1
|
Zhou AQ, Cao Y, Song YN, Zhang BF, Chen KY, Yang SY, Chen HJ. miR-92a-1-5p targets MEF2A to induce insulin resistance in myocardial ischemia/reperfusion injury. Biochem Biophys Res Commun 2025; 768:151938. [PMID: 40345013 DOI: 10.1016/j.bbrc.2025.151938] [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: 02/24/2025] [Revised: 04/15/2025] [Accepted: 05/02/2025] [Indexed: 05/11/2025]
Abstract
PURPOSE Improving myocardial energy metabolism is an important way to alleviate myocardial ischemia/reperfusion injury (MIRI). Myocardial insulin resistance (IR) can occur after MIRI and cause the inhibition of glucose absorption and metabolism. This study aimed to detect the mechanism of miR-92a-1-5p in MIRI-induced myocardial IR. METHODS First, MIRI rat models were established using the Langendorff technique. H9c2 cells were treated with oxygen-glucose deprivation/reperfusion (OGD/R) to establish in vitro cell models. The expression levels of miR-92a-1-5p and myocyte enhancer factor 2A (MEF2A) were detected using RT-qPCR, and the expression of glucose transporter 4 (GLUT4) in the cell membrane and MEF2A was detected using Western blot. Immunofluorescence was used to detect GLUT4 expression in the cell membrane of H9c2 cells. Glucose absorption was detected in H9c2 cells using flow cytometry. H&E staining was used to determine pathological changes in heart tissue. H9c2 cell viability was detected using CCK-8 assay, and the binding affinity between miR-92a-1-5p and MEF2A was verified using dual luciferase reporter assay. RESULTS miR-92a-1-5p expression increased, and MEF2A expression decreased after OGD/R in H9c2 cells or MIRI in rats. Overexpression of miR-92a-1-5p aggravated myocardial tissue and H9c2 cell damage, inhibited the translocation of GLUT4 to the cell membrane, and reduced glucose absorption. Inhibiting the miR-92a-1-5p yielded the opposite results. MEF2A overexpression reversed the injury, which was exacerbated by miR-92a-1-5p, and promoted the translocation of GLUT4 to the cell membrane and glucose absorption. The double luciferase reporter assay results showed that miR-92a-1-5p could negatively regulate the expression of MEF2A. CONCLUSION miR-92a-1-5p expression increased after IR in myocardial tissue and H9c2 cells. Inhibition of miR-92a-1-5p increased MEF2A expression, promoted GLUT4 translocation, and increased glucose absorption, thereby reducing MIRI.
Collapse
Affiliation(s)
- An-Qiang Zhou
- Division of Cardiac Surgery, Guizhou Institute of Precision Medicine, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550009, China; Translational Medicine Research Center, Guizhou Province Key Laboratory of Regenerative Medicine, Tissue Engineering and Stem Cell Experiment Center, Guizhou Medical University, GuiAn, 561113, Guizhou, China
| | - Ying Cao
- Department of Anesthesiology, The Affiliated JinYang Hospital of Guizhou Medical University, The Second People's Hospital of Guiyang, Guiyang, 550023, Guizhou, China
| | - Ying-Nan Song
- Division of Cardiac Surgery, Guizhou Institute of Precision Medicine, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550009, China
| | - Ben-Fa Zhang
- Translational Medicine Research Center, Guizhou Province Key Laboratory of Regenerative Medicine, Tissue Engineering and Stem Cell Experiment Center, Guizhou Medical University, GuiAn, 561113, Guizhou, China
| | - Kai-Yuan Chen
- Division of Cardiac Surgery, Guizhou Institute of Precision Medicine, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550009, China
| | - Si-Yuan Yang
- Division of Cardiac Surgery, Guizhou Institute of Precision Medicine, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550009, China.
| | - Hong-Jin Chen
- Division of Cardiac Surgery, Guizhou Institute of Precision Medicine, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550009, China; Translational Medicine Research Center, Guizhou Province Key Laboratory of Regenerative Medicine, Tissue Engineering and Stem Cell Experiment Center, Guizhou Medical University, GuiAn, 561113, Guizhou, China; Department of Pharmacology, School of Basic Medical Sciences, Guizhou Medical University, GuiAn, 561113, Guizhou, China.
| |
Collapse
|
2
|
De Bartolo A, Romeo N, Angelone T, Rocca C. Specialized Pro-Resolving Mediators as Emerging Players in Cardioprotection: From Inflammation Resolution to Therapeutic Potential. Acta Physiol (Oxf) 2025; 241:e70062. [PMID: 40433738 PMCID: PMC12117521 DOI: 10.1111/apha.70062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/18/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025]
Abstract
AIM Timely myocardial reperfusion is essential for restoring blood flow to post-ischemic tissue, thereby reducing cardiac injury and limiting infarct size. However, this process can paradoxically result in additional, irreversible myocardial damage, known as myocardial ischemia-reperfusion injury (MIRI). The goal of this review is to explore the role of specialized pro-resolving mediators (SPMs) in atherosclerosis and MIRI, and to assess the therapeutic potential of targeting inflammation resolution in these cardiovascular conditions. METHODS This review summarizes current preclinical and clinical evidence on the involvement of SPMs in the pathogenesis of atherosclerosis and MIRI, acknowledging that several cellular and molecular aspects of their mechanisms of action remain to be fully elucidated. RESULTS MIRI is a complex phenomenon in which inflammation, initially triggered during ischemia and further amplified upon reperfusion, plays a central role in its pathogenesis. Various cellular and molecular players mediate the initial pro-inflammatory response and the subsequent anti-inflammatory reparative phase following acute myocardial infarction (AMI), contributing both to ischemia- and reperfusion-induced damage as well as to the healing process. SPMs have emerged as key endogenous immunoresolvents with potent anti-inflammatory, antioxidant, and pro-resolving properties that contribute to limit excessive acute inflammation and promote tissue repair. While dysregulated SPM-related signaling has been linked to various cardiovascular diseases (CVD), their precise role in AMI and MIRI remains incompletely understood. CONCLUSION Targeting inflammation resolution may represent a promising therapeutic strategy for mitigating atheroprogression and addressing a complex condition such as MIRI.
Collapse
Affiliation(s)
- Anna De Bartolo
- Cellular and Molecular Cardiovascular Physiology and Pathophysiology Laboratory, Department of Biology, E. And E. S. (DiBEST)University of CalabriaCosenzaItaly
| | - Naomi Romeo
- Cellular and Molecular Cardiovascular Physiology and Pathophysiology Laboratory, Department of Biology, E. And E. S. (DiBEST)University of CalabriaCosenzaItaly
| | - Tommaso Angelone
- Cellular and Molecular Cardiovascular Physiology and Pathophysiology Laboratory, Department of Biology, E. And E. S. (DiBEST)University of CalabriaCosenzaItaly
- National Institute of Cardiovascular Research (INRC)BolognaItaly
| | - Carmine Rocca
- Cellular and Molecular Cardiovascular Physiology and Pathophysiology Laboratory, Department of Biology, E. And E. S. (DiBEST)University of CalabriaCosenzaItaly
- National Institute of Cardiovascular Research (INRC)BolognaItaly
| |
Collapse
|
3
|
Liu Y, Yin H, Wang T, Chen T, Guo C, Zhang F, Jiang Z. Myeloid SHP2 attenuates myocardial ischemia‑reperfusion injury via regulation of BRD4/SYK/STING/NOX4/NLRP3 signaling. Mol Med Rep 2025; 31:155. [PMID: 40211713 PMCID: PMC12005128 DOI: 10.3892/mmr.2025.13520] [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: 08/16/2024] [Accepted: 01/17/2025] [Indexed: 04/19/2025] Open
Abstract
The objective of the present study was to investigate the impact of myeloid Src homology region 2‑containing protein tyrosine phosphatase 2 (SHP2) on myocardial ischemia reperfusion (MI/R) injury and the underlying mechanism. Bioinformatics was used to analyze genes specifically associated with MI/R. In addition, myeloid‑specific SHP2 knockout mice and wild‑type mice were subjected to MI/R or sham surgery. Echocardiography and Masson's staining were used to observe the myocardial function and infarct area of the mice. In addition, double immunofluorescence staining was used to detect the relative fluorescence intensity of SHP2 and bromodomain‑containing protein 4 (BRD4) in bone marrow‑derived macrophages (BMMs) from the mice. Western blot analysis was conducted to determine the expression levels of SHP2, BRD4, spleen tyrosine kinase (SYK), stimulator of interferon genes (STING), NADPH oxidase 4 (NOX4), NLR family pyrin domain containing 3 (NLRP3), IL‑1β and gasdermin D (GSDMD) in BMMs and mouse myocardial cells co‑cultured with the BMMs. In addition, flow cytometry was employed to assess myocardial cell apoptosis. Bioinformatics analysis revealed the downregulated expression of SHP2 and upregulated expression of BRD4 and SYK in mice with MI/R. The deletion of myeloid SHP2 aggravated MI/R injury, impaired cardiac function and increased the infarct area in mice. In addition, myeloid SHP2 deletion in BMMs promoted the expression of BRD4, SYK, STING, NOX4 and NLRP3 in BMMs, and the expression of IL‑1β and GSDMD in mouse myocardial cells co‑cultured with the BMMs. In addition, the deletion of myeloid SHP2 promoted cardiomyocyte apoptosis. These results indicate that myeloid SHP2 inhibits MI/R injury by regulating BRD4/SYK/STING/NOX4/NLRP3 signaling in BMMs.
Collapse
Affiliation(s)
- Yazhong Liu
- Department of Cardiovascular Disease and Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| | - Hongshan Yin
- Department of Cardiovascular Disease and Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| | - Tao Wang
- Department of Cardiovascular Disease and Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| | - Tao Chen
- Department of Cardiovascular Disease and Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| | - Chengda Guo
- Department of Cardiovascular Disease and Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| | - Fue Zhang
- Department of Cardiovascular Disease and Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| | - Zhian Jiang
- Department of Cardiovascular Disease and Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| |
Collapse
|
4
|
Chen D, Wen J, Zang W, Lin X. Knockdown of Galectin-3 confers myocardial protection against ischemia-reperfusion injury, modulating oxidative stress, inflammatory response, and the peroxisome proliferator-activated receptor g signaling pathway. Cytojournal 2025; 22:49. [PMID: 40539121 PMCID: PMC12178088 DOI: 10.25259/cytojournal_12_2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 03/19/2025] [Indexed: 06/22/2025] Open
Abstract
Objective Ischemia-reperfusion (I-R) injury in the myocardium is a considerable challenge in cardiovascular medicine, posing a severe threat to life. Given that galectin-3 possibly regulates myocardial I-R damage, this study aims to investigate the detailed mechanisms underlying galectin-3's effects on myocardial I-R injury. Material and Methods The expression levels of galectin-3 in vivo and in vitro myocardial I-R models were determined by Western blot and quantitative real-time polymerase chain reaction. The effects of galectin-3 on inflammatory factors and oxidative stress factors in myocardial I-R were measured with an enzyme-linked immunosorbent assay, and the extent of myocardial tissue damage was assessed using hematoxylin-eosin staining. The influence of galectin-3 on peroxisome proliferator-activated receptor g (PPARg) signaling pathway-related proteins in myocardial I-R was determined by Western blot. Results Myocardial I-R damage was associated with increased galectin-3 expression, and the blood levels of creatine kinase-myocardial band and creatine kinase were favorably correlated with the messenger RNA levels of galectin-3 in mice with cardiac I-R damage. The inhibition of galectin-3 alleviated oxidative stress and inflammatory response, and galectin-3 promoted reactive oxygen species production in myocardial I-R cells. Furthermore, the cardiac I-R damage mouse model exhibited decreased expression of proteins linked to the PPARg signaling pathway, but galectin-3 inhibition enhanced the expression of these proteins. Conclusion Galectin-3 plays a crucial role in exacerbating myocardial I-R injury, and its up-regulation is associated with increased oxidative stress, inflammatory responses, and inhibition of the protective PPARg signaling pathway. The alleviation of these harmful effects by galectin-3 inhibition suggests that targeting galectin-3 is a potential therapeutic method for reducing myocardial I-R injury.
Collapse
Affiliation(s)
- Duo Chen
- Department of Cardiovascular Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Jingyu Wen
- Department of Traditional Chinese Medicine, Shandong Rehabilitation Hospital, Jinan, Shandong, China
| | - Wei Zang
- Department of Traditional Chinese Medicine, Jinan Massage Hospital, Jinan, Shandong, China
| | - Xuehong Lin
- Department of Cardiovascular Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| |
Collapse
|
5
|
Dong J, Li Z, Fu C, Yang D, Yang H, Lin L, Liang XJ, Chen Z, Chen L, Guo W. Cardiosplenic axis-targeted immunomodulatory liposome for myocardial ischemia-reperfusion injury treatment. J Control Release 2025; 383:113799. [PMID: 40324533 DOI: 10.1016/j.jconrel.2025.113799] [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: 01/27/2025] [Revised: 04/02/2025] [Accepted: 04/28/2025] [Indexed: 05/07/2025]
Abstract
Monocyte/macrophage (Mo/Mϕ) infiltration is critical in myocardial ischemia-reperfusion injury (MIRI). However, the complex composition of the myocardium severely hinders drug accumulation and makes it challenging to modulate the Mo/Mϕ immune response at the MIRI site. The spleen, acting as a Mo/Mϕ reservoir, plays a crucial role in the development of MIRI along the cardiosplenic axis. Compared to directly delivering medications to the MIRI site, targeting the spleen for Mo/Mϕ immunomodulation provides an alternative strategy to modulate the immunological phenotype on-site. Therefore, we developed a melatonin-loaded liposome (ST-MT@lipo2) that specifically targets the spleen and can effectively regulate the immunological response of splenic monocytes and macrophages, consequently enhancing their immune response at the site of MIRI. Additionally, the splenectomy mouse model revealed that ST-MT@lipo2 regulated MIRI's immune response through the cardiosplenic axis by regulating the MCP-1/CCR2 pathway to reduce circulating inflammatory monocyte migration from the spleen to the MIRI site. Moreover, pathological staining and echocardiography showed that ST-MT@lipo2 reduced myocardial damage and improved cardiac function in MIRI mice. This study demonstrates the crucial importance of modulating the immune response in the cardiosplenic axis for treating MIRI, which also inspired the treatments for inflammatory diseases by controlling the spleen immunological milieu.
Collapse
Affiliation(s)
- Jiankai Dong
- Department of Cardiology, Fujian Medical University Union Hospital, Fuzhou 250001, China
| | - Zechuan Li
- Department of Minimally Invasive Interventional Radiology, The Second Affiliated Hospital, School of Biomedical Engineering, Guangzhou Medical University, Guangzhou 510260, China
| | - Chenxing Fu
- Department of Minimally Invasive Interventional Radiology, The Second Affiliated Hospital, School of Biomedical Engineering, Guangzhou Medical University, Guangzhou 510260, China
| | - Ding Yang
- Department of Radiology, Peking University Cancer Hospital & Institute, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Beijing 100142, China
| | - Huijie Yang
- Department of Neurology, The Fifth Affiliated Hospital of Southern Medical University, Guangzhou 510260, China
| | - Li Lin
- Department of Cardiology, Fujian Medical University Union Hospital, Fuzhou 250001, China
| | - Xing-Jie Liang
- Department of Minimally Invasive Interventional Radiology, The Second Affiliated Hospital, School of Biomedical Engineering, Guangzhou Medical University, Guangzhou 510260, China; Laboratory of Controllable Nanopharmaceuticals, Chinese Academy of Sciences (CAS) Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Zhaoyang Chen
- Department of Cardiology, Fujian Medical University Union Hospital, Fuzhou 250001, China.
| | - Lianglong Chen
- Department of Cardiology, Fujian Medical University Union Hospital, Fuzhou 250001, China.
| | - Weisheng Guo
- Department of Minimally Invasive Interventional Radiology, The Second Affiliated Hospital, School of Biomedical Engineering, Guangzhou Medical University, Guangzhou 510260, China.
| |
Collapse
|
6
|
Yang S, Yan L, Chen L, Su G, Yang L, Gong L, Liu L. Cardiac PDK4 promotes neutrophilic PFKL methylation and drives the innate immune response in diabetic myocardial infarction. Pharmacol Res 2025; 215:107731. [PMID: 40222696 DOI: 10.1016/j.phrs.2025.107731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 03/20/2025] [Accepted: 04/06/2025] [Indexed: 04/15/2025]
Abstract
NETosis plays a pivotal role in the innate immune response after diabetic myocardial infarction (MI), exerting a profound influence on the overall pathological process and potential recovery outcomes. The metabolism of diabetic cardiomyocyte actively creates a specialized micro environment for the innate immune response after MI. However, the mechanism by which cardiac metabolism drives NETosis remains unclear. Utilizing public databases of human MI sc-RNA datasets, we discovered that cardiomyocyte PDK4 expression mediates the intensification of glycolysis, which is strongly correlated with NETosis. Through mass spectrometry imaging and phenotype assessment, we ascertained that specific knockout of PDK4 in cardiomyocytes (PDK4fl/flMyh6Cre, male, 6 weeks) led to a reduction in NETosis by restraining micro environmental lactate (LA) production. In addition, the role of LA in promoting NETosis has been further corroborated by in vivo/in vitro experiments involving LA supplementation and its absence. Moreover, LA redirects neutrophil metabolic flux from glycolysis to the pentose-phosphate pathway (PPP). Mechanistically, LA triggers metabolic remodeling through the PRMT9-mediated methylation of PFKL at the R301 residue, resulting in PFKL inactivation and the consequent restriction of glycolysis. Our findings reveal the crucial role of cardiomyocyte metabolism in NETosis, shedding light on the role of LA as a vital signaling molecule in the crosstalk between cardiomyocytes and neutrophils. Importantly, we screened pitavastatin, a potential inhibitor of PDK4 among the FDA-approved drugs, and verified that it can alleviate NETosis in diabetic MI, which provides a rationale for drug selection in diabetic MI patients.
