HF.
Collapse
Affiliation(s)
- Shuangcui Wang
- Department of Integrative Medicine, Tianjin University of Traditional Chinese Medicine; Tianjin-China
| | - Jiaqi Zhang
- Department of Integrative Medicine, Tianjin University of Traditional Chinese Medicine; Tianjin-China
| | - Yuli Wang
- Department of Integrative Medicine, Tianjin University of Traditional Chinese Medicine; Tianjin-China
| | - Xijuan Jiang
- Department of Integrative Medicine, Tianjin University of Traditional Chinese Medicine; Tianjin-China
| | - Maojuan Guo
- Department of Integrative Medicine, Tianjin University of Traditional Chinese Medicine; Tianjin-China
| | - Zhen Yang
- Department of Chinese Medicine, Tianjin University of Traditional Chinese Medicine; Tianjin-China
| |
Collapse
|
41
|
Mao S, Chen P, Pan W, Gao L, Zhang M. Exacerbated post-infarct pathological myocardial remodelling in diabetes is associated with impaired autophagy and aggravated NLRP3 inflammasome activation. ESC Heart Fail 2021; 9:303-317. [PMID: 34964299 PMCID: PMC8787965 DOI: 10.1002/ehf2.13754] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 10/28/2021] [Accepted: 11/24/2021] [Indexed: 01/14/2023] Open
Abstract
Background Diabetes mellitus (DM) patients surviving myocardial infarction (MI) have substantially higher mortality due to the more frequent development of subsequent pathological myocardial remodelling and concomitant functional deterioration. This study investigates the molecular pathways underlying accelerated cardiac remodelling in a well‐established mouse model of diabetes exposed to MI. Methods and results Myocardial infarction in DM mice was established by ligating the left anterior descending coronary artery. Cardiac function was assessed by echocardiography. Myocardial hypertrophy and cardiac fibrosis were determined histologically 6 weeks post‐MI or sham operation. Autophagy, the NLRP3 inflammasome, and caspase‐1 were evaluated by western blotting or immunofluorescence. Echocardiographic imaging revealed significantly increased left ventricular dilation in parallel with increased mortality after MI in DM mice (53.33%) compared with control mice (26.67%, P < 0.05). Immunoblotting, electron microscopy, and immunofluorescence staining for LC3 and p62 indicated impaired autophagy in DM + MI mice compared with control mice (P < 0.05). Furthermore, defective autophagy was associated with increased NLRP3 inflammasome and caspase‐1 hyperactivation in DM + MI mouse cardiomyocytes (P < 0.05). Consistent with NLRP3 inflammasome and caspase‐I hyperactivation, cardiomyocyte death and IL‐1β and IL‐18 secretion were increased in DM + MI mice (P < 0.05). Importantly, the autophagy inducer and the NLRP3 inhibitor attenuated the cardiac remodelling of DM mice after MI. Conclusion In summary, our results indicate that DM aggravates cardiac remodelling after MI through defective autophagy and associated exaggerated NLRP3 inflammasome activation, proinflammatory cytokine secretion, suggesting that restoring autophagy and inhibiting NLRP3 inflammasome activation may serve as novel targets for the prevention and treatment of post‐infarct remodelling in DM.
