1
|
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
|
2
|
Szabados T, Makkos A, Ágg B, Benczik B, Brenner GG, Szabó M, Váradi B, Vörös I, Gömöri K, Varga ZV, Görbe A, Bencsik P, Ferdinandy P. Pharmacokinetics and cardioprotective efficacy of intravenous miR-125b* microRNA mimic in a mouse model of acute myocardial infarction. Br J Pharmacol 2025; 182:432-450. [PMID: 39472767 DOI: 10.1111/bph.17345] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 07/16/2024] [Accepted: 08/01/2024] [Indexed: 12/13/2024] Open
Abstract
BACKGROUND AND PURPOSE MicroRNA (miRNA) therapy is a promising approach to induce cardioprotection. We have previously identified cardiac microRNA-125b* (microRNA-125b-2-3p; miR-125b*) as a potential cardioprotective miRNA, termed ProtectomiR. We aimed to characterize the pharmacokinetics and pharmacodynamics, and the effect of miR-125b* mimic on infarct size using an in vivo mouse model. EXPERIMENTAL APPROACH To characterize the pharmacokinetics properties of miR-125b* mimic, a single injection of 10-μg miR-125b* mimic or its scramble miRNA control, or vehicle i.v. was given to C57BL/6 mice. MiR-125b* expression was measured from plasma, heart, kidney and liver samples. Effect of miR-125b* on area at risk and infarct size was assessed after 45-min coronary occlusion, followed by 24-h reperfusion; 10-μg miR-125b* mimic or 10-μg non-targeting miRNA mimic control or vehicle were administered via the right jugular vein at 10th mins of coronary occlusion. To assess molecular mechanism involved in cardioprotection, expression of mRNA targets of miR-125b* were measured from ventricular myocardium at 1, 2, 4, 8 or 24 h post-treatment using quantitative real time polymerase chain reaction. KEY RESULTS MiR-125b* expression was markedly increased in plasma and myocardium 1 h, and in the liver 2h after treatment. Infarct size was significantly reduced after miR-125b* mimic treatment when compared to the vehicle. The expression of Ccna2, Eef2k and Cacnb2 target mRNAs was significantly reduced 8 h after injection of miR-125b* mimic. CONCLUSION AND IMPLICATIONS This is the first demonstration of pharmacokinetic and molecular pharmacodynamic properties as well as the cardioprotective effect of miR-125b* mimic in vivo. LINKED ARTICLES This article is part of a themed issue Non-coding RNA Therapeutics. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v182.2/issuetoc.
Collapse
Affiliation(s)
- Tamara Szabados
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - András Makkos
- Pharmahungary Group, Szeged, Hungary
- Cardiometabolic and HUN-REN-SU and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, and Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
| | - Bence Ágg
- Pharmahungary Group, Szeged, Hungary
- Cardiometabolic and HUN-REN-SU and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, and Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
| | - Bettina Benczik
- Pharmahungary Group, Szeged, Hungary
- Cardiometabolic and HUN-REN-SU and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, and Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
| | - Gábor G Brenner
- Cardiometabolic and HUN-REN-SU and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, and Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
| | - Márta Szabó
- Cardiometabolic and HUN-REN-SU and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, and Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
| | - Barnabás Váradi
- Cardiometabolic and HUN-REN-SU and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, and Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
| | - Imre Vörös
- Cardiometabolic and HUN-REN-SU and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, and Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
| | - Kamilla Gömöri
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Zoltán V Varga
- Cardiometabolic and HUN-REN-SU and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, and Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
| | - Anikó Görbe
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- Pharmahungary Group, Szeged, Hungary
- Cardiometabolic and HUN-REN-SU and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, and Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
| | - Péter Bencsik
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Péter Ferdinandy
- Pharmahungary Group, Szeged, Hungary