Collapse
Affiliation(s)
- Song Yang
- China-Japan Friendship Hospital, Beijing 100029, China
| | - Longxin Yan
- China-Japan Friendship Hospital, Beijing 100029, China
| | - Lang Chen
- China-Japan Friendship Hospital, Beijing 100029, China
| | - Gaijuan Su
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100007, China
| | - Long Yang
- Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Lili Gong
- China-Japan Friendship Hospital, Beijing 100029, China.
| | - Lihong Liu
- China-Japan Friendship Hospital, Beijing 100029, China.
| |
Collapse
|
7
|
Li J, Guo Y, Yang Y, Xue Q, Cao H, Yang G, Sun Z, Jia LL, Yu HB. The cardioprotective effects of acteoside in myocardial ischemia reperfusion injury and the underlying mechanism. BMC Cardiovasc Disord 2025; 25:329. [PMID: 40287651 PMCID: PMC12032671 DOI: 10.1186/s12872-025-04762-0] [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: 11/18/2024] [Accepted: 04/13/2025] [Indexed: 04/29/2025] Open
Abstract
INTRODUCTION We observed the cardioprotective effects of Acteoside (AC) on myocardial ischemia reperfusion injury (MIRI) and discussed the possible mechanisms. METHODS Before MIRI model was established successfully, AC was administrated to SD rats by gastric route for 7 d. Punctuate paw withdrawal threshold (PWT) was recorded to reflect the pain threshold. Blood samples were collected to measure the levels of oxidative stress, myocardial enzymes and Norepinephrine (NE). Hematoxylin and eosin (HE) staining was performed to observe the pathological changes of myocardial tissues. Apoptosis of myocardial cell was determined by transferase-mediated dUTP nick end labeling (TUNEL) assay, and the expressions of Bcl-2 and Bax were determined by Western blotting. Using network pharmacological analysis, the PI3K/Akt signaling pathway was screened to be associated with both AC and MIRI. Subsequently, the expressions of PI3K, p-Akt and caspase-3 were detected by immunochemistry in myocardial tissues. RESULTS We found that pre-administration of AC improved pain threshold and pathological change of myocardial structure caused by MIRI. AC reduced serum levels of myocardial enzymes and NE in MIRI. Compared with the Sham group, rats in MIRI group showed enhanced oxidative stress levels. These changes were partly reversed by AC. In addition, AC inhibited apoptosis, regulated the expression of apoptosis-related proteins. Immunochemistry analysis confirmed that AC increased the expressions of PI3K and p-Akt in myocardial tissue. CONCLUSION The cardioprotective effects of AC in MIRI were related with pain alleviation, oxidative stress, apoptosis and sympathetic nerve activity inhibition, the PI3K/Akt signal pathway activation. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Jing Li
- Department of Physiology, Basic medical college, Jiamusi University, 148 Xuefu Street, Jiamusi, Heilongjiang Province, 154000, P. R. China
| | - Yuxin Guo
- Department of Physiology, Basic medical college, Jiamusi University, 148 Xuefu Street, Jiamusi, Heilongjiang Province, 154000, P. R. China
| | - Yang Yang
- Department of Physiology, Basic medical college, Jiamusi University, 148 Xuefu Street, Jiamusi, Heilongjiang Province, 154000, P. R. China
| | - Qing Xue
- Department of Cardiology, the First affiliated hospital to Jiamusi University, 348 Dexiang Street Jiamusi, Jiamusi, Heilongjiang Province, 154000, P. R. China
| | - Hong Cao
- Department of Cardiology, the First affiliated hospital to Jiamusi University, 348 Dexiang Street Jiamusi, Jiamusi, Heilongjiang Province, 154000, P. R. China
| | - Guangyuan Yang
- Department of Cardiology, the First affiliated hospital to Jiamusi University, 348 Dexiang Street Jiamusi, Jiamusi, Heilongjiang Province, 154000, P. R. China
| | - Zhiqi Sun
- Daqing Oilfield General Hospital, No. 195, Zhongqiao Road, Saltu District, Daqing City, Heilongjiang Province, 163000, P. R. China
| | - Lin-Lin Jia
- Department of Physiology, Basic medical college, Jiamusi University, 148 Xuefu Street, Jiamusi, Heilongjiang Province, 154000, P. R. China.
- Zhangzhou Health Vocational College, No.29 Xiyangping Road, Xiangcheng District, Zhangzhou, Fujian Province, 363000, P. R. China.
| | - Hai-Bo Yu
- Department of Cardiology, the First affiliated hospital to Jiamusi University, 348 Dexiang Street Jiamusi, Jiamusi, Heilongjiang Province, 154000, P. R. China.
| |
Collapse
|
8
|
Zhang J, Feng S, Geng Y, Wang X, Wang Z, Liu Y. Anti-inflammatory phenotypes of immune cells after myocardial infarction and prospects of therapeutic strategy. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04167-y. [PMID: 40278891 DOI: 10.1007/s00210-025-04167-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Accepted: 04/09/2025] [Indexed: 04/26/2025]
Abstract
Often causing negative cardiac remodeling and heart failure, a major threat to human life and health, myocardial infarction (MI) is a cardiovascular disease with a high morbidity and fatality rate worldwide. Maintaining ordinary heart function depends significantly on the immune system. Necrotic cardiomyocyte signals promote specific immunity and activate general immunity as the disease progresses in MI. Complex immune cells play a key role in all stages of MI progression by removing necrotic cardiomyocytes and tissue and promoting the healing of damaged tissue cells. Immune cells can help to regrow injured heart muscle as well as enable both inflammation and cardiomyocyte death. Immune cells are essential elements that help the immune system carry out its protective function. There are two types of immunity: nonspecific immunity and specific immunity. Developed throughout the long-term evolution of species, nonspecific immunity (including macrophages, myeloid-derived suppressor cells MDSC, natural killer cells NK, neutrophils, and dendritic cells DC) offers immediate and conservative host defense that might destroy healthy tissues because of its nonspecific nature. Precisely acquired immunity, specific immunity helps humoral and cellular immunity mediated through B and T cells correspondingly. These findings offer crucial information needed for the creation of effective immunomodulatory treatment, as discussed in this article.
Collapse
Affiliation(s)
- Jiacheng Zhang
- Department of Cardiovascular, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250002, Shandong, China
| | - Shuai Feng
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250300, Shandong, China
| | - Yannan Geng
- Pharmaceutical Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250002, Shandong, China
| | - Xiaoli Wang
- Pharmaceutical Department, Liaocheng People's Hospital, Liaocheng, 252002, Shandong, China
| | - Zhen Wang
- Department of Cardiovascular, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250002, Shandong, China.
| | - Yang Liu
- Department of Cardiovascular, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250002, Shandong, China.
| |
Collapse
|
9
|
Cen W, Pan Y, Tang Y, Yu J, Xuan Y, Huang J, Wei S, Zhang J. Novel diagnostic biomarkers regulating macrophages autophagy in ischemic cardiomyopathy: An analysis integrating bulk RNA sequencing with single-cell RNA sequencing. Immunobiology 2025; 230:152907. [PMID: 40300424 DOI: 10.1016/j.imbio.2025.152907] [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: 01/09/2025] [Revised: 04/01/2025] [Accepted: 04/23/2025] [Indexed: 05/01/2025]
Abstract
Macrophage autophagy plays a pivotal role in ischemia cardiomyopathy (ICM). However, the underlying mechanisms and macrophage autophagy-related biomarkers in ICM have not been elucidated. Therefore, this study was designed to explore novel macrophage autophagy-related biomarkers for ICM. The autophagy-related genes were downloaded from the Human Autophagy Modulator and intersected with the differentially expressed genes (DEGs) of GSE46224 identified with "limma" package in R to obtain the autophagy-related DEGs. Immune infiltration analysis showed that macrophages were the dominant immune cells in ICM tissue. Then the macrophage autophagy-related DEGs were identified using the weighted gene co-expression network analysis (WGCNA). A total of six hub genes were obtained from the PPI network. All of the hub genes showed specific diagnostic significance with AUCs higher than 0.7, as also validated in the external dataset GSE116250. RT-qPCR was conducted to detect the mRNA expression levels of hub genes in vivo ICM rat model. Single-cell RNA sequencing analysis was also performed to investigate gene expression profiles. Our study explored the macrophage autophagy-related biomarkers and their relative pathways in ICM, provided novel diagnostic biomarkers for ICM, and gave new insight into the progression mechanism of ICM.
Collapse
Affiliation(s)
- Weiluan Cen
- Department of Emergency, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yajin Pan
- Department of Cardiology, National Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Yaohan Tang
- Department of Emergency, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jianing Yu
- Department of General Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yixuan Xuan
- Department of General Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jingyu Huang
- Department of General Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Shanshan Wei
- Department of General Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jianfeng Zhang
- Department of Emergency, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China; Department of General Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China.
| |
Collapse
|
10
|
Du X, Liu J, Zhang Z, Ren Y, Chen L, Lu Y, Zhang Z. Predictive value of lymphocyte-to-C-reactive protein ratio for left ventricular thrombus in patients with ST-segment elevation myocardial infarction. Front Cardiovasc Med 2025; 12:1465350. [PMID: 40342978 PMCID: PMC12058794 DOI: 10.3389/fcvm.2025.1465350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 04/07/2025] [Indexed: 05/11/2025] Open
Abstract
Background and purpose Current evidence suggested a correlation between inflammation and Left Ventricular Thrombus (LVT). The lymphocyte to C-reactive protein ratio (LCR) has been established as be a reliable inflammation marker and is associated with the prognosis of patients with ST-segment elevation myocardial infarction (STEMI). However, its relationship with the occurrence of LVT remains unclear. This study aims to evaluate the effectiveness of LCR in predicting LVT in patients with STEMI after undergoing primary percutaneous coronary intervention (pPCI). Methods A total of 564 STEMI patients who underwent pPCI at the Affiliated Hospital of Xuzhou Medical University from September 2019 to June 2024 were included. Cardiac magnetic resonance imaging (CMR) was used to assess myocardial infarction characteristics and the presence of LVT. The definition of LCR is the lymphocyte to C-reactive protein ratio. Results Out of 564 patients, 57 were diagnosed with LVT. The median time for CMR testing was 5 (4, 6) days. Univariate regression analysis showed significant differences in left ventricular ejection fraction (LVEF), peak N-terminal pro B-type natriuretic peptide (peak NT-proBNP), peak high-sensitivity troponin T (peak hsTnT), LCR, Late Gadolinium Enhancement% (LGE%), and Microvascular Obstruction% (MVO%) (p < 0.05). Multivariate regression analysis indicated that LCR was an independent predictor for LVT (P = 0.007, OR: 0.001 95% CI: 0.00-0.123). Receiver operating characteristic (ROC) curve analysis showed that LCR has good predictive ability for LVT (Area under the curve: 0.704, p < 0.001). Integration of integral LCR could significantly improve the discrimination and reclassification accuracy for LVT after STEMI (NRI = 0.517, IDI = 0.030; p < 0.001). Conclusion Lower LCR is independently associated with the risk of LVT in patients with STEMI after pPCI. Integration of LCR can significantly improve the risk model for LVT.
Collapse
Affiliation(s)
- Xinjia Du
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Jiahua Liu
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Zeqing Zhang
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yanfei Ren
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Lei Chen
- Department of Cardiology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuan Lu
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Zhuoqi Zhang
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
11
|
Liu X, Zhan Z, Zhang R, Wang Y, Xu Q. Network pharmacology-based prediction and molecular docking-based strategy to investigate the potential mechanism of Leonurus japonicus Houtt. Against myocardial ischemia reperfusion injury. J Cardiothorac Surg 2025; 20:204. [PMID: 40241120 PMCID: PMC12001406 DOI: 10.1186/s13019-025-03425-7] [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: 11/13/2024] [Accepted: 04/06/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Leonurus japonicus Houtt. (LJH) has multiple pharmacological effects. OBJECTIVE To investigate the potential mechanism of LJH in the treatment of myocardial ischemia-reperfusion injury (MIRI) using network pharmacology, molecular docking technology, and in vitro experiments. METHODS Herbs for ischemic heart disease were identified with the help of herb-disease databases. The TCMSP database was used to find the potential targets of LJH. Disease targets of MIRI were identified with the help of Disgenet, Genecard, Alliance of Genome Resources databases. The common targets were obtained with the help of VENN diagram, and the common targets were analyzed by GO function and KEEG pathway enrichment to predict the potential mechanism of action of LJH in treating MIRI. With the help of STRING database and Cytoscape software, we constructed a visual protein-protein interaction (PPI) network model to screen the core targets and then docked the core targets with the corresponding ligand molecules. AC16 cells were used to simulate MIRI by glucose-oxygen deprivation, and apoptosis was detected by Annexin V-FITC/PI double staining; protein expression was detected by Western blot. RESULTS LJH was one of the herbal remedies for the treatment of ischemic heart disease. LJH had 247 potential targets of action and 26 targets in common with MIRI. These 26 targets were enriched in the TNF signaling pathway and NF-kappa B signaling pathway, and the core targets screened by the PPI results included TNF, VCAM1, and MMP9. Molecular docking results showed that the compounds in LJH docked well with the core target proteins. In vitro experiments showed that LJH could inhibit the elevation of TNF, VCAM1, and MMP9 after MIRI, reduce apoptosis, and inhibit inflammation. CONCLUSION The mechanism of LJH in the treatment of MIRI was mainly related to the activation of TNF signaling pathway and NF-kappa B signaling pathway, and the regulation of TNF, VCAM1, and MMP9 protein expression.
Collapse
Affiliation(s)
- Xuan Liu
- Department of Cardiology, The Second Affiliated Hospital of Tianjin University of TCM, No. 69, Zengchan Road, Hebei District, Tianjin, 300150, China
| | - Zilian Zhan
- Department of Cardiology, The Second Affiliated Hospital of Tianjin University of TCM, No. 69, Zengchan Road, Hebei District, Tianjin, 300150, China
| | - Rui Zhang
- Department of Cardiology, The Second Affiliated Hospital of Tianjin University of TCM, No. 69, Zengchan Road, Hebei District, Tianjin, 300150, China
| | - Yadong Wang
- Department of Cardiology, The Second Affiliated Hospital of Tianjin University of TCM, No. 69, Zengchan Road, Hebei District, Tianjin, 300150, China
| | - Qiang Xu
- Department of Cardiology, The Second Affiliated Hospital of Tianjin University of TCM, No. 69, Zengchan Road, Hebei District, Tianjin, 300150, China.
| |
Collapse
|
12
|
He X, Wang Q, Long Q, Zhong Y, Qi Z, Zhang Y, Chang L, Qian B, Huang S, Wang X, Chen X, Li F, Yang X, Gao WD, Song Z, Xu L, Zhao Q. The single-cell atlas of short-chain fatty acid receptors in human and mice hearts. Front Immunol 2025; 16:1538384. [PMID: 40308581 PMCID: PMC12040890 DOI: 10.3389/fimmu.2025.1538384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 03/18/2025] [Indexed: 05/02/2025] Open
Abstract
Introduction The gut microbiota metabolite, short-chain fatty acids (SCFAs), can protect against multiple cardiovascular diseases, while the molecular targets and underlying mechanisms need to be elucidated. One of the primary mechanisms of SCFA benefits was the direct activation of a group of G-protein-coupled receptors (GPCRs), termed free fatty acid receptors (FFARs), the FFAR2 (GPR43), and FFAR3 (GPR41). At present, the distribution of FFAR2/3 in cardiac cells has not been entirely clarified. Methods Using 18 public single-cell RNA-seq and single-nuclear RNA-seq data of human and mouse hearts, we illustrate the entire atlas of FFAR2/3 distribution in different regions and cell types in normal and infarcted hearts. Results and discussion We present the atlas of FFAR2/3 in the whole human body, normal and infarcted hearts at single-cell resolution. We also illustrated the entire atlas of FFAR2/3 in normal/ischemic hearts of newborn and adult mice by combining public and newly built sc/snRNA-seq datasets. These findings provide valuable information on the possible effect of SCFAs via FFAR2/3 in the heart and valuable references for future studies.