Collapse
Affiliation(s)
- Shuai Mao
- Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Critical Care Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510120, China.,Guangdong Provincial Branch of National Clinical Research Centre for Chinese Medicine Cardiology, Guangzhou, China
| | - Peipei Chen
- Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Critical Care Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510120, China
| | - Wenjun Pan
- Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Critical Care Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510120, China
| | - Lei Gao
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Minzhou Zhang
- Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Critical Care Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510120, China.,Guangdong Provincial Branch of National Clinical Research Centre for Chinese Medicine Cardiology, Guangzhou, China
| |
Collapse
|
42
|
Melatonin Alleviates LPS-Induced Pyroptotic Cell Death in Human Stem Cell-Derived Cardiomyocytes by Activating Autophagy. Stem Cells Int 2021; 2021:8120403. [PMID: 34873405 PMCID: PMC8643260 DOI: 10.1155/2021/8120403] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 10/22/2021] [Accepted: 10/29/2021] [Indexed: 11/18/2022] Open
Abstract
Endotoxemia in sepsis remains a problem due to a lack of effective strategies. Our previous studies have demonstrated that melatonin (Mel) protects against ischemic heart injury and arteriosclerosis. However, its role in endotoxemia-exposed cardiomyocytes remains poorly understood. This study explored, for the first time, the protective effect of Mel on the pyroptosis of human stem cell-derived cardiomyocytes (hiPSC-CMs) exposed to lipopolysaccharide (LPS). Our results showed that treatment with 1 μM or 10 μM Mel for 12 h significantly improved 1 μg/ml LPS-induced hiPSC-CM injuries, as reflected by drastically decreased LDH release and increased cell viability, which was accompanied by the overt induction of autophagy. Specifically, Mel profoundly alleviated LPS-induced cell pyroptosis, as evidenced by decreased propidium iodide (PI) and active caspase-1 double-positive cell rates; suppressed the expression of NLRP3, cleaved caspase-1 (activated form of caspase-1), and GSDMD-NT (functional N-terminal fragment of GSDMD) expression; and inhibited the production of the cleaved IL-1β and cleaved IL-18 cytokines. Additionally, double-membrane autophagosomes were observed in LPS-injured hiPSC-CMs treated with 1 μM or 10 μM Mel. The hiPSC-CMs treated with LPS exhibited considerably fewer acidic vesicles (as revealed by LAMP1 staining) and autophagosomes (as revealed by LC3-II staining); however, Mel reversed this outcome in a dose-dependent manner. Furthermore, coincubation with rapamycin (an autophagy activator) or 3-MA (an autophagy inhibitor) accentuated and attenuated the antipyroptotic actions of Mel, respectively. Collectively, our findings demonstrate that Mel shields hiPSC-CMs against pyroptosis during endotoxemia by activating autophagy.
Collapse
|
43
|
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: 5.7] [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
|
44
|
Wei D, Li R, Si T, He H, Wu W. Screening and bioinformatics analysis of key biomarkers in acute myocardial infarction. Pteridines 2021. [DOI: 10.1515/pteridines-2020-0031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Abstract
Acute myocardial infarction (AMI) is the most severe manifestation of coronary artery disease. Considerable efforts have been made to elucidate its etiology and pathology, but the genetic factors that play a decisive role in the occurrence of AMI are still unclear. To determine the molecular mechanism of the occurrence and development of AMI, four microarray datasets, namely, GSE29111, GSE48060, GSE66360, and GSE97320, were downloaded from the Gene Expression Omnibus (GEO) database. We analyzed the four GEO datasets to obtain the differential expression genes (DEGs) of patients with AMI and patients with non-AMI and then performed gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis, and Protein-protein interaction (PPI) network analysis. A total of 41 DEGs were identified, including 39 upregulated genes and 2 downregulated genes. The enriched functions and pathways of the DEGs included the inflammatory response, neutrophil chemotaxis, immune response, extracellular space, positive regulation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) transcription factor activity, response to lipopolysaccharide, receptor for advanced glycation end products (RAGE) receptor binding, innate immune response, defense response to bacterium, and receptor activity. The cytoHubba plug-in in Cytoscape was used to select the most significant hub gene from the PPI network. Ten hub genes were identified, and GO enrichment analysis revealed that these genes were mainly enriched in inflammatory response, neutrophil chemotaxis, immune response, RAGE receptor binding, and extracellular region. In conclusion, this study integrated four datasets and used bioinformatics methods to analyze the gene chips of AMI samples and control samples and identified DEGs that may be involved in the occurrence and development of AMI. The study provides reliable molecular biomarkers for AMI screening, diagnosis, and prognosis.