- Cardiometabolic and HUN-REN-SU and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, and Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
| |
Collapse
|
3
|
Tan J, Min J, Jiang Y, Liu S, Ke M, Wang Z, Yang HT. CircCHSY1 protects hearts against ischaemia/reperfusion injury by enhancing heme oxygenase 1 expression via miR-24-3p. Cardiovasc Res 2024; 120:1924-1938. [PMID: 39082269 DOI: 10.1093/cvr/cvae162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 03/14/2024] [Accepted: 05/03/2024] [Indexed: 12/11/2024] Open
Abstract
AIMS Circular RNAs (circRNAs) are important players involved in a variety of physiological and pathological processes. However, their functions and mechanisms during myocardial ischaemic injury and protection remain largely unknown. We recently found significant alterations of many circRNAs including circCHSY1 following myocardial ischaemia/reperfusion (I/R) injury, whereas their exact functions are unclear. Here, we investigated the roles of circCHSY1 in the acute myocardial I/R injury and the potential mechanisms involved. METHODS AND RESULTS The expression of circCHSY1 was detected in cardiomyocytes from mouse, rat, and human embryonic stem cells (hESC-CMs). It was further up-regulated in mouse I/R (30 min/24 h) hearts, oxygen glucose deprivation/reperfusion (OGD/R, 6 h/2 h) primary neonatal rat ventricular cardiomyocytes (NRCMs) and OGD/R (48 h/2 h) hESC-CMs. Adenovirus-mediated circCHSY1 overexpression significantly decreased infarct size and lactate dehydrogenase (LDH) release in mouse I/R hearts. Consistently, circCHSY1 overexpression reduced the LDH release in the OGD/R NRCMs and hESC-CMs, improved cell viability, and preserved mitochondrial function in the OGD/R NRCMs, whereas there were no significant differences in cell viability and LDH release between the OGD/R NRCMs with and without small interfering RNA (siRNA)-mediated circCHSY1 knockdown. Mechanistically, circCHSY1 was detected to bind with miR-24-3p analysed by dual-luciferase assay and RNA pull-down assays. CircCHSY1 overexpression-mediated protective effects on cells and mitochondria in OGD/R NRCMs were reversed by the miR-24-3p mimic. Furthermore, dual-luciferase assay showed that miR-24-3p was directly bound to heme oxygenase 1 (HO1) via its 3'UTR. The protein level of HO1 was down-regulated by miR-24-3p mimic in OGD/R NRCMs but enhanced by the circCHSY1 overexpression in vitro and in vivo. Functionally, the HO1 knockdown by adenovirus in vivo and by siRNA in vitro eliminated cardioprotective effects of circCHSY1 overexpression. CONCLUSION CircCHSY1 is up-regulated following myocardial I/R injury. The higher level of circCHSY1 protects I/R hearts and cardiomyocytes. The protection of circCHSY1 is mediated through enhancement of the HO1 level, resulting in preserving mitochondrial homoeostasis via targeting miR-24-3p in cardiomyocytes. These findings suggest circCHSY1 as a protective factor.
Collapse
MESH Headings
- Animals
- MicroRNAs/metabolism
- MicroRNAs/genetics
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Myocardial Reperfusion Injury/enzymology
- Myocardial Reperfusion Injury/pathology
- Myocardial Reperfusion Injury/genetics
- Myocardial Reperfusion Injury/metabolism
- Myocardial Reperfusion Injury/prevention & control
- Humans
- RNA, Circular/metabolism
- RNA, Circular/genetics
- Disease Models, Animal
- Mice, Inbred C57BL
- Male
- Cells, Cultured
- Human Embryonic Stem Cells/metabolism
- Human Embryonic Stem Cells/enzymology
- Human Embryonic Stem Cells/pathology
- Myocardial Infarction/enzymology
- Myocardial Infarction/pathology
- Myocardial Infarction/metabolism
- Myocardial Infarction/genetics
- Myocardial Infarction/prevention & control
- Signal Transduction
- Rats, Sprague-Dawley
- Heme Oxygenase-1/metabolism
- Heme Oxygenase-1/genetics
- Mitochondria, Heart/enzymology
- Mitochondria, Heart/metabolism
- Mitochondria, Heart/pathology
- Rats
- 3' Untranslated Regions
- Heme Oxygenase (Decyclizing)
- Membrane Proteins
Collapse
Affiliation(s)
- Jiliang Tan
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Laboratory of Molecular Cardiology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences (CAS), CAS, 320 Yue Yang Road, Shanghai 200031, P.R. China
| | - Jie Min
- Department of Cardiovascular Surgery, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai 200003, P.R. China
| | - Yun Jiang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Laboratory of Molecular Cardiology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences (CAS), CAS, 320 Yue Yang Road, Shanghai 200031, P.R. China