Collapse
Affiliation(s)
- Xiaojun He
- Cardiovascular Medical Center, Department of Cardiovascular Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Qiang Wang
- Cardiovascular Medical Center, Department of Cardiovascular Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Qiang Long
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yiming Zhong
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhaoxi Qi
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yecen Zhang
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Lan Chang
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Bei Qian
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Shixing Huang
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xinming Wang
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xuemei Chen
- Department of Anesthesiology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Feifei Li
- Department of Cardiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiaomei Yang
- Department of Anesthesiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Cardiology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Wei Dong Gao
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Zhizhao Song
- Cardiovascular Medical Center, Department of Cardiovascular Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Li Xu
- Clinical Trial Institution, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Qiang Zhao
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
13
|
Liu X, Shui G, Wang Y, Chen T, Zhang P, Liu L, Li C, Li T, Wang X. Remimazolam alleviates myocardial ischemia/reperfusion injury and inflammation via inhibition of the NLRP3/IL‑1β pathway in mice. Int J Mol Med 2025; 55:57. [PMID: 39886966 PMCID: PMC11819765 DOI: 10.3892/ijmm.2025.5498] [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: 07/19/2024] [Accepted: 11/25/2024] [Indexed: 02/01/2025] Open
Abstract
Remimazolam (Rema) is a novel anesthetic that is widely used in anesthesia and sedation in critically ill patients. Notably, Rema exerts effects in patients through activation of the γ‑aminobutyric acid (GABA) receptor. GABA may alleviate myocardial ischemia/reperfusion (I/R) injury; however, the impact of Rema and underlying molecular mechanism in myocardial I/R injury remain to be fully understood. Therefore, the present study aimed to investigate the effects of Rema on cardiac I/R injury and to determine the underlying mechanisms. An acute myocardial I/R model was established by ligating the left anterior descending artery in adult male C57BL/6 mice (8‑10 weeks). Cultured Raw264.7 cells treated with lipopolysaccharide (LPS) were also used to investigate the effect of Rema on macrophages. The results of the present study revealed that Rema improved I/R‑induced cardiac dysfunction by increasing the ejection fraction value and reducing the myocardial infarction area. In addition, Rema also alleviated I/R‑induced cardiac inflammatory cell infiltration based on H&E and immunofluorescence staining. Transmission electron microscopy and ROS measurements showed that Rema improved I/R‑induced mitochondrial structural disruption and oxidative stress in cardiomyocytes. Transcriptomics analysis and reverse transcription‑quantitative PCR revealed that Rema alleviated I/R‑induced release of inflammatory factors and cytokines by inhibiting the expression of IL‑1β, IL‑6, C‑C chemokine receptor 2 and C‑X‑C motif chemokine ligand 5. Rema also inhibited I/R‑induced CD68+ cell proliferation, IL‑1β release, and NOD‑like receptor thermal protein domain associated protein 3 (NLRP3) and IL‑1β expression. The results of in vitro assays revealed that Rema inhibited LPS‑induced increases in IL‑1β, IL‑6 and TNF‑α expression and release in cultured RAW264.7 macrophages. In conclusion, the present study revealed that Rema may alleviate I/R‑induced cardiac dysfunction and myocardial injury by inhibiting oxidative stress and inflammatory responses via the NLRP3/IL‑1β pathway.
Collapse
Affiliation(s)
- Xueru Liu
- Department of Anesthesiology, Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Guojing Shui
- Department of Anesthesiology, Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Yan Wang
- Key Laboratory of Medical Electrophysiology of The Ministry of Education, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Tangting Chen
- Key Laboratory of Medical Electrophysiology of The Ministry of Education, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
- Department of Cardiology, Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Peng Zhang
- Key Laboratory of Medical Electrophysiology of The Ministry of Education, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Li Liu
- Department of Anesthesiology, Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Tao Li
- Key Laboratory of Medical Electrophysiology of The Ministry of Education, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
- Department of Cardiology, Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Xiaobin Wang
- Department of Anesthesiology, Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| |
Collapse
|
14
|
Mo C, Tang X, Wei Y, Han H, Wei G, Wei L, Lin X. miRNA-148a-3p targets to regulate the lipid metabolism gene SOCS3 to reduce myocardial ischemia/reperfusion injury. Minerva Cardiol Angiol 2025; 73:136-146. [PMID: 39535526 DOI: 10.23736/s2724-5683.24.06578-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
BACKGROUND Acute myocardial infarction (AMI) is a major cause of death in cardiovascular patients. SOCS3's protective role in cardiac I/R-I is being explored, and miRNAs, particularly miRNA-148a-3p, are suspected to target SOCS3. To elucidate the role of miRNA-148a-3p targeting lipid metabolism gene SOCS3 in cardiac ischemia-reperfusion injury (I/R-I) in rats. METHODS Derived mRNA expression data GSE59867 from GEO, identified 558 lipid metabolism genes from KEGG and GSEA, and screened for differentially expressed genes in acute myocardial infarction (AMI). Predicted miRNA-148a-3p targeting SOCS3 using TargetScanHuman, validated binding via luciferase assay and 3'UTR mutation. Established a rat I/R-I model to assess miRNA-148a-3p and SOCS3 expression, and investigated SOCS3 regulation by miRNA-148a-3p overexpression. Analyzed expression of NF-κB p65, IL-1β, and TNF-α-related proteins, and evaluated cardiac hemodynamics post-SOCS3 regulation by miRNA-148a-3p. RESULTS In GSE59867, TSPO, SOCS3, LRP1, PLB1, CYP1B1, PPARG, ACSL1, and CYP27A1 were identified as differentially expressed lipid metabolism genes in AMI. The results of immune infiltration showed a close relationship between the differential lipid metabolism genes and the infiltration of immune cells such as macrophages and monocytes. The random forest algorithm identified SOCS3 as the key gene. The luciferase reporter gene demonstrated the participation of miRNA-148a-3p in the regulation of SOCS3 by binding to its 3'UTR. In vivo experiments revealed low expression of miRNA-148a-3p in myocardial I/R, while SOCS3 was highly expressed. Elevated miRNA-148a-3p expression led to a decrease in SOCS3, NF-κB p65, IL-1β, and TNF-α levels during cardiac I/R-I. Overexpression of miRNA-148a-3p enhanced the cardiac performance in rats experiencing cardiac I/R-I. CONCLUSIONS Overexpression of miRNA-148a-3p regulates NF-κB signaling pathway by targeting lipid metabolism gene SOCS3, reduces inflammatory response, and then reduces cardiac I/R-I in rats.
Collapse
Affiliation(s)
- Changgan Mo
- The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
- Hechi Hospital Affiliated to Youjiang Medical University for Nationalities, Hechi, Guangxi, China
| | - Xiuge Tang
- Hechi Hospital Affiliated to Youjiang Medical University for Nationalities, Hechi, Guangxi, China
| | - Ying Wei
- Hechi Hospital Affiliated to Youjiang Medical University for Nationalities, Hechi, Guangxi, China
| | - Hui Han
- Hechi Hospital Affiliated to Youjiang Medical University for Nationalities, Hechi, Guangxi, China
| | - Guangsuo Wei
- Hechi Hospital Affiliated to Youjiang Medical University for Nationalities, Hechi, Guangxi, China
| | - Liyuan Wei
- Hechi Hospital Affiliated to Youjiang Medical University for Nationalities, Hechi, Guangxi, China
| | - Xu Lin
- Guangxi Key Laboratory of Basic Medical Research Support for Immune-related Diseases, Baise, Guangxi, China -
- Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| |
Collapse
|
15
|
Bi L, Liu Y, Zhang L, Zhang X, Wang D. High expression of SERPINE1 and CTSL in keratinocytes in pressure injury caused by ischemia-reperfusion injury. Tissue Cell 2025; 93:102746. [PMID: 39864211 DOI: 10.1016/j.tice.2025.102746] [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: 07/25/2024] [Revised: 01/14/2025] [Accepted: 01/15/2025] [Indexed: 01/28/2025]
Abstract
INTRODUCTION Pressure Injury (PI) is a complex disease process which is influenced by multiple factors, among which ischemia-reperfusion (I/R) injury is closely related to the progression of PI. But its biomarkers are still unclearly. Understanding its physiological mechanisms and related molecular biomarkers is a key to developing effective prevention and therapeutic strategies. METHODS This study through obtained the candidate genes of the differentially expressed genes (DEGs) from the PI rat model by transcriptome sequencing, PI single-cell sequencing database, and genes related to I/R injury from GeneCards database to analyze and screen prognostic related target genes. Gene Ontology (GO) and Kyoto Encyclopedia of Genomes (KEGG) pathway analysis were performed using clusterProfiler package, and a protein-protein interaction (PPI) network was constructed to identify hub genes. The genes related to I/R injury were identified and analyzed using three machine learning algorithms. Then, the hub genes were evaluated using nomogram and receiver operating characteristic (ROC) curves, and validated using immunohistochemistry in the PI rat model. RESULTS There were finally 7 candidate genes obtained from the intersection of the three datasets. GO and KEGG pathway analysis revealed that the DEGs were enriched in complement and coagulation cascades, and the keratinocyte differentiation is a significant factor. Then, two hub genes Serine protease inhibitor clade E member 1 (SERPINE1) and Cathepsin L (CTSL) were identified through three machine learning algorithms. The two hub genes were indicated had a high prognosis value by nomogram and ROC curves. SERPINE1 and CTSL both play crucial roles in vasculogenesis, coagulation and degradation of the extracellular matrix, which is essential for wound healing. The results of immunohistochemistry demonstrated that SERPINE1 and CTSL are significantly upregulated in skin tissue from PI caused by I/R injury, and their mRNA expression levels significantly correlate with PI outcomes. CONCLUSION According to our research we referred that the SERPINE1 and CTSL might be the potential biomarkers of PI.
Collapse
Affiliation(s)
- Lexuan Bi
- School of Nursing, Binzhou Medical University, Yantai, Shandong 264003, PR China; Institute of Regenerative Medicine, Binzhou Medical University, Yantai, Shandong 264003, PR China
| | - Yaxin Liu
- School of Nursing, Binzhou Medical University, Yantai, Shandong 264003, PR China
| | - Lei Zhang
- Second Clinical Medical College, Binzhou Medical University, Yantai, Shandong 264003, PR China
| | - Xiaoli Zhang
- School of Nursing, Binzhou Medical University, Yantai, Shandong 264003, PR China.
| | - Dong Wang
- Institute of Regenerative Medicine, Binzhou Medical University, Yantai, Shandong 264003, PR China; Department of Histology and Embryology, Binzhou Medical University, Yantai, Shandong 264003, PR China.
| |
Collapse
|
16
|
Inocencio I, Rai A, Donner D, Greening DW. The Proteomic Landscape of the Coronary Accessible Heart Cell Surfaceome. Proteomics 2025; 25:e202400320. [PMID: 39790063 PMCID: PMC11962585 DOI: 10.1002/pmic.202400320] [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: 10/02/2024] [Revised: 12/11/2024] [Accepted: 12/12/2024] [Indexed: 01/12/2025]
Abstract
Cell surface proteins (surfaceome) represent key signalling and interaction molecules for therapeutic targeting, biomarker profiling and cellular phenotyping in physiological and pathological states. Here, we employed coronary artery perfusion with membrane-impermeant biotin to label and capture the surface-accessible proteome in the neo-native (intact) heart. Using quantitative proteomics, we identified 701 heart cell surfaceome accessible by the coronary artery, including receptors, cell surface enzymes, adhesion and junctional molecules. This surfaceome comprises to 216 cardiac cell-specific surface proteins, including 29 proteins reported in cardiomyocytes (CXADR, CACNA1C), 12 in cardiac fibroblasts (ITGA8, COL3A1) and 63 in multiple cardiac cell types (ICAM1, SLC3A2, CDH2). Further, this surfaceome comprises to 53 proteins enriched in heart tissue compared to other tissues in humans and implicated in cardiac cell signalling networks involving cardiomyopathy (CDH2, DTNA, PTKP2, SNTA1, CAM, K2D/B), cardiac muscle contraction and development (ENG, SNTA1, SGCG, MYPN), calcium ion binding (SGCA, MASP1, THBS4, FBLN2, GSN) and cell metabolism (SDHA, NUDFS1, GYS1, ACO2, IDH2). This method offers a powerful tool for dissecting the molecular landscape of the coronary artery accessible heart cell surfaceome, its role in maintaining cardiac and vascular function, and potential molecular leads for studying cardiac cell interactions and systemic delivery to the neo-native heart.
Collapse
Affiliation(s)
- Iasmin Inocencio
- Baker Heart and Diabetes InstituteMelbourneVictoriaAustralia
- Baker Department of Cardiovascular Research Translation and ImplementationLa Trobe UniversityMelbourneVictoriaAustralia
| | - Alin Rai
- Baker Heart and Diabetes InstituteMelbourneVictoriaAustralia
- Baker Department of Cardiovascular Research Translation and ImplementationLa Trobe UniversityMelbourneVictoriaAustralia
- Baker Department of Cardiometabolic HealthUniversity of MelbourneMelbourneVictoriaAustralia
- Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
| | - Daniel Donner
- Baker Heart and Diabetes InstituteMelbourneVictoriaAustralia
| | - David W. Greening
- Baker Heart and Diabetes InstituteMelbourneVictoriaAustralia
- Baker Department of Cardiovascular Research Translation and ImplementationLa Trobe UniversityMelbourneVictoriaAustralia
- Baker Department of Cardiometabolic HealthUniversity of MelbourneMelbourneVictoriaAustralia
- Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
| |
Collapse
|
17
|
Li Q, Yang X, Li T. Natural flavonoids from herbs and nutraceuticals as ferroptosis inhibitors in central nervous system diseases: current preclinical evidence and future perspectives. Front Pharmacol 2025; 16:1570069. [PMID: 40196367 PMCID: PMC11973303 DOI: 10.3389/fphar.2025.1570069] [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: 02/02/2025] [Accepted: 02/24/2025] [Indexed: 04/09/2025] Open
Abstract
Flavonoids are a class of important polyphenolic compounds, renowned for their antioxidant properties. However, recent studies have uncovered an additional function of these natural flavonoids: their ability to inhibit ferroptosis. Ferroptosis is a key mechanism driving cell death in central nervous system (CNS) diseases, including both acute injuries and chronic neurodegenerative disorders, characterized by iron overload-induced lipid peroxidation and dysfunction of the antioxidant defense system. This review discusses the therapeutic potential of natural flavonoids from herbs and nutraceuticals as ferroptosis inhibitors in CNS diseases, focusing on their molecular mechanisms, summarizing findings from preclinical animal models, and providing insights for clinical translation. We specifically highlight natural flavonoids such as Baicalin, Baicalein, Chrysin, Vitexin, Galangin, Quercetin, Isoquercetin, Eriodictyol, Proanthocyanidin, (-)-epigallocatechin-3-gallate, Dihydromyricetin, Soybean Isoflavones, Calycosin, Icariside II, and Safflower Yellow, which have shown promising results in animal models of acute CNS injuries, including ischemic stroke, cerebral ischemia-reperfusion injury, intracerebral hemorrhage, subarachnoid hemorrhage, traumatic brain injury, and spinal cord injury. Among these, Baicalin and its precursor Baicalein stand out due to extensive research and favorable outcomes in acute injury models. Mechanistically, these flavonoids not only regulate the Nrf2/ARE pathway and activate GPX4/GSH-related antioxidant pathways but also modulate iron metabolism proteins, thereby alleviating iron overload and inhibiting ferroptosis. While flavonoids show promise as ferroptosis inhibitors for CNS diseases, especially in acute injury settings, further studies are needed to evaluate their efficacy, safety, pharmacokinetics, and blood-brain barrier penetration for clinical application.