Collapse
Affiliation(s)
- Dongmei Wei
- Department of Cardiovasology, Liuzhou Traditional Chinese Medical Hospital , Liuzhou , Guangxi Province, 545001 , People’s Republic of China
| | - Rui Li
- Guangzhou University of Chinese Medicine , Guangzhou , Guangdong Province, 510405 , People’s Republic of China
| | - Tao Si
- Guangzhou University of Chinese Medicine , Guangzhou , Guangdong Province, 510405 , People’s Republic of China
| | - Hankang He
- Department of Cardiovasology, Liuzhou Traditional Chinese Medical Hospital , Liuzhou , Guangxi Province, 545001 , People’s Republic of China
| | - Wei Wu
- Guangzhou University of Chinese Medicine , Guangzhou , Guangdong Province, 510405 , People’s Republic of China
| |
Collapse
|
45
|
Luo Y, Zheng D, Mou T, Pu J, Huang Z, Chen W, Zhang Y, Wu Z. CMPK2 accelerates liver ischemia/reperfusion injury via the NLRP3 signaling pathway. Exp Ther Med 2021; 22:1358. [PMID: 34659504 PMCID: PMC8515557 DOI: 10.3892/etm.2021.10793] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 03/15/2021] [Indexed: 12/12/2022] Open
Abstract
Cytidine monophosphate kinase 2 (CMPK2) is a mitochondrial nucleotide monophosphate kinase which is important for the substrates of mitochondrial DNA synthesis and has been reported to participate in macrophage activation and the inflammatory response. The purpose of the present research was to determine the potential role of CMPK2 in hepatic ischemia/reperfusion (I/R) injury and to elucidate the underlying molecular mechanisms. The present study investigated the role of CMPK2 in regulating the NLRP3 pathway and liver dysfunction induced by hepatic I/R both in vivo and in vitro. It was revealed that hypoxia/reoxygenation (H/R) treatment enhanced the mRNA expression levels of CMPK2, NLRP3, IL-18, IL-1β and TNF-α in RAW 264.7 cells. The protein expression levels of IL-18, IL-1β and cleaved-caspase-1 were decreased following CMPK2 knockdown. Furthermore, the inhibition of AIM2 downregulated the expression level of IL-1β, IL-18 and cleaved-caspase-1 in the CMPK2 knockdown group followed by H/R treatment, while the inhibition of NLRP3 did not. CMPK2 deficiency also decreased alanine aminotransferase and aspartate aminotransferase expression in mice serum, as well as the pathological changes in the liver. Similarly, the release of IL-18 and IL-1β in mouse serum was also restrained with the decline of CMPK2. In conclusion, the results of the present study demonstrate that CMPK2 is indispensable for NLRP3 inflammasome activation, making CMPK2 an effective target to relieve the liver from I/R injury. In addition, the function of CMPK2 is closely associated with NLRP3 inflammasome activation, instead of AIM2.
Collapse
Affiliation(s)
- Yunhai Luo
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Daofeng Zheng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Tong Mou
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Junliang Pu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Zuotian Huang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Wei Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Yuke Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Zhongjun Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
46
|
Abstract
Purpose of Review Pericarditis is a generally benign disease, although complications and/or recurrences may occur in up to 30% of cases. New evidence on the pathophysiology of the disease has accumulated in recent years. Recent Findings Recently, it has been shown that the activation of the NLRP3 (NACHT, leucine-rich repeat, and pyrin domain-containing protein 3) inflammasome is central in the pathophysiology of pericarditis. These findings derive from clinical data, an experimental animal model of acute pericarditis supporting a role for the NLRP3 inflammasome in pericarditis, and from indirect evidence of inhibitors of NLRP3 inflammasome in clinical trials. Summary Pericarditis is regarded as a stereotypical response to an acute damage of the mesothelial cells of the pericardial layers. NLRP3 inflammasome, a macromolecular structure sensing damage and releasing pro-inflammatory cytokines, is centrally involved as it releases interleukin (IL)-1β, whose auto-induction feeds an autoinflammatory disease, mostly responsible for recurrences. Colchicine, an inhibitor of NLRP3 inflammasome formation, and IL-1-targeted therapies, such as anakinra and rilonacept, were found to effectively blunt the acute inflammation and reduce the risk for recurrences.
Collapse
|
47
|
Melatonin Exerts Cardioprotective Effects by Inhibiting NLRP3 Inflammasome-Induced Pyroptosis in Mice following Myocardial Infarction. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5387799. [PMID: 34512865 PMCID: PMC8429019 DOI: 10.1155/2021/5387799] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 07/12/2021] [Accepted: 08/16/2021] [Indexed: 12/26/2022]
Abstract
Myocardial infarction- (MI-) induced myocardial damage is mainly attributed to the loss of cardiomyocytes. Pyroptosis is a newly recognized form of programmed cell necrosis that is associated with the progression of MI. Melatonin has been shown to exert cardioprotective effects against cardiac damage in multiple cardiovascular diseases. However, the effect of melatonin on pyroptosis-induced cardiac injury in MI has not been elucidated. Herein, we found that melatonin administration ameliorated cardiac dysfunction and reduced cardiomyocyte death both in mice following coronary artery ligation and in H9C2 cells exposed to hypoxia. The results also showed that pyroptosis was induced both in vivo and in vitro, as evidenced by increased NLRP3, cleaved caspase-1, GSDMD-N, and mature IL-1β and IL-18 levels, and these changes were decreased by melatonin treatment. Furthermore, we observed that TLR4 and NF-κB levels were increased by MI or hypoxia, and these increases were reversed by melatonin. The antipyroptotic action of melatonin was abrogated by treatment with an agonist of the TLR4/NF-κB signaling pathway. Our results indicate that melatonin can exert cardioprotective effects by inhibiting NLRP3 inflammasome-induced pyroptosis through modulation of the TLR4/NF-κB signaling pathway and provide strong evidence for the utility of melatonin in the treatment of MI.