| | - Shenyan Liu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Laboratory of Molecular Cardiology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences (CAS), CAS, 320 Yue Yang Road, Shanghai 200031, P.R. China
| | - Minxia Ke
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Laboratory of Molecular Cardiology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences (CAS), CAS, 320 Yue Yang Road, Shanghai 200031, P.R. China
| | - Zhinong Wang
- Department of Cardiovascular Surgery, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai 200003, P.R. China
| | - Huang-Tian Yang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Laboratory of Molecular Cardiology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences (CAS), CAS, 320 Yue Yang Road, Shanghai 200031, P.R. China
| |
Collapse
|
4
|
Joghataie P, Ardakani MB, Sabernia N, Salary A, Khorram S, Sohbatzadeh T, Goodarzi V, Amiri BS. The Role of Circular RNA in the Pathogenesis of Chemotherapy-Induced Cardiotoxicity in Cancer Patients: Focus on the Pathogenesis and Future Perspective. Cardiovasc Toxicol 2024; 24:1151-1167. [PMID: 39158829 DOI: 10.1007/s12012-024-09914-w] [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: 09/21/2023] [Accepted: 08/11/2024] [Indexed: 08/20/2024]
Abstract
Cardiotoxicity is a serious challenge cancer patients face today. Various factors are involved in cardiotoxicity. Circular RNAs (circRNAs) are one of the effective factors in the occurrence and prevention of cardiotoxicity. circRNAs can lead to increased proliferation, apoptosis, and regeneration of cardiomyocytes by regulating the molecular pathways, as well as increasing or decreasing gene expression; some circRNAs have a dual role in cardiomyocyte regeneration or death. Identifying each of the pathways related to these processes can be effective on managing patients and preventing cardiotoxicity. In this study, an overview of the molecular pathways involved in cardiotoxicity by circRNAs and their effects on the downstream factors have been discussed.
Collapse
Affiliation(s)
- Pegah Joghataie
- Department of Cardiology, School of Medicine, Hazrat-E Rasool General Hospital, Iran University of Medical Sciences, Tehran, Iran
| | | | - Neda Sabernia
- Department of Internal Medicine, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | | | - Tooba Sohbatzadeh
- Student Research Committee, School of Medicine, Alborz University of Medical Sciences, Alborz, Iran
| | - Vahid Goodarzi
- Department of Anesthesiology, Rasoul-Akram Medical Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Bahareh Shateri Amiri
- Assistant Professor of Internal Medicine, Department of Internal Medicine, School of Medicine, Hazrat-E Rasool General Hospital, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Mei S, Ma X, Zhou L, Wuyun Q, Cai Z, Yan J, Ding H. Circular RNA in Cardiovascular Diseases: Biogenesis, Function and Application. Biomolecules 2024; 14:952. [PMID: 39199340 PMCID: PMC11352787 DOI: 10.3390/biom14080952] [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: 06/30/2024] [Revised: 07/31/2024] [Accepted: 08/05/2024] [Indexed: 09/01/2024] Open
Abstract
Cardiovascular diseases pose a significant public health challenge globally, necessitating the development of effective treatments to mitigate the risk of cardiovascular diseases. Recently, circular RNAs (circRNAs), a novel class of non-coding RNAs, have been recognized for their role in cardiovascular disease. Aberrant expression of circRNAs is closely linked with changes in various cellular and pathophysiological processes within the cardiovascular system, including metabolism, proliferation, stress response, and cell death. Functionally, circRNAs serve multiple roles, such as acting as a microRNA sponge, providing scaffolds for proteins, and participating in protein translation. Owing to their unique properties, circRNAs may represent a promising biomarker for predicting disease progression and a potential target for cardiovascular drug development. This review comprehensively examines the properties, biogenesis, and potential mechanisms of circRNAs, enhancing understanding of their role in the pathophysiological processes impacting cardiovascular disease. Furthermore, the prospective clinical applications of circRNAs in the diagnosis, prognosis, and treatment of cardiovascular disease are addressed.