Collapse
Affiliation(s)
- Qiuhe Li
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaohang Yang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Tiegang Li
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
18
|
Marquard JM, Lønborg J, Obling LER, Beske RP, Zhou Y, Nepper-Christensen L, Vejlstrup N, Bang LE, Hassager C, Folke F, Andersen LB, Christensen HC, Holmvang L, Pedersen F, Ahlehoff O, Jabbari R, Minkkinen M, Sørensen R, Tilsted HH, Engstrøm T. Prehospital pulse-dose glucocorticoid on index of microvascular resistance in patients with ST-segment elevation myocardial infarction: a sub-study of the PULSE-MI trial. J Inflamm (Lond) 2025; 22:12. [PMID: 40102868 PMCID: PMC11921491 DOI: 10.1186/s12950-025-00440-2] [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: 09/26/2024] [Accepted: 03/08/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND Microvascular injury in patients with ST-segment elevation myocardial infarction (STEMI) occurs in up to 50%, yet no therapeutic target exists. Inflammation contributes directly to myocardial damage in STEMI and may also cause deleteriously effects on the microcirculation. The aim of this prespecified sub-study was to determine the effect of prehospital pulse-dose glucocorticoid on the microcirculation determined by index of microvascular resistance (IMR) and its relation to inflammation. The PULSE-MI trial was a 1:1 randomized, blinded, placebo-controlled clinical trial in patients with STEMI transferred for primary percutaneous coronary intervention (PCI) investigating the cardioprotective effects of prehospital pulse-dose glucocorticoid (methylprednisolone 250 mg) compared with placebo. In this prespecified sub-study, we investigated microvascular function as IMR by thermodilution after primary PCI and inflammation defined by C-reactive protein (CRP) at 24 hours after onset of STEMI. RESULTS Of 530 patients included in the PULSE-MI trial, 295 (56%) were assessed with coronary physiology of whom 142 (48%) were treated with glucocorticoid and 153 (52%) with placebo. Baseline characteristics were overall well-balanced in both groups. The median IMR in the glucocorticoid group was 23 (interquartile range (IQR), 11-38) and 18 (IQR, 11-42) in the placebo group (p=0.49). CRP upon arrival did not differ between treatment groups (p=0.81), but CRP at 24 hours was significantly lower in the glucocorticoid group compared to placebo (p<0.001). CONCLUSIONS Prehospital glucocorticoid did not impact IMR assessed immediately after primary PCI, albeit this compound, demonstrated significant anti-inflammatory effects as determined by CRP levels at 24 hours. TRIAL REGISTRATION http://www. CLINICALTRIALS gov ; Unique Identifier: NCT05462730.
Collapse
Affiliation(s)
- Jasmine Melissa Marquard
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Inge Lehmanns Vej 7, Copenhagen, 2100, Denmark.
| | - Jacob Lønborg
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Inge Lehmanns Vej 7, Copenhagen, 2100, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Laust Emil Roelsgaard Obling
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Inge Lehmanns Vej 7, Copenhagen, 2100, Denmark
| | - Rasmus Paulin Beske
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Inge Lehmanns Vej 7, Copenhagen, 2100, Denmark
| | - Yan Zhou
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Inge Lehmanns Vej 7, Copenhagen, 2100, Denmark
| | - Lars Nepper-Christensen
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Inge Lehmanns Vej 7, Copenhagen, 2100, Denmark
| | - Niels Vejlstrup
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Inge Lehmanns Vej 7, Copenhagen, 2100, Denmark
| | - Lia Evi Bang
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Inge Lehmanns Vej 7, Copenhagen, 2100, Denmark
| | - Christian Hassager
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Inge Lehmanns Vej 7, Copenhagen, 2100, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Fredrik Folke
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Copenhagen Emergency Medical Services, University of Copenhagen, Copenhagen, Denmark
- Department of Cardiology, Copenhagen University Hospital Herlev, Copenhagen, Denmark
| | | | - Helle Collatz Christensen
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Region Zealand Emergency Medical Services, Naestved, Denmark
| | - Lene Holmvang
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Inge Lehmanns Vej 7, Copenhagen, 2100, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Frants Pedersen
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Inge Lehmanns Vej 7, Copenhagen, 2100, Denmark
| | - Ole Ahlehoff
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Inge Lehmanns Vej 7, Copenhagen, 2100, Denmark
| | - Reza Jabbari
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Inge Lehmanns Vej 7, Copenhagen, 2100, Denmark
| | - Mikko Minkkinen
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Inge Lehmanns Vej 7, Copenhagen, 2100, Denmark
| | - Rikke Sørensen
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Inge Lehmanns Vej 7, Copenhagen, 2100, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Hans-Henrik Tilsted
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Inge Lehmanns Vej 7, Copenhagen, 2100, Denmark
| | - Thomas Engstrøm
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Inge Lehmanns Vej 7, Copenhagen, 2100, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
19
|
Evan M, Dong F. The role of long non-coding RNA in myocardial ischemia/reperfusion injury. Int J Cardiol 2025; 423:132983. [PMID: 39824290 DOI: 10.1016/j.ijcard.2025.132983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 01/08/2025] [Accepted: 01/10/2025] [Indexed: 01/20/2025]
Affiliation(s)
- Madeline Evan
- School of Medicine, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Feng Dong
- School of Medicine, Northeast Ohio Medical University, Rootstown, OH, USA.
| |
Collapse
|
20
|
Chen DX, Feng YY, Wang HY, Lu CH, Liu DZ, Gong C, Xue Y, Na N, Huang F. Metrnl ameliorates myocardial ischemia-reperfusion injury by activating AMPK-mediated M2 macrophage polarization. Mol Med 2025; 31:98. [PMID: 40082768 PMCID: PMC11907862 DOI: 10.1186/s10020-025-01150-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 03/03/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND Meteorin-like hormone (Metrnl) is prominently expressed in activated M2 macrophages and has demonstrated potential therapeutic effects in a range of cardiovascular diseases by modulating inflammatory responses. Nevertheless, its precise role and the underlying mechanisms in myocardial ischemia/reperfusion injury (MI/RI) are not fully understood. This study examined whether Metrnl can mitigate MI/RI through the AMPK-mediated polarization of M2 macrophages. METHODS In vivo, adeno-associated virus 9 containing the F4/80 promoter (AAV9-F4/80) was utilized to overexpress Metrnl in mouse cardiac macrophages before MI/RI surgery. In vitro, mouse bone marrow-derived macrophages (BMDMs) were treated with recombinant protein Metrnl, and the human cardiomyocyte cell line AC16 was subjected to hypoxia/reoxygenation (H/R) after co-culture with the supernatant of these macrophages. Cardiac function was assessed via echocardiography, H&E staining, and Evans blue-TTC staining. Inflammatory infiltration was evaluated by RT-qPCR and ELISA, apoptosis by Western blotting and TUNEL staining, and macrophage polarization by immunofluorescence staining and flow cytometry. RESULTS In vivo, Metrnl overexpression in cardiac macrophages significantly attenuated MI/RI, as evidenced by reduced myocardial infarct size, enhancement of cardiac function, diminished inflammatory cell infiltration, and decreased cardiomyocyte apoptosis. Furthermore, Metrnl overexpression promoted M1 to M2 macrophage polarization. In vitro, BMDMs treated with Metrnl shifted towards M2 polarization, characterized by decreased expression of inflammatory cytokines (IL-1β, MCP-1, TNF-α) and increased expression of the anti-inflammatory cytokine IL-10. Additionally, supernatant from Metrnl-treated macrophages protected AC16 cells from apoptosis under H/R conditions, as evidenced by decreased BAX expression and increased BCL-2 expression. However, these effects of Metrnl were inhibited by the AMPK inhibitor Compound C. CONCLUSIONS Metrnl alleviates MI/RI by activating AMPK-mediated M2 macrophage polarization to attenuate inflammatory response and cardiomyocyte apoptosis. This study highlights the therapeutic potential of Metrnl in MI/RI, and identifies it as a promising target for the treatment of ischemic heart disease.
Collapse
Affiliation(s)
- De-Xin Chen
- Department of Cardiology, Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Yang-Yi Feng
- Department of Cardiology, Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Hai-Yan Wang
- Department of Cardiology, Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Chuang-Hong Lu
- Department of Cardiology, Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - De-Zhao Liu
- Department of Cardiology, Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Chen Gong
- Department of Cardiology, Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Yan Xue
- Department of Cardiology, Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Na Na
- Department of Cardiology, Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, 530021, Guangxi, China
- Department of Neuroscience, Scripps Research Institute, No.10550 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Feng Huang
- Department of Cardiology, Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, 530021, Guangxi, China.
| |
Collapse
|
21
|
Zhao A, Lei W, Tian J, Wu X, Li M, Zhang Y, Wu X, Xu X, Tang J, Yang Y, Jin Z. Mangiferin Attenuates Myocardial Ischemia Reperfusion Injury by Regulating the GAS6/Axl Signaling Pathway. Phytother Res 2025; 39:1388-1402. [PMID: 39780746 DOI: 10.1002/ptr.8423] [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: 06/27/2024] [Revised: 11/18/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025]
Abstract
Ischemia reperfusion-induced myocardial injury is a prominent pathological feature in patients with coronary artery disease, contributing to significant mortality and morbidity rates. Mangiferin (MGF), the main active ingredient extracted from Anemarrhena asphodeloides Bge, has anti-inflammatory, anti-oxidation, anti-diabetes, and anti-tumor effects. The present study confirmed that the GAS6/Axl pathway was identified as a promising novel target for the treatment of myocardial ischemia reperfusion (IR) injury. However, whether MGF exerts anti-myocardial ischemia reperfusion injury through GAS6/Axl is still unclear. In this study, BALB/c male mice and HL-1 cardiomyocytes were used to construct a model of IR and hypoxia-reoxygenation (HR) (or H2O2) injury in vivo and in vitro, respectively. MGF significantly improved cardiac function indicators, myocardial structure, myocardial enzymes, and mitochondrial function, together with reduced oxidative stress and apoptosis in IR-injured mice. In vitro, MGF significantly increased cell viability, inhibited the release of LDH, reduced oxidative stress and apoptosis, and improved mitochondrial function in both HR and H2O2-injured HL-1 cells. In particular, the GAS6/Axl signaling pathway plays an important role in this process. Additionally, we also demonstrated that GAS6 gene knockout reversed the protective effect of MGF against HR and H2O2-injured cardiomyocytes. The present study confirmed that MGF has protective effects against myocardial IR injury by activating the GAS6/Axl pathway, providing a theoretical basis for MGF as a potential cardioprotective drug in the clinical setting of myocardial IR injury.
Collapse
Affiliation(s)
- Aizhen Zhao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Wangrui Lei
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Jiayin Tian
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Xiaopeng Wu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Northwest University First Hospital, Northwest University, Xi'an, China
| | - Mengyu Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Yan Zhang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Xue Wu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Xuezeng Xu
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jiayou Tang
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yang Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Northwest University First Hospital, Northwest University, Xi'an, China
| | - Zhenxiao Jin
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
22
|
Tong C, Zhou B. Cardioprotective strategies in myocardial ischemia-reperfusion injury: Implications for improving clinical translation. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2025; 11:100278. [PMID: 40182153 PMCID: PMC11967023 DOI: 10.1016/j.jmccpl.2024.100278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 11/11/2024] [Accepted: 12/13/2024] [Indexed: 04/05/2025]
Abstract
Ischemic heart disease is the most common cause of death and disability globally which is caused by reduced or complete cessation of blood flow to a portion of the myocardium. One of its clinical manifestations is myocardial infarction, which is commonly treated by restoring of blood flow through reperfusion therapies. However, serious ischemia-reperfusion injury (IRI) can occur, significantly undermining clinical outcomes, for which there is currently no effective therapy. This review revisits several potential pharmacological IRI intervention strategies that have entered preclinical or clinical research phases. Here, we discuss what we have learned through translational failures over the years, and propose possible ways to enhance translation efficiency.
Collapse
Affiliation(s)
- Chao Tong
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Bingying Zhou
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen 518063, China
| |
Collapse
|
23
|
Wu JW, Lan Q, Zhang DS, Jian YH, Yu L, Hao R, Liu P, Luo G, Chen MT, Liu MN. Frontiers and Hotspot Evolution of NLRP3 Inflammasome in Myocardial Infarction Research: A Bibliometric Analysis From 2013 to 2024. Cardiovasc Ther 2025; 2025:5178894. [PMID: 40018377 PMCID: PMC11865457 DOI: 10.1155/cdr/5178894] [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] [Received: 09/10/2024] [Accepted: 02/08/2025] [Indexed: 03/01/2025] Open
Abstract
The NACHT, leucine-rich repeat, and pyrin domain-containing protein 3 (NLRP3) inflammasome plays an essential role in myocardial infarction (MI) development. Up to now, no bibliometric analyses of NLRP3 in MI have been performed. Publications related to NLRP3 in MI from 1 January 2013 to 20 August 2024 were extracted from the Web of Science Core Collection (WoSCC). HistCite Pro, CiteSpace, VOSviewer, Scimago Graphica, and bibliometric online analysis program were used for bibliometric analysis and visualization. The impact of publications was assessed using the total global citation score (TGCS). A total of 324 articles (284 articles and 40 reviews) were included. China has published the most in this field, followed by the United States. Harbin Medical University was the leading institution for research related to NLRP3 in MI. Professor Abbate A. from the United States has made significant achievements in this field. International Immunopharmacology was the most active journal and Journal of Cardiovascular Pharmacology was the most cited journal. This study systematically summarizes the research results of NLRP3 in MI over the past 12 years. NLRP3 in myocardial ischemia-reperfusion injury (MIRI) will become a hot research topic, and translational research on NLRP3 inhibitors in MIRI will benefit a greater number of patients.
Collapse
Affiliation(s)
- Jin-wen Wu
- Department of Cardiovascular Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Qi Lan
- Department of Cardiovascular Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Ding-shan Zhang
- Department of Cardiovascular Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Yu-hong Jian
- Department of Cardiovascular Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Lin Yu
- Department of Orthopaedics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, Sichuan, China
| | - Rui Hao
- School of Nursing, Southwest Medical University, Luzhou, Sichuan, China
| | - Ping Liu
- Department of Cardiovascular Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Gang Luo
- Department of Cardiovascular Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Ming-tai Chen
- Department of Cardiovascular Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Meng-nan Liu
- Department of Cardiovascular Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
24
|
Zhu K, Wang K, Zhang R, Zhu Z, Wang W, Yang B, Zhao J, Shen Y. Iron chelators loaded on myocardiocyte mitochondria-targeted nanozyme system for treating myocardial ischemia-reperfusion injury in mouse models. J Nanobiotechnology 2025; 23:112. [PMID: 39955554 PMCID: PMC11829476 DOI: 10.1186/s12951-025-03197-1] [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: 12/11/2024] [Accepted: 02/02/2025] [Indexed: 02/17/2025] Open
Abstract
Ferroptosis plays a critical role in myocardial ischemia-reperfusion injury (MIRI), posing a significant clinical challenge. Nanoenzymes like cerium oxide (CeO2) hold promise for mitigating oxidative damage and inhibiting ferroptosis, but their delivery efficiency and biological activity require optimization. This study aims to develop a targeted nanozyme delivery system for MIRI treatment by integrating CeO2 with mesoporous polydopamine (mPDA) and dexrazoxane (DXZ) to achieve synergistic therapeutic effects. A biomineralization technique was used to synthesize CeO2 nanoparticles (2-3 nm) within mPDA, forming ~ 130 nm composite nanoparticles (Ce@mPDA). Surface modifications with cardiac homing peptide (CHP) and triphenylphosphine (TPP) enabled hierarchical targeting to injured myocardium and mitochondria. DXZ-loaded Ce@mPDA-C/P nanoparticles (D/Ce@mPDA-C/P) were evaluated in vitro and in a MIRI mouse model for their effects on oxidative stress, ferroptosis, apoptosis, inflammation, and cardiac function. D/Ce@mPDA-C/P nanoparticles exhibited robust ROS scavenging, sustained DXZ release, and efficient myocardial and mitochondrial targeting. The D/Ce@mPDA-C/P system significantly reduced oxidative stress, upregulated GPX4 expression, inhibited ferroptosis, and modulated the inflammatory microenvironment. Long-term studies in a MIRI mouse model demonstrated reductions in myocardial fibrosis and improvements in cardiac function, including enhanced fractional shortening and ejection fraction. This hierarchical targeting delivery system effectively combines the antioxidant properties of CeO2 with the iron-chelating effects of DXZ, providing a promising therapeutic strategy for MIRI. This approach may expand the clinical use of DXZ and advance nanomedicine-based interventions for myocardial repair.