Collapse
|
48
|
Xu H, Yu W, Sun S, Li C, Ren J, Zhang Y. TAX1BP1 protects against myocardial infarction-associated cardiac anomalies through inhibition of inflammasomes in a RNF34/MAVS/NLRP3-dependent manner. Sci Bull (Beijing) 2021; 66:1669-1683. [PMID: 36654301 DOI: 10.1016/j.scib.2021.01.030] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/03/2020] [Accepted: 01/18/2021] [Indexed: 02/03/2023]
Abstract
Acute myocardial infarction (MI), one of the most common cardiovascular emergencies, is a leading cause of morbidity and mortality. Ample evidence has revealed an essential role for inflammasome activation and autophagy in the pathogenesis of acute MI. Tax1-binding protein 1 (TAX1BP1), an adaptor molecule involved in termination of proinflammatory signaling, serves as an important selective autophagy adaptor, but its role in cardiac ischemia remains elusive. This study examined the role of TAX1BP1 in myocardial ischemic stress and the underlying mechanisms involved. Levels of TAX1BP1 were significantly downregulated in heart tissues of patients with ischemic heart disease and in a left anterior descending (LAD) ligation-induced model of acute MI. Adenovirus carrying TAX1BP1 was delivered into the myocardium. The acute MI induced procedure elicited an infarct and cardiac dysfunction, the effect of which was mitigated by TAX1BP1 overexpression with little effect from viral vector alone. TAX1BP1 nullified acute MI-induced activation of the NLRP3 inflammasome and associated mitochondrial dysfunction. TAX1BP1 overexpression suppressed NLRP3 mitochondrial localization by inhibiting the interaction of NLRP3 with mitochondrial antiviral signaling protein (MAVS). Further investigation revealed that ring finger protein 34 (RNF34) was recruited to interact with TAX1BP1 thereby facilitating autophagic degradation of MAVS through K27-linked polyubiquitination of MAVS. Knockdown of RNF34 using siRNA nullified TAX1BP1 yielded protection against hypoxia-induced MAVS mitochondrial accumulation, NLRP3 inflammasome activation and associated loss of mitochondrial membrane potential. Taken together, our results favor a cardioprotective role for TAX1BP1 in acute MI through repression of inflammasome activation in a RNF34/MAVS-dependent manner.
Collapse
Affiliation(s)
- Haixia Xu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; Department of Cardiology, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Wenjun Yu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Shiqun Sun
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Congye Li
- Department of Cardiology, Xijing Hospital, Air Force Military Medical University, Xi'an 710038, China
| | - Jun Ren
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; Department of Pathology, University of Washington, Seattle WA 98195, USA.
| | - Yingmei Zhang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China.