Collapse
Affiliation(s)
- Shuai Mei
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave., Wuhan 430030, China; (S.M.); (X.M.); (L.Z.); (Q.W.); (Z.C.)
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Xiaozhu Ma
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave., Wuhan 430030, China; (S.M.); (X.M.); (L.Z.); (Q.W.); (Z.C.)
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Li Zhou
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave., Wuhan 430030, China; (S.M.); (X.M.); (L.Z.); (Q.W.); (Z.C.)
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Qidamugai Wuyun
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave., Wuhan 430030, China; (S.M.); (X.M.); (L.Z.); (Q.W.); (Z.C.)
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Ziyang Cai
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave., Wuhan 430030, China; (S.M.); (X.M.); (L.Z.); (Q.W.); (Z.C.)
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Jiangtao Yan
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave., Wuhan 430030, China; (S.M.); (X.M.); (L.Z.); (Q.W.); (Z.C.)
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Hu Ding
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave., Wuhan 430030, China; (S.M.); (X.M.); (L.Z.); (Q.W.); (Z.C.)
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave., Wuhan 430030, China
| |
Collapse
|
6
|
Abubakar M, Hajjaj M, Naqvi ZEZ, Shanawaz H, Naeem A, Padakanti SSN, Bellitieri C, Ramar R, Gandhi F, Saleem A, Abdul Khader AHS, Faraz MA. Non-Coding RNA-Mediated Gene Regulation in Cardiovascular Disorders: Current Insights and Future Directions. J Cardiovasc Transl Res 2024; 17:739-767. [PMID: 38092987 DOI: 10.1007/s12265-023-10469-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 11/23/2023] [Indexed: 09/04/2024]
Abstract
Cardiovascular diseases (CVDs) pose a significant burden on global health. Developing effective diagnostic, therapeutic, and prognostic indicators for CVDs is critical. This narrative review explores the role of select non-coding RNAs (ncRNAs) and provides an in-depth exploration of the roles of miRNAs, lncRNAs, and circRNAs in different aspects of CVDs, offering insights into their mechanisms and potential clinical implications. The review also sheds light on the diverse functions of ncRNAs, including their modulation of gene expression, epigenetic modifications, and signaling pathways. It comprehensively analyzes the interplay between ncRNAs and cardiovascular health, paving the way for potential novel interventions. Finally, the review provides insights into the methodologies used to investigate ncRNA-mediated gene regulation in CVDs, as well as the implications and challenges associated with translating ncRNA research into clinical applications. Considering the broader implications, this research opens avenues for interdisciplinary collaborations, enhancing our understanding of CVDs across scientific disciplines.
Collapse
Affiliation(s)
- Muhammad Abubakar
- Department of Internal Medicine, Ameer-Ud-Din Medical College, Lahore General Hospital, Lahore, Punjab, Pakistan.