Collapse
Affiliation(s)
- Ke Zhu
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Kun Wang
- Department of Nuclear Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Rongting Zhang
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Ziyang Zhu
- Department of Nuclear Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Wenyuan Wang
- Department of Ultrasound, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Biao Yang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jun Zhao
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
- Department of Nuclear Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| | - Yunli Shen
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| |
Collapse
|
25
|
Zheng X, Zheng X, Zhang C, Liu M. Geriatric nutritional risk index as a predictor of 30-day and 365-day mortality in patients with acute myocardial infarction: a retrospective cohort study using the MIMIC-IV database. Front Nutr 2025; 12:1544382. [PMID: 39973920 PMCID: PMC11835668 DOI: 10.3389/fnut.2025.1544382] [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: 12/12/2024] [Accepted: 01/24/2025] [Indexed: 02/21/2025] Open
Abstract
Background The Geriatric Nutritional Risk Index (GNRI) is a clinical indicator for evaluating the nutritional status of patients, but its role in the short-term prognosis of patients with acute myocardial infarction is still not fully understood. This study aims to explore the correlation between the GNRI and the overall mortality within 30 days and 365 days in those with acute myocardial infarction (AMI). Methods A retrospective analysis was performed utilizing the Medical Information Mart for Intensive Care IV (MIMIC-IV) database. The study included 895 patients diagnosed with AMI and identified through ICD9 and ICD10 codes (410, I21, I23) who were hospitalized for the first time due to AMI. Subjects were classified into four groups according to GNRI: high (GNRI <82, n = 110), moderate (82 ≤ GNRI <92, n = 205), low (92 ≤ GNRI <98, n = 225), and no nutritional risk (GNRI ≥98, n = 355). Restricted cubic splines (RCS) and threshold effect analyses were applied to explore the non-linear relationship between GNRI and mortality. Subgroup analyses were conducted based on gender, hypertension, diabetes, stroke, hyperlipidemia, chronic obstructive pulmonary disease, and age. A mediation study was conducted to investigate the impact of lymphocytes on the association between GNRI and mortality. Results In an overall sample of 895 patients, an elevated GNRI correlated with reduced 30-day (HR = 0.937, 95% CI: 0.917-0.957, p < 0.001) and 365-day mortality (HR = 0.937, 95% CI: 0.923-0.950, p < 0.001). The trend analysis for GNRI categories indicated a significant decline in mortality associated with rising GNRI (P for trend <0.001). Subgroup analysis validated the consistency of such results throughout diverse patient characteristics. The lymphocytes significantly mediated the relationship between GNRI and 30-day mortality (ACME: 0.022, 95% CI: 0.003-0.180, p < 0.001). A landmark analysis at 20 days after admission further demonstrated the impact of GNRI on mortality during different phases of recovery. Conclusion This study highlights the prognostic value of GNRI in predicting short-term and long-term mortality in AMI patients, emphasizing the significance of nutritional status and inflammatory indicators in the therapy and risk assessment of these individuals.
Collapse
Affiliation(s)
- Xiaolong Zheng
- Emergency Department, The First Hospital Affiliated to Army Medical University, Chongqing, China
- Department of Orthopedics, the 963rd Hospital of the Joint Service Support Force of the PLA, Jiamusi, China
| | - Xin Zheng
- Emergency Department, The First Hospital Affiliated to Army Medical University, Chongqing, China
- Emergency Department, the 963rd Hospital of the Joint Service Support Force of the PLA, Jiamusi, China
| | - Changgui Zhang
- Emergency Department, The First Hospital Affiliated to Army Medical University, Chongqing, China
| | - Minghua Liu
- Emergency Department, The First Hospital Affiliated to Army Medical University, Chongqing, China
| |
Collapse
|
26
|
Xu H, Chen X, Luo S, Jiang J, Pan X, He Y, Deng B, Liu S, Wan R, Lin L, Tan Q, Chen X, Yao Y, He B, An Y, Li J. Cardiomyocyte-specific Piezo1 deficiency mitigates ischemia-reperfusion injury by preserving mitochondrial homeostasis. Redox Biol 2025; 79:103471. [PMID: 39740362 PMCID: PMC11750285 DOI: 10.1016/j.redox.2024.103471] [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: 11/17/2024] [Accepted: 12/15/2024] [Indexed: 01/02/2025] Open
Abstract
Ca2+ overload and mitochondrial dysfunction play crucial roles in myocardial ischemia-reperfusion (I/R) injury. Piezo1, a mechanosensitive cation channel, is essential for intracellular Ca2+ homeostasis. The objective of this research was to explore the effects of Piezo1 on mitochondrial function during myocardial I/R injury. We showed that the expression of myocardial Piezo1 was elevated in the infracted area of I/R and cardiomyocyte-specific Piezo1 deficiency (Piezo1△Myh6) mice attenuated I/R by decreasing infarct size and cardiac dysfunction. Piezo1△Myh6 regulated mitochondrial fusion and fission to improve mitochondrial function and decrease inflammation and oxidative stress in vivo and in vitro. Mechanistically, myocardial Piezo1 knockout alleviated intracellular calcium overload to normalize calpain-associated mitochondrial homeostasis. Our findings indicated that Piezo1 depletion in cardiomyocytes partially restored mitochondrial homeostasis during cardiac ischemia/reperfusion (I/R) injury. This study suggests an innovative therapeutic strategy to alleviate cardiac I/R injury.
Collapse
Affiliation(s)
- Honglin Xu
- Innovation Research Center, Shandong University of Traditional Chinese Medicine, Jinan, 250307, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Xin Chen
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Shangfei Luo
- Innovation Research Center, Shandong University of Traditional Chinese Medicine, Jinan, 250307, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Jintao Jiang
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Xianmei Pan
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Yu He
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710049, China
| | - Bo Deng
- The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510260, China
| | - Silin Liu
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Rentao Wan
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Liwen Lin
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Qiaorui Tan
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Xiaoting Chen
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Youfen Yao
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Bin He
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Yajuan An
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Jing Li
- Innovation Research Center, Shandong University of Traditional Chinese Medicine, Jinan, 250307, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, LS2 9JT, UK.
| |
Collapse
|
27
|
Bo X, Li Q, Chen S, Zhou T, Yin N, Song W, Zhao D, Liu J, Fan Q. Evidence and perspectives on miRNA, circRNA, and lncRNA in myocardial ischemia-reperfusion injury: a bibliometric study. J Cardiothorac Surg 2025; 20:66. [PMID: 39815292 PMCID: PMC11736979 DOI: 10.1186/s13019-024-03238-0] [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: 09/25/2024] [Accepted: 12/24/2024] [Indexed: 01/18/2025] Open
Abstract
OBJECTIVE miRNA, circRNA, and lncRNA play crucial roles in the pathogenesis and progression of myocardial ischemia-reperfusion injury (MI/RI). This study aims to provide valuable insights into miRNA, circRNA, lncRNA, and MI/RI from a bibliometric standpoint, with the goal of fostering further advancements in this area. METHODS The relevant literature in the field of miRNA, circRNA, lncRNA, and MI/RI was retrieved from the Science Citation Index Expanded (SCI-E) database within Web of Science. The "Analyze Results" and "Citation Report" functions in WOS were utilized to compile the annual publication and citation counts in this field. Microsoft Office Excel 2019 was used to organize and visualize the data. Furthermore, bibliometric and visualization analyses of countries/regions, institutions, authors, keywords, and references were conducted using the bibliometric visualization software CiteSpace. RESULTS A total of 858 publications were included for further analysis in this field. The literature was published across 297 journals, with Molecular Medicine Reports contributing the highest number of publications. Researchers from 45 countries participated in studies within this field, with those from China contributing the most publications. The research hotspots in this field primarily focus on three areas: the role of miRNA, circRNA, and lncRNA in the pathogenesis of MI/RI, their potential as therapeutic targets, and their role as biomarkers. Among these, circular RNA, therapy target, inflammatory response, and cardiomyocyte ferroptosis are likely to emerge as emerging trends in this field. CONCLUSION The overall development of research in this field is on the rise. The compilation of research hotspots and emerging trends in this area may provide researchers with more references and assistance in selecting research directions, ultimately benefiting MI/RI patients.
Collapse
Affiliation(s)
- Xiaowen Bo
- Department of Cardiology, Beijing Anzhen Hospital, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Capital Medical University, Beijing, 100069, China
| | - Qiuyu Li
- Department of Cardiology, Beijing Anzhen Hospital, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Capital Medical University, Beijing, 100069, China
| | - Siyuan Chen
- Department of Cardiology, Beijing Anzhen Hospital, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Capital Medical University, Beijing, 100069, China
| | - Tian Zhou
- Department of Cardiology, Beijing Anzhen Hospital, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Capital Medical University, Beijing, 100069, China
| | - Ning Yin
- Department of Cardiology, Beijing Anzhen Hospital, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Capital Medical University, Beijing, 100069, China
| | - Wenpeng Song
- Department of Stomatology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Donghui Zhao
- Department of Cardiology, Beijing Anzhen Hospital, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Capital Medical University, Beijing, 100069, China
| | - Jinghua Liu
- Department of Cardiology, Beijing Anzhen Hospital, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Capital Medical University, Beijing, 100069, China
| | - Qian Fan
- Department of Cardiology, Beijing Anzhen Hospital, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
28
|
Hu C, Francisco J, Del Re DP, Sadoshima J. Decoding the Impact of the Hippo Pathway on Different Cell Types in Heart Failure. Circ J 2024; 89:6-15. [PMID: 38644191 DOI: 10.1253/circj.cj-24-0171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
The evolutionarily conserved Hippo pathway plays a pivotal role in governing a variety of biological processes. Heart failure (HF) is a major global health problem with a significant risk of mortality. This review provides a contemporary understanding of the Hippo pathway in regulating different cell types during HF. Through a systematic analysis of each component's regulatory mechanisms within the Hippo pathway, we elucidate their specific effects on cardiomyocytes, fibroblasts, endothelial cells, and macrophages in response to various cardiac injuries. Insights gleaned from both in vitro and in vivo studies highlight the therapeutic promise of targeting the Hippo pathway to address cardiovascular diseases, particularly HF.
Collapse
Affiliation(s)
- Chengchen Hu
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School
| | - Jamie Francisco
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School
| | - Dominic P Del Re
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School
| |
Collapse
|
29
|
Jose A, Fernando JJ, Kienesberger PC. Lysophosphatidic acid metabolism and signaling in heart disease. Can J Physiol Pharmacol 2024; 102:685-696. [PMID: 38968609 DOI: 10.1139/cjpp-2024-0077] [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] [Indexed: 07/07/2024]
Abstract
Lysophosphatidic acid (LPA) is a bioactive lipid that is mainly produced by the secreted lysophospholipase D, autotaxin (ATX), and signals through at least six G protein-coupled receptors (LPA1-6). Extracellular LPA is degraded through lipid phosphate phosphatases (LPP1, LPP2, and LPP3) at the plasmamembrane, terminating LPA receptor signaling. The ATX-LPA-LPP3 pathway is critically involved in a wide range of physiological processes, including cell survival, migration, proliferation, angiogenesis, and organismal development. Similarly, dysregulation of this pathway has been linked to many pathological processes, including cardiovascular disease. This review summarizes and interprets current literature examining the regulation and role of the ATX-LPA-LPP3 axis in heart disease. Specifically, the contribution of altered LPA metabolism via ATX and LPP3 and resulting changes to LPA receptor signaling in obesity cardiomyopathy, cardiac mitochondrial dysfunction, myocardial infarction/ischemia-reperfusion injury, hypertrophic cardiomyopathy, and aortic valve stenosis is discussed.
Collapse
Affiliation(s)
- Anu Jose
- Department of Biochemistry and Molecular Biology, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, NB, Canada
| | - Jeffy J Fernando
- Department of Biochemistry and Molecular Biology, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, NB, Canada
| | - Petra C Kienesberger
- Department of Biochemistry and Molecular Biology, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, NB, Canada
| |
Collapse
|
30
|
Johnson CF, Schafer CM, Burge KY, Coon BG, Chaaban H, Griffin CT. Endothelial RIPK3 minimizes organotypic inflammation and vascular permeability in ischemia-reperfusion injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.25.625188. [PMID: 39651150 PMCID: PMC11623548 DOI: 10.1101/2024.11.25.625188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Recent studies have revealed a link between endothelial receptor-interacting protein kinase 3 (RIPK3) and vascular integrity. During mouse embryonic development, hypoxia can trigger elevated endothelial RIPK3 that contributes to lethal vascular rupture. However, it is unknown whether RIPK3 regulate endothelial barrier function in adult vasculature under hypoxic injury conditions such as ischemia-reperfusion (I/R) injury. Here we performed inducible genetic deletion of endothelial Ripk3 ( Ripk iECKO ) in mice, which led to elevated vascular permeability in the small intestine and multiple distal organs after intestinal I/R injury. Mechanistically, this vascular permeability correlated with increased endothelial secretion of IL-6 and organ-specific expression of VCAM-1 and ICAM-1 adhesion molecules. Circulating monocyte depletion with clodronate liposomes reduced permeability in organs with elevated adhesion molecules, highlighting the contribution of monocyte adhesion and extravasation to Ripk iECKO barrier dysfunction. These results elucidate mechanisms by which RIPK3 regulates endothelial inflammation to minimize vascular permeability in I/R injury. GRAPHICAL ABSTRACT
Collapse
|
31
|
Chen X, Yang Y, Zhou Z, Yu H, Zhang S, Huang S, Wei Z, Ren K, Jin Y. Unraveling the complex interplay between Mitochondria-Associated Membranes (MAMs) and cardiovascular Inflammation: Molecular mechanisms and therapeutic implications. Int Immunopharmacol 2024; 141:112930. [PMID: 39146786 DOI: 10.1016/j.intimp.2024.112930] [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: 07/04/2024] [Revised: 07/26/2024] [Accepted: 08/10/2024] [Indexed: 08/17/2024]
Abstract
Cardiovascular diseases (CVDs) represent a significant public health concern because of their associations with inflammation, oxidative stress, and abnormal remodeling of the heart and blood vessels. In this review, we discuss the intricate interplay between mitochondria-associated membranes (MAMs) and cardiovascular inflammation, highlighting their role in key cellular processes such as calcium homeostasis, lipid metabolism, oxidative stress management, and ERS. We explored how these functions impact the pathogenesis and progression of various CVDs, including myocardial ischemia-reperfusion injury, atherosclerosis, diabetic cardiomyopathy, cardiovascular aging, heart failure, and pulmonary hypertension. Additionally, we examined current therapeutic strategies targeting MAM-related pathways and proteins, emphasizing the potential of MAMs as therapeutic targets. Our review aims to provide new insights into the mechanisms of cardiovascular inflammation and propose novel therapeutic approaches to improve cardiovascular health outcomes.
Collapse
Affiliation(s)
- Xing Chen
- Department of Translational Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Yang Yang
- Department of Translational Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Zheng Zhou
- Department of Geriatric Endocrinology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Haihan Yu
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Shuwei Zhang
- Department of Translational Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Siyuan Huang
- Department of Translational Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Ziqing Wei
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China.
| | - Kaidi Ren
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China.
| | - Yage Jin
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China.
| |
Collapse
|
32
|
Han R, Huang H, Zhu J, Jin X, Wang Y, Xu Y, Xia Z. Adipokines and their potential impacts on susceptibility to myocardial ischemia/reperfusion injury in diabetes. Lipids Health Dis 2024; 23:372. [PMID: 39538244 PMCID: PMC11558907 DOI: 10.1186/s12944-024-02357-w] [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: 07/21/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
Coronary artery disease has a high mortality rate and is a striking public health concern, affecting a substantial portion of the global population. On the early onset of myocardial ischemia, thrombolytic therapy and coronary revascularization could promptly restore the bloodstream and nutrient supply to the ischemic tissue, efficiently preserving less severely injured myocardium. However, the abrupt re-establishment of blood flow triggers the significant discharge of previously accumulated oxidative substances and inflammatory cytokines, leading to further harm referred to as ischemia/reperfusion (I/R) injury. Diabetes significantly raises the vulnerability of the heart to I/R injury due to disrupted glucose and lipid processing, impaired insulin sensitivity and metabolic signaling, and increased inflammatory responses. Numerous studies have indicated that adipokines are crucial in the etiology and pathogenesis of obesity, diabetes, hyperlipidemia, hypertension, and coronary artery disease. Adipokines such as adiponectin, adipsin, visfatin, chemerin, omentin, and apelin, which possess protective properties against inflammatory activity and insulin resistance, have been shown to confer myocardial protection in conditions such as atherosclerosis, myocardial hypertrophy, myocardial I/R injury, and diabetic complications. On the other hand, adipokines such as leptin and resistin, known for their pro-inflammatory characteristics, have been linked to elevated cardiac lipid deposition, insulin resistance, and fibrosis. Meteorin-like (metrnl) exhibits opposite effects in various pathological conditions. However, the data on adipokines in myocardial I/R, especially in diabetes, is still incomplete and controversial. This review focuses on recent research regarding the categorization and function of adipokines in the heart muscle, and the identification of different signaling pathways involved in myocardial I/R injury under diabetic conditions, aiming to facilitate the exploration of therapeutic strategies against myocardial I/R injury in diabetes.
Collapse
Affiliation(s)
- Ronghui Han
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao, PR, China
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, China
| | - Hemeng Huang
- Department of Emergency, Affiliated Hospital of Guangdong Medical University, Guangdong, China
| | - Jianyu Zhu
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, China
| | - Xiaogao Jin
- Department of Anesthesiology, The Second Affiliated Hospital of Guangdong Medical University, Guangdong, China
| | - Yongyan Wang
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao, PR, China
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, China
| | - Youhua Xu
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao, PR, China.
- Macau University of Science and Technology Zhuhai MUST Science and Technology Research Institute, Hengqin, Zhuhai, People's Republic of China.
- Faculty of Pharmacy, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao, People's Republic of China.
| | - Zhengyuan Xia
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao, PR, China.
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, China.