| |
Collapse
|
49
|
Keskin H, Keskin F, Tavaci T, Halici H, Yuksel TN, Ozkaraca M, Bilen A, Halici Z. Neuroprotective effect of roflumilast under cerebral ischaemia/reperfusion injury in juvenile rats through NLRP-mediated inflammatory response inhibition. Clin Exp Pharmacol Physiol 2021; 48:1103-1110. [PMID: 33686709 DOI: 10.1111/1440-1681.13493] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 03/03/2021] [Accepted: 03/05/2021] [Indexed: 12/24/2022]
Abstract
This study aims to investigate the protective effect of roflumilast, a phosphodiesterase (PDE)-4 enzyme inhibitor, and demonstrate its possible role in the development prevention of cerebral ischemia/reperfusion injury (CI/RI) after stroke induced by carotid artery ligation in juvenile rats. The rats were randomly divided into five groups: healthy group without any treatment, healthy group administered with 1 mg/kg roflumilast, CI group not administered with roflumilast, CI group administered with 0.5 mg/kg roflumilast, and CI group administered with 1 mg/kg roflumilast. In the CI groups, reperfusion was achieved 2h after ischemia induction; in the roflumilast groups, this drug was intraperitoneally administered immediately after reperfusion and at the 12th hour. At the end of 24h, the rats were sacrificed and their brain tissues removed for examination. The mRNA expressions obtained with real-time PCR of IL-1β, TNF-α, and NLRP3 significantly increased in the CI/RI-induced groups compared with the control group, and this increase was significantly lower in the groups administered with roflumilast compared with the CI/RI-induced groups. Moreover, ELISA revealed that both IL-1 β and IL-6 brain levels were significantly higher in the CI/RI-induced groups than in the controls. This increase was significantly lower in the groups administered with roflumilast compared with the CI/RI-induced groups. Histopathological studies revealed that the values closest to those of the healthy group were obtained from the roflumilast groups. Nissl staining revealed that the Nissl bodies manifested normal density in the healthy and roflumilast-administered healthy groups, but were rare in the CI/RI-induced groups. Roflumilast treatment increased these decreased Nissl bodies with increasing doses. Observations indicated that the Nissl body density was close to the value in the healthy group in the CI/RI-induced group administered with 1 mg/kg roflumilast. Overall, roflumilast reduced cellular damage caused by CI/RI in juvenile rats, and this effect may be mediated by NLRP3.
Collapse
Affiliation(s)
- Halil Keskin
- Division of Paediatric Intensive Care Unit, Department of Paediatrics, Ataturk University Faculty of Medicine, Erzurum, Turkey
| | - Filiz Keskin
- Division of Paediatric Neurology, Department of Paediatrics, Ataturk University Faculty of Medicine, Erzurum, Turkey
| | - Taha Tavaci
- Department of Pharmacology, Ataturk University Faculty of Medicine, Erzurum, Turkey
| | - Hamza Halici
- Department of Pharmacology, Ataturk University Faculty of Medicine, Erzurum, Turkey
| | - Tugba Nurcan Yuksel
- Department of Pharmacology, Namik Kemal University Faculty of Medicine, Tekirdag, Turkey
| | - Mustafa Ozkaraca
- Department of Pathology, Cumhuriyet University Faculty of Veterinary, Sivas, Turkey
| | - Arzu Bilen
- Division of Endocrinology, Department of Internal Medicine, Ataturk University Faculty of Medicine, Erzurum, Turkey
| | - Zekai Halici
- Department of Pharmacology, Ataturk University Faculty of Medicine, Erzurum, Turkey
- Clinical Research, Development and Design Application and Research Center, Ataturk University, Erzurum, Turkey
| |
Collapse
|
50
|
Cole J, Htun N, Lew R, Freilich M, Quinn S, Layland J. Colchicine to Prevent Periprocedural Myocardial Injury in Percutaneous Coronary Intervention: The COPE-PCI Pilot Trial. Circ Cardiovasc Interv 2021; 14:e009992. [PMID: 34003667 DOI: 10.1161/circinterventions.120.009992] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Justin Cole
- Peninsula Heart Service, Peninsula Health, Frankston, Australia (J.C., N.H., R.L., M.F., J.L.).,Peninsula Clinical School, Monash University, Frankston, Australia (J.C., N.H., J.L.)
| | - Nay Htun
- Peninsula Heart Service, Peninsula Health, Frankston, Australia (J.C., N.H., R.L., M.F., J.L.).,Peninsula Clinical School, Monash University, Frankston, Australia (J.C., N.H., J.L.)
| | - Robert Lew
- Peninsula Heart Service, Peninsula Health, Frankston, Australia (J.C., N.H., R.L., M.F., J.L.)
| | - Mark Freilich
- Peninsula Heart Service, Peninsula Health, Frankston, Australia (J.C., N.H., R.L., M.F., J.L.)
| | - Stephen Quinn
- Department of Health Science and Biostatistics, Swinburne University of Technology, Hawthorn, Victoria, Australia (S.Q.)
| | - Jamie Layland
- Peninsula Heart Service, Peninsula Health, Frankston, Australia (J.C., N.H., R.L., M.F., J.L.).,Peninsula Clinical School, Monash University, Frankston, Australia (J.C., N.H., J.L.)
| |
Collapse
|