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
33
|
Qiu ZY, Shi KN, Li HH, Zhang B. CBR-470-1 protects against cardiomyocyte death in ischaemia/reperfusion injury by activating the Nrf2-GPX4 cascade. Toxicol Appl Pharmacol 2024; 492:117113. [PMID: 39343043 DOI: 10.1016/j.taap.2024.117113] [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: 07/16/2024] [Revised: 09/14/2024] [Accepted: 09/25/2024] [Indexed: 10/01/2024]
Abstract
Cardiac ischaemia/reperfusion (I/R) impairs mitochondrial function, resulting in excessive oxidative stress and cardiomyocyte ferroptosis and death. Nuclear factor E2-related factor 2 (Nrf2) is a key regulator of redox homeostasis and has cardioprotective effects against various stresses. Here, we tested whether CBR-470-1, a noncovalent Nrf2 activator, can protect against cardiomyocyte death caused by I/R stress. Compared with vehicle treatment, the administration of CBR-470-1 (2 mg/kg) to mice significantly increased Nrf2 protein levels and ameliorated the infarct size, the I/R-induced decrease in cardiac contractile performance, and the I/R-induced increases in cell apoptosis, ROS levels, and inflammation. Consistently, the beneficial effects of CBR-470-1 on cardiomyocytes were verified in a hypoxia/reoxygenation (H/R) model in vitro, but this cardioprotection was dramatically attenuated by the GPX4 inhibitor RSL3. Mechanistically, CBR-470-1 upregulated Nrf2 expression, which increased the expression levels of antioxidant enzymes (NQO1, SOD1, Prdx1, and Gclc) and antiferroptotic proteins (SLC7A11 and GPX4) and downregulated the protein expression of p53 and Nlrp3, leading to the inhibition of ROS production and inflammation and subsequent cardiomyocyte death (apoptosis, ferroptosis and pyroptosis). In summary, CBR-470-1 prevented I/R-mediated cardiac injury possibly through inhibiting cardiomyocyte apoptosis, ferroptosis and pyroptosis via Nrf2-mediated inhibition of p53 and Nlrp3 and activation of the SLC7A11/GPX4 pathway. Our data also highlight that CBR-470-1 may serve as a valuable agent for treating ischaemic heart disease.
Collapse
Affiliation(s)
- Ze-Yang Qiu
- Department of Cardiology, First Affiliated Hospital, Dalian Medical University, Dalian, Liaoning 116011, China
| | - Kai-Na Shi
- Department of Emergency Medicine, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Hui-Hua Li
- Department of Emergency Medicine, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China.
| | - Bo Zhang
- Department of Cardiology, First Affiliated Hospital, Dalian Medical University, Dalian, Liaoning 116011, China.
| |
Collapse
|
34
|
Yan R, Yang H, Jiang X, Lai X. Renal Protective Effect of Umbelliferone on Acute Kidney Injury in Rats via Alteration of HO-1/Nrf2 and NF-κB Signaling Pathway. DOKL BIOCHEM BIOPHYS 2024; 518:442-451. [PMID: 39196533 DOI: 10.1134/s160767292460043x] [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: 05/10/2024] [Revised: 05/30/2024] [Accepted: 05/30/2024] [Indexed: 08/29/2024]
Abstract
Acute kidney injury (AKI), formerly known as acute renal failure, refers to a sudden and often reversible decline in kidney function. Inflammatory reaction and oxidative stress play a crucial role in the expansion of renal disease. In this experimental study, we scrutinized the renal protective effect of umbelliferone against gentamicin induced renal injury in the rats and explore the mechanism. Wistar rats were used in this study and Gentamicin was used for the induction the AKI in the rats and rats were received the oral administration of umbelliferone. The body weight, organ weight, renal, oxidative stress, cytokines, inflammatory parameters were estimated. The mRNA expression caspase-3, Bax, Bcl-2, TNF-α, IL-1β, IL-6, IL-10, HO-1, and Nrf2 were estimated. Umbelliferone remarkably improved the body weight and altered the absolute and relative weight of hepatic and renal tissue. Umbelliferone significantly suppressed the level of BUN, Scr, magnesium, calcium, phosphorus, sodium, and potassium along with altered the level of oxidative stress parameters like CAT, SOD, GSH, LPO, and GPx. Umbelliferone altered the level of cytokines viz., TNF-α, Il-1β, IL-6, IL-10; inflammatory parameters like PGE2, COX-2, TGF-β, NF-κB, respectively. Umbelliferone significantly altered the mRNA expression of caspase-3, Bax, Bcl-2, TNF-α, IL-1β, IL-6, IL-10, HO-1, and Nrf2. The result showed the renal protective effect of umbelliferone against gentamycin induced renal disease via alteration of HO-1/Nrf2 and NF-κB Signaling Pathway.
Collapse
Affiliation(s)
- RuiJuan Yan
- Department of Emergency Medicine, Shandong Provincial Third Hospital, Shandong University, 250031, Jinan, China
| | - Hui Yang
- Department of Respiratory, Qingdao, Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Hospital), 266042, Qingdao, China
| | - XiaoQi Jiang
- Department of Respiratory, Qingdao, Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Hospital), 266042, Qingdao, China
| | - XiaoDong Lai
- Department of Urology Surgery, Qingdao, Qingdao Traditional Chinese Medicine Hospital (Qingdao Hiser Hospital), 266000, Qingdao, China.
| |
Collapse
|
35
|
Guo Z, Tian Y, Liu N, Chen Y, Chen X, Yuan G, Chang A, Chang X, Wu J, Zhou H. Mitochondrial Stress as a Central Player in the Pathogenesis of Hypoxia-Related Myocardial Dysfunction: New Insights. Int J Med Sci 2024; 21:2502-2509. [PMID: 39439461 PMCID: PMC11492880 DOI: 10.7150/ijms.99359] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 09/13/2024] [Indexed: 10/25/2024] Open
Abstract
Hypoxic injury is a critical pathological factor in the development of various cardiovascular diseases, such as congenital heart disease, myocardial infarction, and heart failure. Mitochondrial quality control is essential for protecting cardiomyocytes from hypoxic damage. Under hypoxic conditions, disruptions in mitochondrial homeostasis result in excessive reactive oxygen species (ROS) production, imbalances in mitochondrial dynamics, and initiate pathological processes including oxidative stress, inflammatory responses, and apoptosis. Targeted interventions to enhance mitochondrial quality control, such as coenzyme Q10 and statins, have shown promise in mitigating hypoxia-induced mitochondrial dysfunction. These treatments offer potential therapeutic strategies for hypoxia-related cardiovascular diseases by regulating mitochondrial fission and fusion, restoring mitochondrial biogenesis, reducing ROS production, and promoting mitophagy.
Collapse
Affiliation(s)
- Zhijiang Guo
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Yingjie Tian
- Beijing University of Chinese Medicine, Beijing, 100028, China
| | - Nanyang Liu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ye Chen
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaohan Chen
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Guoxing Yuan
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - An Chang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Xing Chang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Jie Wu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Hao Zhou
- Senior Department of Cardiology, The Sixth Medical Center of People's Liberation Army General Hospital, Beijing, China
| |
Collapse
|
36
|
Kang M, Jia H, Feng M, Ren H, Gao J, Liu Y, Zhang L, Zhou MS. Cardiac macrophages in maintaining heart homeostasis and regulating ventricular remodeling of heart diseases. Front Immunol 2024; 15:1467089. [PMID: 39372400 PMCID: PMC11449765 DOI: 10.3389/fimmu.2024.1467089] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 09/03/2024] [Indexed: 10/08/2024] Open
Abstract
Macrophages are most important immune cell population in the heart. Cardiac macrophages have broad-spectrum and heterogeneity, with two extreme polarization phenotypes: M1 pro-inflammatory macrophages (CCR2-ly6Chi) and M2 anti-inflammatory macrophages (CCR2-ly6Clo). Cardiac macrophages can reshape their polarization states or phenotypes to adapt to their surrounding microenvironment by altering metabolic reprogramming. The phenotypes and polarization states of cardiac macrophages can be defined by specific signature markers on the cell surface, including tumor necrosis factor α, interleukin (IL)-1β, inducible nitric oxide synthase (iNOS), C-C chemokine receptor type (CCR)2, IL-4 and arginase (Arg)1, among them, CCR2+/- is one of most important markers which is used to distinguish between resident and non-resident cardiac macrophage as well as macrophage polarization states. Dedicated balance between M1 and M2 cardiac macrophages are crucial for maintaining heart development and cardiac functional and electric homeostasis, and imbalance between macrophage phenotypes may result in heart ventricular remodeling and various heart diseases. The therapy aiming at specific target on macrophage phenotype is a promising strategy for treatment of heart diseases. In this article, we comprehensively review cardiac macrophage phenotype, metabolic reprogramming, and their role in maintaining heart health and mediating ventricular remodeling and potential therapeutic strategy in heart diseases.
Collapse
Affiliation(s)
- Mengjie Kang
- Science and Experiment Research Center, Shenyang Medical College & Shenyang Key Laboratory of Vascular Biology, Science and Experimental Research Center, Shenyang Medical College, Shenyang, China
| | - Hui Jia
- Science and Experiment Research Center, Shenyang Medical College & Shenyang Key Laboratory of Vascular Biology, Science and Experimental Research Center, Shenyang Medical College, Shenyang, China
- School of Traditional Chinese Medicine, Shenyang Medical College, Shenyang, China
| | - Mei Feng
- Science and Experiment Research Center, Shenyang Medical College & Shenyang Key Laboratory of Vascular Biology, Science and Experimental Research Center, Shenyang Medical College, Shenyang, China
| | - Haolin Ren
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Junjia Gao
- Department of Cardiology, Second Affiliated Hospital, Shenyang Medical College, Shenyang, China
| | - Yueyang Liu
- Science and Experiment Research Center, Shenyang Medical College & Shenyang Key Laboratory of Vascular Biology, Science and Experimental Research Center, Shenyang Medical College, Shenyang, China
- School of Pharmacy, Shenyang Medical College, Shenyang, China
| | - Lu Zhang
- Science and Experiment Research Center, Shenyang Medical College & Shenyang Key Laboratory of Vascular Biology, Science and Experimental Research Center, Shenyang Medical College, Shenyang, China
| | - Ming-Sheng Zhou
- Science and Experiment Research Center, Shenyang Medical College & Shenyang Key Laboratory of Vascular Biology, Science and Experimental Research Center, Shenyang Medical College, Shenyang, China
| |
Collapse
|
37
|
Cao Y, Fan R, Zhu K, Gao Y. Advances in Functionalized Hydrogels in the Treatment of Myocardial Infarction and Drug-Delivery Strategies. ACS APPLIED MATERIALS & INTERFACES 2024; 16:48880-48894. [PMID: 39227344 DOI: 10.1021/acsami.4c09623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Myocardial infarction (MI) is a serious cardiovascular disease with high morbidity and mortality rates, posing a significant threat to patient's health and quality of life. Following a MI, the damaged myocardial tissue is typically not fully repaired, leading to permanent impairment of myocardial function. While traditional treatments can alleviate symptoms and reduce pain, their ability to repair damaged heart muscle tissue is limited. Functionalized hydrogels, a broad category of materials with diverse functionalities, can enhance the properties of hydrogels to cater to the needs of tissue engineering, drug delivery, medical dressings, and other applications. Recently, functionalized hydrogels have emerged as a promising new therapeutic approach for the treatment of MI. Functionalized hydrogels possess outstanding biocompatibility, customizable mechanical properties, and drug-release capabilities. These properties enable them to offer scaffold support, drug release, and tissue regeneration promotion, making them a promising approach for treating MI. This paper aims to evaluate the advancements and delivery methods of functionalized hydrogels for treating MI, while also discussing their potential and the challenges they may pose for future clinical use.
Collapse
Affiliation(s)
- Yuchen Cao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Rong Fan
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Kaiyi Zhu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
- Shanxi Academy of Advanced Research and Innovation, Taiyuan 030032, China
| | - Yuping Gao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Taiyuan 030032, China
- Key Laboratory of Cellular Physiology, Shanxi Province, Taiyuan 030032, China
| |
Collapse
|
38
|
Tan Z, Li Y, Wu Y, Yang H, Zhang H, Liu Z, Cheng Y, Wu P. Chemical components with biological activities in the roots of Ilex pubescens. Fitoterapia 2024; 177:106076. [PMID: 38897247 DOI: 10.1016/j.fitote.2024.106076] [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: 03/10/2024] [Revised: 06/06/2024] [Accepted: 06/16/2024] [Indexed: 06/21/2024]
Abstract
Two new triterpenoids, ilexsaponin U (1) and ilexsaponin V (2), and three new phenylpropanoids, pubescenoside S (3), pubescenoside T (38), and pubescenoside U (39), along with thirty-four existing compounds were isolated from the roots of Ilex pubescens. The elucidation of their structures involved comprehensive spectroscopic techniques, including IR, UV, HR-ESI-MS, and NMR experiments. The anti-inflammatory effects of almost all the compounds were evaluated in LPS-induced RAW264.7 cells. Among these, compounds 1, 4, 8, 11, 12, 26, 27, 29 and 33 exhibited varying degrees of inhibition of inflammatory factors. Notably, compounds 1, 4 and 8 significantly inhibited the mRNA levels of iNOS, IL-6, IL-1β and TNFα, comparable to or exceeding the effect of the positive control (dexamethasone, DEX). We also evaluated the cardioprotective effects of these compounds in OGD/R-induced H9c2 cells. The results revealed that compounds 2, 3, 7, 8, 26, 35, 36 and 37 at 20 μM significantly increased cell viability by 24.9 ± 3.4%, 28.0 ± 0.3%, 37.6 ± 0.2%, 44.86 ± 0.5%, 9.47 ± 2.1%, 23.9 ± 0.4%, 39.5 ± 3.1% and 28.2 ± 0.1%, respectively. Some of them exhibited effects equal to or greater than that of the positive control (diazoxide, DZ) at 100 μM, showing a 21.9 ± 3.0% increase.
Collapse
Affiliation(s)
- Zihao Tan
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yongkang Li
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuhong Wu
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Han Yang
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hongli Zhang
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhongqiu Liu
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuanyuan Cheng
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China..
| | - Peng Wu
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China..
| |
Collapse
|
39
|
Tibenda JJ, Wang N, Li N, Dang Y, Zhu Y, Wang X, Zhang Z, Zhao Q. Research progress of circular RNAs in myocardial ischemia. Life Sci 2024; 352:122809. [PMID: 38908786 DOI: 10.1016/j.lfs.2024.122809] [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: 02/08/2024] [Revised: 05/18/2024] [Accepted: 06/04/2024] [Indexed: 06/24/2024]
Abstract
Circular RNAs (circRNAs) are a type of single-stranded RNA that forms a covalently closed continuous loop. Its structure, stability, properties, and cell- and tissue-specificity have gained considerable recognition in the research and clinical sectors, as its role has been observed in different diseases, such as cardiovascular diseases, cancers, and central nervous system diseases, etc. Cardiovascular disease is still named as the number one cause of death globally, with myocardial ischemia (MI) accounting for 15 % of mortality annually. A number of circRNAs have been identified and are being studied for their ability to reduce MI by inhibiting the molecular mechanisms associated with myocardial ischemia reperfusion injury, such as inflammation, oxidative stress, autophagy, apoptosis, and so on. CircRNAs play a significant role as crucial regulatory elements at transcriptional levels, regulating different proteins, and at posttranscriptional levels, having interactions with RNA-binding proteins, ribosomal proteins, micro-RNAS, and long non-coding RNAS, making it possible to exert their effects through the circRNA-miRNA-mRNA axis. CircRNAs are a potential novel biomarker and therapeutic target for myocardial ischemia and cardiovascular diseases in general. The purpose of this review is to summarize the relationship, function, and mechanism observed between circRNAs and MI injury, as well as to provide directions for future research and clinical trials.
Collapse
Affiliation(s)
- Jonnea Japhet Tibenda
- School of Pharmacy, Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, Ningxia, China
| | - Niuniu Wang
- School of Pharmacy, Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, Ningxia, China
| | - Nuan Li
- School of Pharmacy, Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, Ningxia, China
| | - Yanning Dang
- School of Pharmacy, Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, Ningxia, China
| | - Yafei Zhu
- School of Pharmacy, Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, Ningxia, China
| | - Xiaobo Wang
- Innovative Institute of Chinese Medicine and Pharmacy/Academy for Interdiscipline, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Zhengjun Zhang
- Department of Cardiology, General Hospital of Ningxia Medical University, Ningxia, China.
| | - Qipeng Zhao
- School of Pharmacy, Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, Ningxia, China.
| |
Collapse
|
40
|
Slotabec L, Seale B, Wang H, Wen C, Filho F, Rouhi N, Adenawoola MI, Li J. Platelets at the intersection of inflammation and coagulation in the APC-mediated response to myocardial ischemia/reperfusion injury. FASEB J 2024; 38:e23890. [PMID: 39143722 PMCID: PMC11373610 DOI: 10.1096/fj.202401128r] [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: 05/21/2024] [Revised: 07/23/2024] [Accepted: 08/05/2024] [Indexed: 08/16/2024]
Abstract
Thromboinflammation is a complex pathology associated with inflammation and coagulation. In cases of cardiovascular disease, in particular ischemia-reperfusion injury, thromboinflammation is a common complication. Increased understanding of thromboinflammation depends on an improved concept of the mechanisms of cells and proteins at the axis of coagulation and inflammation. Among these elements are activated protein C and platelets. This review summarizes the complex interactions of activated protein C and platelets regulating thromboinflammation in cardiovascular disease. By unraveling the pathways of platelets and APC in the inflammatory and coagulation cascades, this review summarizes the role of these vital mediators in the development and perpetuation of heart disease and the thromboinflammation-driven complications of cardiovascular disease. Furthermore, this review emphasizes the significance of the counteracting effects of platelets and APC and their combined role in disease states.
Collapse
Affiliation(s)
- Lily Slotabec
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
- G.V. (Sonny) Montgomery VA Medical Center, Jackson, Mississippi, USA
| | - Blaise Seale
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Hao Wang
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Changhong Wen
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Fernanda Filho
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Nadiyeh Rouhi
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Michael I Adenawoola
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Ji Li
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
- G.V. (Sonny) Montgomery VA Medical Center, Jackson, Mississippi, USA
| |
Collapse
|
41
|
Liu L, Yao Y, Liu Y, Hong B, Li Z, Chen X, Zhang Y, Fu H, Yang D, Yang C. Targeted H 2S-Mediated Gas Therapy with pH-Sensitive Release Property for Myocardial Ischemia-Reperfusion Injury by Platelet Membrane. Biomater Res 2024; 28:0061. [PMID: 39161346 PMCID: PMC11330987 DOI: 10.34133/bmr.0061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 07/02/2024] [Indexed: 08/21/2024] Open
Abstract
Management of myocardial ischemia-reperfusion injury (MIRI) in reperfusion therapy remains a major obstacle in the field of cardiovascular disease, but current available therapies have not yet been achieved in mitigating myocardial injury due to the complex pathological mechanisms of MIRI. Exogenous delivery of hydrogen sulfide (H2S) to the injured myocardium can be an effective strategy for treating MIRI due to the multiple physiologic functions of H2S, including anti-inflammatory, anti-apoptotic, and mitochondrial protective effects. Here, to realize the precise delivery and release of H2S, we proposed the targeted H2S-mediated gas therapy with pH-sensitive release property mediated by platelet membranes (PMs). In this study, a biomimetic functional poly(lactic-co-ethanolic acid) nanoparticle (RAPA/JK-1-PLGA@PM) was fabricated by loading rapamycin (RAPA; mTOR inhibitor) and JK-1 (H2S donor) and then coated with PM. In vitro observations were conducted including pharmaceutical evaluation, H2S release behaviors, hemolysis analysis, serum stability, cellular uptake, cytotoxicity, inhibition of myocardial apoptosis, and anti-inflammation. In vivo examinations were performed including targeting ability, restoration of cardiac function, inhibition of pathological remodeling, and anti-inflammation. RAPA/JK-1-PLGA@PM was successfully prepared with good size distribution and stability. Utilizing the natural infarct-homing ability of PM, RAPA/JK-1-PLGA@PM could be effectively targeted to the damaged myocardium. RAPA/JK-1-PLGA@PM continuously released H2S triggered by inflammatory microenvironment, which could inhibit cardiomyocyte apoptosis, realize the transition of pro-inflammation, and alleviate myocardial injury demonstrated in hypoxia/reoxygenation myocardial cell in vitro. Precise delivery and release of H2S attenuated inflammatory response and cardiac damage, promoted cardiac repair, and ameliorated cardiac function proven in MIRI mouse model in vivo. This research outlined the novel nanoplatform that combined immunosuppressant agents and H2S donor with the pH-sensitive release property, offering a promising therapeutic for MIRI treatment that leveraged the synergistic effects of gas therapy.
Collapse
Affiliation(s)
- Lin Liu
- Department of Pharmacy,
The Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
- Department of Pharmacy,
Shantou University Medical College, Shantou 515041, China
| | - Yucen Yao
- Department of Pharmacy,
Shantou University Medical College, Shantou 515041, China
- College of Pharmacy, Jiamusi University, Jiamusi 154007, China
| | - Yang Liu
- Department of Pharmacy,
The Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
- Department of Pharmacy,
Shantou University Medical College, Shantou 515041, China
| | - Bingrong Hong
- Department of Pharmacy,
The Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
- Department of Pharmacy,
Shantou University Medical College, Shantou 515041, China
| | - Ziqing Li
- Department of Pharmacy,
The Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
- Department of Pharmacy,
Shantou University Medical College, Shantou 515041, China
| | - Xuejun Chen
- Department of Pharmacy,
The Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
- Department of Pharmacy,
Shantou University Medical College, Shantou 515041, China
| | - Yaofeng Zhang
- Department of Pharmacy,
The Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Hongbo Fu
- Department of Pharmacy,
The Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Degong Yang
- Department of Pharmacy,
Shantou University Medical College, Shantou 515041, China
- Department of Pharmacy, Department of Dermatology,
The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Chunrong Yang
- Department of Pharmacy,
The Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
- Department of Pharmacy,
Shantou University Medical College, Shantou 515041, China
| |
Collapse
|
42
|
Dong WS, Hu C, Hu M, Gao YP, Hu YX, Li K, Ye YJ, Zhang X. Metrnl: a promising biomarker and therapeutic target for cardiovascular and metabolic diseases. Cell Commun Signal 2024; 22:389. [PMID: 39103830 PMCID: PMC11301845 DOI: 10.1186/s12964-024-01767-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 07/27/2024] [Indexed: 08/07/2024] Open
Abstract
Modern human society is burdened with the pandemic of cardiovascular and metabolic diseases. Metrnl is a widely distributed secreted protein in the body, involved in regulating glucose and lipid metabolism and maintaining cardiovascular system homeostasis. In this review, we present the predictive and therapeutic roles of Metrnl in various cardiovascular and metabolic diseases, including atherosclerosis, ischemic heart disease, cardiac remodeling, heart failure, hypertension, chemotherapy-induced myocardial injury, diabetes mellitus, and obesity.
Collapse
Affiliation(s)
- Wen-Sheng Dong
- Department of Geriatrics, Hubei Key Laboratory of Metabolic and Chronic Diseases, Renmin Hospital of Wuhan University, Wuhan University at Jiefang Road 238, Wuhan, 430060, China
| | - Can Hu
- Department of Ultrasound, Union Hospital, Tongji Medical College, Clinical Research Center for Medical Imaging in Hubei Province, Hubei Province Key Laboratory of Molecular Imaging, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Min Hu
- Department of Cardiology, Hubei Key Laboratory of Metabolic and Chronic Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yi-Peng Gao
- Department of Cardiology, Hubei Key Laboratory of Metabolic and Chronic Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yu-Xin Hu
- Department of Geriatrics, Hubei Key Laboratory of Metabolic and Chronic Diseases, Renmin Hospital of Wuhan University, Wuhan University at Jiefang Road 238, Wuhan, 430060, China
| | - Kang Li
- Department of Geriatrics, Hubei Key Laboratory of Metabolic and Chronic Diseases, Renmin Hospital of Wuhan University, Wuhan University at Jiefang Road 238, Wuhan, 430060, China
| | - Yun-Jia Ye
- Department of Geriatrics, Hubei Key Laboratory of Metabolic and Chronic Diseases, Renmin Hospital of Wuhan University, Wuhan University at Jiefang Road 238, Wuhan, 430060, China
| | - Xin Zhang
- Department of Geriatrics, Hubei Key Laboratory of Metabolic and Chronic Diseases, Renmin Hospital of Wuhan University, Wuhan University at Jiefang Road 238, Wuhan, 430060, China.
| |
Collapse
|
43
|
Jiang C, Meng A, Shi X, Fu Z, Wang Y, Zhou J, Zhang X, Liu C. Preparation of antioxidant peptides from yak skin gelatin and their protective effect on myocardial ischemia reperfusion injury. Food Funct 2024; 15:7961-7973. [PMID: 38982985 DOI: 10.1039/d4fo00458b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/11/2024]
Abstract
We herein report a study on the antioxidant peptides that show potential in alleviating myocardial ischemia reperfusion injury (MI/RI). Yak skin gelatin fraction Ac (YSG-Ac), obtained through ultrafiltration and gel filtration with Sephadex G-15, exhibits a favorable nutrient composition, high foaming capacity and stability, and resistance against gastrointestinal digestion. LC-MS/MS analysis reveals that YSG-Ac contains 26 peptide segments with sequence lengths of 8 to 12 amino acids. Online screening suggests that the antioxidant capacity of YSG-Ac is mainly attributed to the presence of hydrophobic and antioxidant amino acids. In vitro, our results demonstrate the MI/RI protective effects of YSG-Ac by effectively repairing H2O2-induced oxidative damage in H9c2 cells, which is achieved by inhibiting malondialdehyde (MDA) levels, and increasing glutathione peroxidase (GSH-pX) and superoxide dismutase (SOD) activity. In vivo, our results further confirm the effectiveness of YSG-Ac in narrowing the area of myocardial infarction, decreasing MDA levels, increasing SOD activity, and reducing the content of lactate dehydrogenase (LDH) in a mouse MI/RI model. Molecular docking analysis indicates that PGADGQPGAK with xanthine dehydrogenase (XDH) and GAAGPTGPIGS with tumor necrosis factor-alpha (TNF-α) exhibit strong bonding capability, and other related targets also show certain binding ability toward YSG-Ac. This suggests that YSG-Ac can regulate MI/RI through multiple targets and pathways. Overall, our findings highlight the potential of YSG-Ac as a functional food ingredient with antioxidant and MI/RI protective characteristics.
Collapse
Affiliation(s)
- Chongning Jiang
- School of Pharmacy, North China University of Science and Technology, 21 Bohai Avenue, Caofeidian New Town, Tangshan, 063210 Hebei Province, China.
| | - Aiguo Meng
- Affiliated Hospital, North China University of Science and Technology, 21 Bohai Avenue, Caofeidian New Town, Tangshan, 063210 Hebei Province, China
| | - Xiaoyu Shi
- School of Pharmacy, North China University of Science and Technology, 21 Bohai Avenue, Caofeidian New Town, Tangshan, 063210 Hebei Province, China.
| | - Zhiping Fu
- School of Pharmacy, North China University of Science and Technology, 21 Bohai Avenue, Caofeidian New Town, Tangshan, 063210 Hebei Province, China.
| | - Yali Wang
- School of Pharmacy, North China University of Science and Technology, 21 Bohai Avenue, Caofeidian New Town, Tangshan, 063210 Hebei Province, China.
| | - Jingjing Zhou
- School of Pharmacy, North China University of Science and Technology, 21 Bohai Avenue, Caofeidian New Town, Tangshan, 063210 Hebei Province, China.
| | - Xiaowei Zhang
- Hebei General Hospital, No. 348 Heping West Road, Xinhua District, Shijiazhuang City, 050000, Hebei Province, China
| | - Chunyan Liu
- School of Pharmacy, North China University of Science and Technology, 21 Bohai Avenue, Caofeidian New Town, Tangshan, 063210 Hebei Province, China.
| |
Collapse
|
44
|
Guan H, Chen Y, Liu X, Huang L. Research and application of hydrogel-encapsulated mesenchymal stem cells in the treatment of myocardial infarction. Colloids Surf B Biointerfaces 2024; 239:113942. [PMID: 38729022 DOI: 10.1016/j.colsurfb.2024.113942] [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: 01/05/2024] [Revised: 04/19/2024] [Accepted: 05/02/2024] [Indexed: 05/12/2024]
Abstract
Myocardial infarction (MI) stands out as a highly lethal disease that poses a significant threat to global health. Worldwide, heart failure resulting from MI remains a leading cause of human mortality. Mesenchymal stem cell (MSC) therapy has emerged as a promising therapeutic approach, leveraging its intrinsic healing properties. Nevertheless, pervasive issues, including a low cell retention rate, suboptimal survival rate, and incomplete differentiation of MSCs, present formidable challenges for further research. The introduction and advancement of biomaterials have offered a novel avenue for the exploration of MSC therapy in MI, marking considerable progress thus far. Notably, hydrogels, among the representative biomaterials, have garnered extensive attention within the biomedical field. This review delves into recent advancements, specifically focusing on the application of hydrogels to augment MSC therapy for cardiac tissue regeneration in MI.
Collapse
Affiliation(s)
- Haien Guan
- Center of Stem Cell and Regenerative Medicine, Gaozhou People's Hospital, Gaozhou 525200, China
| | - Yuehua Chen
- Center of Stem Cell and Regenerative Medicine, Gaozhou People's Hospital, Gaozhou 525200, China
| | - Xuanyu Liu
- Center of Stem Cell and Regenerative Medicine, Gaozhou People's Hospital, Gaozhou 525200, China
| | - Li Huang
- Center of Stem Cell and Regenerative Medicine, Gaozhou People's Hospital, Gaozhou 525200, China.
| |
Collapse
|
45
|
Li T, Li Y, Zeng Y, Zhou X, Zhang S, Ren Y. Construction of preclinical evidence for propofol in the treatment of reperfusion injury after acute myocardial infarction: A systematic review and meta-analysis. Biomed Pharmacother 2024; 174:116629. [PMID: 38640712 DOI: 10.1016/j.biopha.2024.116629] [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: 01/17/2024] [Revised: 04/04/2024] [Accepted: 04/17/2024] [Indexed: 04/21/2024] Open
Abstract
Propofol, a commonly used intravenous anesthetic, has demonstrated potential in protecting against myocardial ischemia/reperfusion injury (MIRI) based on preclinical animal studies. However, the clinical benefits of propofol in this context are subject to debate. We conducted a systematic search across eight databases to identify all relevant animal studies investigating the preventive effects of propofol on MIRI until October 30, 2023. We assessed the methodological quality of the included studies using SYRCLE's bias risk tool. Statistical analysis was performed using STATA 15.1. The primary outcome measures analyzed in this study were myocardial infarct size (IS) and myocardial injury biomarkers. This study presents a comprehensive analysis of 48 relevant animal studies investigating propofol's preventive effects on MIRI. Propofol administration demonstrated a reduction in myocardial IS and decreased levels of myocardial injury biomarkers (CK-MB, LDH, cTnI). Moreover, propofol improved myocardial function parameters (+dp/dtmax, -dP/dtmax, LVEF, LVFS), exhibited favorable effects on inflammatory markers (IL-6, TNF-α) and oxidative stress markers (SOD, MDA), and reduced myocardial cell apoptotic index (AI). These findings suggest propofol exerts cardioprotective effects by reducing myocardial injury, decreasing infarct size, and improving heart function. However, the absence of animal models that accurately represent comorbidities such as aging and hypertension, as well as inconsistent administration methods that align with clinical practice, may hinder its clinical translation. Further robust investigations are required to validate these findings, elucidate the underlying mechanisms of propofol, and facilitate its potential translation into clinical practice.
Collapse
Affiliation(s)
- Tao Li
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yanwei Li
- Cardiology Department, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yiwei Zeng
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xin Zhou
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Su Zhang
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yulan Ren
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China; School of Chinese Classics, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
46
|
Tratnig-Frankl P, Andrews AR, Berkane Y, Guinier C, Goutard M, Lupon E, Lancia HH, Morrison ML, Roth MB, Randolph MA, Cetrulo CL, Lellouch AG. Exploring Iodide and Hydrogen Sulfide as ROS Scavengers to Delay Acute Rejection in MHC-Defined Vascularized Composite Allografts. Antioxidants (Basel) 2024; 13:531. [PMID: 38790636 PMCID: PMC11118872 DOI: 10.3390/antiox13050531] [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: 03/31/2024] [Revised: 04/19/2024] [Accepted: 04/24/2024] [Indexed: 05/26/2024] Open
Abstract
Vascularized composite allografts (VCA) face ischemic challenges due to their limited availability. Reperfusion following ischemia triggers oxidative stress and immune reactions, and scavenger molecules could mitigate ischemia-reperfusion injuries and, therefore, immune rejection. We compared two scavengers in a myocutaneous flap VCA model. In total, 18 myocutaneous flap transplants were performed in Major histocompatibility complex (MHC)-defined miniature swine. In the MATCH group (n = 9), donors and recipients had minor antigen mismatch, while the animals were fully mismatched in the MISMATCH group (n = 9). Grafts were pretreated with saline, sodium iodide (NaI), or hydrogen sulfide (H2S), stored at 4 °C for 3 h, and then transplanted. Flaps were monitored until clinical rejection without immunosuppression. In the MATCH group, flap survival did not significantly differ between the saline and hydrogen sulfide treatments (p = 0.483) but was reduced with the sodium iodide treatment (p = 0.007). In the MISMATCH group, survival was similar between the saline and hydrogen sulfide treatments (p = 0.483) but decreased with the sodium iodide treatment (p = 0.007). Rhabdomyolysis markers showed lower but non-significant levels in the experimental subgroups for both the MATCH and MISMATCH animals. This study provides insightful data for the field of antioxidant-based approaches in VCA and transplantation.
Collapse
Affiliation(s)
- Philipp Tratnig-Frankl
- Division of Plastic, Reconstructive and Aesthetic Surgery, Vienna General Hospital, Medical University of Vienna, 1090 Vienna, Austria;
- Vascularized Composite Allotransplantation Laboratory, Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA; (A.R.A.); (C.G.); (M.G.); (E.L.); (H.H.L.); (M.A.R.); (A.G.L.)
- Division of Plastic and Reconstructive Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA
- Shriners Children’s Boston, Boston, MA 02114, USA
| | - Alec R. Andrews
- Vascularized Composite Allotransplantation Laboratory, Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA; (A.R.A.); (C.G.); (M.G.); (E.L.); (H.H.L.); (M.A.R.); (A.G.L.)
- Division of Plastic and Reconstructive Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA
- Shriners Children’s Boston, Boston, MA 02114, USA
| | - Yanis Berkane
- Vascularized Composite Allotransplantation Laboratory, Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA; (A.R.A.); (C.G.); (M.G.); (E.L.); (H.H.L.); (M.A.R.); (A.G.L.)
- Division of Plastic and Reconstructive Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA
- Shriners Children’s Boston, Boston, MA 02114, USA
- Department of Plastic, Reconstructive and Aesthetic Surgery, CHU de Rennes, University of Rennes, 35000 Rennes, France
| | - Claire Guinier
- Vascularized Composite Allotransplantation Laboratory, Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA; (A.R.A.); (C.G.); (M.G.); (E.L.); (H.H.L.); (M.A.R.); (A.G.L.)
- Division of Plastic and Reconstructive Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA
- Shriners Children’s Boston, Boston, MA 02114, USA
- Department of Plastic Surgery, NOVO Hospital, 95300 Pontoise, France
| | - Marion Goutard
- Vascularized Composite Allotransplantation Laboratory, Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA; (A.R.A.); (C.G.); (M.G.); (E.L.); (H.H.L.); (M.A.R.); (A.G.L.)
- Division of Plastic and Reconstructive Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA
- Shriners Children’s Boston, Boston, MA 02114, USA
| | - Elise Lupon
- Vascularized Composite Allotransplantation Laboratory, Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA; (A.R.A.); (C.G.); (M.G.); (E.L.); (H.H.L.); (M.A.R.); (A.G.L.)
- Division of Plastic and Reconstructive Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA
- Shriners Children’s Boston, Boston, MA 02114, USA
- Department of Plastic and Reconstructive Surgery, Institut Universitaire Locomoteur et du Sport, Pasteur 2 Hospital, University Côte d’Azur, 06300 Nice, France
| | - Hyshem H. Lancia
- Vascularized Composite Allotransplantation Laboratory, Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA; (A.R.A.); (C.G.); (M.G.); (E.L.); (H.H.L.); (M.A.R.); (A.G.L.)
- Shriners Children’s Boston, Boston, MA 02114, USA
| | - Michael L. Morrison
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (M.L.M.); (M.B.R.)
| | - Mark B. Roth
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (M.L.M.); (M.B.R.)
| | - Mark A. Randolph
- Vascularized Composite Allotransplantation Laboratory, Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA; (A.R.A.); (C.G.); (M.G.); (E.L.); (H.H.L.); (M.A.R.); (A.G.L.)
- Division of Plastic and Reconstructive Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA
- Shriners Children’s Boston, Boston, MA 02114, USA
| | - Curtis L. Cetrulo
- Vascularized Composite Allotransplantation Laboratory, Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA; (A.R.A.); (C.G.); (M.G.); (E.L.); (H.H.L.); (M.A.R.); (A.G.L.)
- Division of Plastic and Reconstructive Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA
- Shriners Children’s Boston, Boston, MA 02114, USA
| | - Alexandre G. Lellouch
- Vascularized Composite Allotransplantation Laboratory, Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA; (A.R.A.); (C.G.); (M.G.); (E.L.); (H.H.L.); (M.A.R.); (A.G.L.)
- Division of Plastic and Reconstructive Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA
- Shriners Children’s Boston, Boston, MA 02114, USA
- INSERM UMRS 1140 Innovation Thérapeutique en Hémostase, University of Paris, 75006 Paris, France
| |
Collapse
|
47
|
Zhang MJ, Karachenets S, Gyberg DJ, Puccini S, Healy CL, Wu SC, Shearer GC, O’Connell TD. Free fatty acid receptor 4 in cardiac myocytes ameliorates ischemic cardiomyopathy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.12.589280. [PMID: 38659901 PMCID: PMC11042222 DOI: 10.1101/2024.04.12.589280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Aims Free fatty acid receptor 4 (Ffar4) is a receptor for long-chain fatty acids that attenuates heart failure driven by increased afterload. Recent findings suggest that Ffar4 prevents ischemic injury in brain, liver, and kidney, and therefore, we hypothesized that Ffar4 would also attenuate cardiac ischemic injury. Methods and Results Using a mouse model of ischemia-reperfusion (I/R), we found that mice with systemic deletion of Ffar4 (Ffar4KO) demonstrated impaired recovery of left ventricular systolic function post-I/R with no effect on initial infarct size. To identify potential mechanistic explanations for the cardioprotective effects of Ffar4, we performed bulk RNAseq to compare the transcriptomes from wild-type (WT) and Ffar4KO infarcted myocardium 3-days post-I/R. In the Ffar4KO infarcted myocardium, gene ontology (GO) analyses revealed augmentation of glycosaminoglycan synthesis, neutrophil activation, cadherin binding, extracellular matrix, rho signaling, and oxylipin synthesis, but impaired glycolytic and fatty acid metabolism, cardiac repolarization, and phosphodiesterase activity. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis indicated impaired AMPK signaling and augmented cellular senescence in the Ffar4KO infarcted myocardium. Interestingly, phosphodiesterase 6c (PDE6c), which degrades cGMP, was the most upregulated gene in the Ffar4KO heart. Further, the soluble guanylyl cyclase stimulator, vericiguat, failed to increase cGMP in Ffar4KO cardiac myocytes, suggesting increased phosphodiesterase activity. Finally, cardiac myocyte-specific overexpression of Ffar4 prevented systolic dysfunction post-I/R, defining a cardioprotective role of Ffa4 in cardiac myocytes. Conclusions Our results demonstrate that Ffar4 in cardiac myocytes attenuates systolic dysfunction post-I/R, potentially by attenuating oxidative stress, preserving mitochondrial function, and modulation of cGMP signaling.
Collapse
Affiliation(s)
- Michael J. Zhang
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN
- Cardiovascular Division, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN
- Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis, MN
| | - Sergey Karachenets
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN
| | - Dylan J. Gyberg
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN
| | - Sara Puccini
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN
| | - Chastity L. Healy
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN
| | - Steven C. Wu
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN
| | - Gregory C. Shearer
- Department of Nutritional Sciences, Pennsylvania State University, University Park, PA
| | - Timothy D. O’Connell
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN
- Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis, MN
| |
Collapse
|
48
|
Deng J, Liu Q, Ye L, Wang S, Song Z, Zhu M, Qiang F, Zhou Y, Guo Z, Zhang W, Chen T. The Janus face of mitophagy in myocardial ischemia/reperfusion injury and recovery. Biomed Pharmacother 2024; 173:116337. [PMID: 38422659 DOI: 10.1016/j.biopha.2024.116337] [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: 01/04/2024] [Revised: 02/20/2024] [Accepted: 02/23/2024] [Indexed: 03/02/2024] Open
Abstract
In myocardial ischemia/reperfusion injury (MIRI), moderate mitophagy is a protective or adaptive mechanism because of clearing defective mitochondria accumulates during MIRI. However, excessive mitophagy lead to an increase in defective mitochondria and ultimately exacerbate MIRI by causing overproduction or uncontrolled production of mitochondria. Phosphatase and tensin homolog (PTEN)-induced kinase 1 (Pink1), Parkin, FUN14 domain containing 1 (FUNDC1) and B-cell leukemia/lymphoma 2 (BCL-2)/adenovirus E1B19KD interaction protein 3 (BNIP3) are the main mechanistic regulators of mitophagy in MIRI. Pink1 and Parkin are mitochondrial surface proteins involved in the ubiquitin-dependent pathway, while BNIP3 and FUNDC1 are mitochondrial receptor proteins involved in the non-ubiquitin-dependent pathway, which play a crucial role in maintaining mitochondrial homeostasis and mitochondrial quality. These proteins can induce moderate mitophagy or inhibit excessive mitophagy to protect against MIRI but may also trigger excessive mitophagy or insufficient mitophagy, thereby worsening the condition. Understanding the actions of these mitophagy mechanistic proteins may provide valuable insights into the pathological mechanisms underlying MIRI development. Based on the above background, this article reviews the mechanism of mitophagy involved in MIRI through Pink1/Parkin pathway and the receptor mediated pathway led by FUNDC1 and BNIP3, as well as the related drug treatment, aim to provide effective strategies for the prevention and treatment of MIRI.
Collapse
Affiliation(s)
- Jiaxin Deng
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Qian Liu
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Linxi Ye
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Shuo Wang
- State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae for the Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zhenyan Song
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Mingyan Zhu
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Fangfang Qiang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Yulin Zhou
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Zhen Guo
- Hunan Provincial Key Laboratory of the Fundamental and Clinical Research on Functional Nucleic Acid, Changsha Medical University, Changsha 410219, China; Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha 410219, China; Hunan Provincial Key Laboratory of the TCM Agricultural Biogenomics, Changsha Medical University, Changsha 410219, China.
| | - Wei Zhang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China.
| | - Ting Chen
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China; National Key Laboratory Cultivation Base of Chinese Medicinal Powder & Innovative Medicinal Jointly Established by Province and Ministry, Changsha 410208, China.
| |
Collapse
|
49
|
Wang Y, Xu R, Yan Y, He B, Miao C, Fang Y, Wan H, Zhou G. Exosomes-Mediated Signaling Pathway: A New Direction for Treatment of Organ Ischemia-Reperfusion Injury. Biomedicines 2024; 12:353. [PMID: 38397955 PMCID: PMC10886966 DOI: 10.3390/biomedicines12020353] [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: 12/31/2023] [Revised: 01/24/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
Ischemia reperfusion (I/R) is a common pathological process which occurs mostly in organs like the heart, brain, kidney, and lung. The injury caused by I/R gradually becomes one of the main causes of fatal diseases, which is an urgent clinical problem to be solved. Although great progress has been made in therapeutic methods, including surgical, drug, gene therapy, and transplant therapy for I/R injury, the development of effective methods to cure the injury remains a worldwide challenge. In recent years, exosomes have attracted much attention for their important roles in immune response, antigen presentation, cell migration, cell differentiation, and tumor invasion. Meanwhile, exosomes have been shown to have great potential in the treatment of I/R injury in organs. The study of the exosome-mediated signaling pathway can not only help to reveal the mechanism behind exosomes promoting reperfusion injury recovery, but also provide a theoretical basis for the clinical application of exosomes. Here, we review the research progress in utilizing various exosomes from different cell types to promote the healing of I/R injury, focusing on the classical signaling pathways such as PI3K/Akt, NF-κB, Nrf2, PTEN, Wnt, MAPK, toll-like receptor, and AMPK. The results suggest that exosomes regulate these signaling pathways to reduce oxidative stress, regulate immune responses, decrease the expression of inflammatory cytokines, and promote tissue repair, making exosomes a competitive emerging vector for treating I/R damage in organs.
Collapse
Affiliation(s)
- Yanying Wang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou 310053, China; (Y.W.); (B.H.); (C.M.)
| | - Ruojiao Xu
- College of Life Science, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou 310053, China; (R.X.); (Y.Y.); (Y.F.)
| | - Yujia Yan
- College of Life Science, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou 310053, China; (R.X.); (Y.Y.); (Y.F.)
| | - Binyu He
- The Second Clinical Medical College, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou 310053, China; (Y.W.); (B.H.); (C.M.)
| | - Chaoyi Miao
- The Second Clinical Medical College, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou 310053, China; (Y.W.); (B.H.); (C.M.)
| | - Yifeng Fang
- College of Life Science, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou 310053, China; (R.X.); (Y.Y.); (Y.F.)
| | - Haitong Wan
- College of Life Science, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou 310053, China; (R.X.); (Y.Y.); (Y.F.)
| | - Guoying Zhou
- College of Life Science, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou 310053, China; (R.X.); (Y.Y.); (Y.F.)
| |
Collapse
|
50
|
Kumphune S, Seenak P, Paiyabhrom N, Songjang W, Pankhong P, Jumroon N, Thaisakun S, Phaonakrop N, Roytrakul S, Malakul W, Jiraviriyakul A, Nernpermpisooth N. Cardiac endothelial ischemia/reperfusion injury-derived protein damage-associated molecular patterns disrupt the integrity of the endothelial barrier. Heliyon 2024; 10:e24600. [PMID: 38312663 PMCID: PMC10835233 DOI: 10.1016/j.heliyon.2024.e24600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 01/05/2024] [Accepted: 01/10/2024] [Indexed: 02/06/2024] Open
Abstract
Human cardiac microvascular endothelial cells (HCMECs) are sensitive to ischemia and vulnerable to damage during reperfusion. The release of damage-associated molecular patterns (DAMPs) during reperfusion induces additional tissue damage. The current study aimed to identify early protein DAMPs in human cardiac microvascular endothelial cells subjected to ischemia-reperfusion injury (IRI) using a proteomic approach and their effect on endothelial cell injury. HCMECs were subjected to 60 min of simulated ischemia and 6 h of reperfusion, which can cause lethal damage. DAMPs in the culture media were subjected to liquid chromatography-tandem mass spectrometry proteomic analysis. The cells were treated with endothelial IRI-derived DAMP medium for 24 h. Endothelial injury was assessed by measuring lactate dehydrogenase activity, morphological features, and the expression of endothelial cadherin, nitric oxide synthase (eNOS), and caveolin-1. The top two upregulated proteins, DNAJ homolog subfamily B member 11 and pyrroline-5-carboxylate reductase 2, are promising and sensitive predictors of cardiac microvascular endothelial damage. HCMECs expose to endothelial IRI-derived DAMP, the lactate dehydrogenase activity was significantly increased compared with the control group (10.15 ± 1.03 vs 17.67 ± 1.19, respectively). Following treatment with endothelial IRI-derived DAMPs, actin-filament dysregulation, and downregulation of vascular endothelial cadherin, caveolin-1, and eNOS expressions were observed, along with cell death. In conclusion, the early protein DAMPs released during cardiac microvascular endothelial IRI could serve as novel candidate biomarkers for acute myocardial IRI. Distinct features of impaired plasma membrane integrity can help identify therapeutic targets to mitigate the detrimental consequences mediated of endothelial IRI-derived DAMPs.
Collapse
Affiliation(s)
- Sarawut Kumphune
- Biomedical Engineering and Innovation Research Centre, Chiang Mai University, Muang, Chiang Mai, 50200, Thailand
- Biomedical Engineering Institute, Chiang Mai University, Muang, Chiang Mai, 50200, Thailand
- Integrative Biomedical Research Unit (IBRU), Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, 65000, Thailand
| | - Porrnthanate Seenak
- Integrative Biomedical Research Unit (IBRU), Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, 65000, Thailand
- Department of Cardio-Thoracic Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, 65000, Thailand
| | - Nitchawat Paiyabhrom
- Department of Medical Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, 65000, Thailand
| | - Worawat Songjang
- Integrative Biomedical Research Unit (IBRU), Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, 65000, Thailand
- Department of Medical Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, 65000, Thailand
| | - Panyupa Pankhong
- Integrative Biomedical Research Unit (IBRU), Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, 65000, Thailand
- Department of Medical Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, 65000, Thailand
| | - Noppadon Jumroon
- Integrative Biomedical Research Unit (IBRU), Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, 65000, Thailand
- Department of Medical Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, 65000, Thailand
| | - Siriwan Thaisakun
- National Centre for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathum Thani, 12120, Thailand
| | - Narumon Phaonakrop
- National Centre for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathum Thani, 12120, Thailand
| | - Sittiruk Roytrakul
- National Centre for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathum Thani, 12120, Thailand
| | - Wachirawadee Malakul
- Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok, 65000, Thailand
| | - Arunya Jiraviriyakul
- Integrative Biomedical Research Unit (IBRU), Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, 65000, Thailand
- Department of Medical Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, 65000, Thailand
| | - Nitirut Nernpermpisooth
- Integrative Biomedical Research Unit (IBRU), Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, 65000, Thailand
- Department of Cardio-Thoracic Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, 65000, Thailand
| |
Collapse
|