| | - Mohsin Hajjaj
- Department of Internal Medicine, Jinnah Hospital, Lahore, Punjab, Pakistan
| | - Zil E Zehra Naqvi
- Department of Internal Medicine, Jinnah Hospital, Lahore, Punjab, Pakistan
| | - Hameed Shanawaz
- Department of Internal Medicine, Windsor University School of Medicine, Cayon, Saint Kitts and Nevis
| | - Ammara Naeem
- Department of Cardiology, Heart & Vascular Institute, Dearborn, Michigan, USA
| | | | | | - Rajasekar Ramar
- Department of Internal Medicine, Rajah Muthiah Medical College, Chidambaram, Tamil Nadu, India
| | - Fenil Gandhi
- Department of Family Medicine, Lower Bucks Hospital, Bristol, PA, USA
| | - Ayesha Saleem
- Department of Internal Medicine, Jinnah Hospital, Lahore, Punjab, Pakistan
| | | | - Muhammad Ahmad Faraz
- Department of Forensic Medicine, Postgraduate Medical Institute, Lahore, Punjab, Pakistan
| |
Collapse
|
7
|
Tang L, Nyarige V, Li P, Wang J, Zhu W. Identification of circular RNAs regulating cardiomyocyte proliferation in neonatal pig hearts. JCI Insight 2024; 9:e175625. [PMID: 38916964 PMCID: PMC11383601 DOI: 10.1172/jci.insight.175625] [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: 09/08/2023] [Accepted: 06/20/2024] [Indexed: 06/27/2024] Open
Abstract
Little is known about the expression patterns and functions of circular RNAs (circRNAs) in the heart of large mammals. In this study, we examined the expression profiles of circRNAs, microRNAs (miRNAs), and messenger RNAs (mRNAs) in neonatal pig hearts. Pig heart samples collected on postnatal days 1 (P1), 3 (P3), 7 (P7), and 28 (P28) were sent for total RNA sequencing. Our data revealed a total of 7,000 circRNAs in the 24 pig hearts. Pathway enrichment analysis of hallmark gene sets demonstrated that differentially expressed circRNAs were engaged in different pathways. The most significant difference was observed between P1 and the other 3 groups (P3, P7, and P28) in pathways related to cell cycle and muscle development. Out of the 10 circRNAs that were validated through real-time quantitative PCR to verify their expression, 6 exhibited significant effects on cell cycle activity in human induced pluripotent stem cell-derived cardiomyocytes following small interfering RNA-mediated knockdown. circRNA-miRNA-mRNA networks were constructed to understand the potential mechanisms of circRNAs in the heart. In conclusion, our study provided a data set for exploring the roles of circRNAs in pig hearts. In addition, we identified several circRNAs that regulate cardiomyocyte cell cycle.
Collapse
Affiliation(s)
- Ling Tang
- Departments of Cardiovascular Medicine and Physiology and Biomedical Engineering, Center for Regenerative Biotherapeutics, and
| | - Verah Nyarige
- Departments of Cardiovascular Medicine and Physiology and Biomedical Engineering, Center for Regenerative Biotherapeutics, and
- Department of Quantitative Health Sciences Research, Center for Individualized Medicine, Mayo Clinic Arizona, Scottsdale, Arizona, USA
| | - Pengsheng Li
- Departments of Cardiovascular Medicine and Physiology and Biomedical Engineering, Center for Regenerative Biotherapeutics, and
| | - Junwen Wang
- Department of Quantitative Health Sciences Research, Center for Individualized Medicine, Mayo Clinic Arizona, Scottsdale, Arizona, USA
- Division of Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, the University of Hong Kong, Hong Kong SAR, China
| | - Wuqiang Zhu
- Departments of Cardiovascular Medicine and Physiology and Biomedical Engineering, Center for Regenerative Biotherapeutics, and
| |
Collapse
|
8
|
Liu Y, Guo Y, Wang N, Dong Z, Yang X, Liu Q, Wang G, Qin M, Song J, Li J, Xu L, Dong Y, Zhong J. Plasma levels of Sirtuin 7 are decreased in patients with essential hypertension. Clin Biochem 2024; 127-128:110766. [PMID: 38679273 DOI: 10.1016/j.clinbiochem.2024.110766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 04/22/2024] [Accepted: 04/23/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND Sirtuin 7 (SIRT7), as a nicotinamide adenine dinucleotide-dependent protein/histone deacetylase, has been implicated in the pathogenesis of cardiovascular diseases. However, whether SIRT7 is related to hypertension remains largely unclear. Thus, this study aims to explore the effects and correlation between SIRT7 and hypertension. METHODS A total of 72 patients with essential hypertension and 82 controls with non-hypertension were recruited at Beijing Tongren Hospital Affiliated with Capital Medical University from July 2022 to June 2023. Plasma SIRT7 expression was measured using enzyme-linked immunosorbent assay analysis. Clinical baseline characteristics, laboratory measurements, echocardiographic data, and medical therapy were collected. RESULTS Plasma levels of SIRT7 were lower in hypertensive patients compared with non-hypertensive patients [0.97 (0.58-1.30) vs. 1.24 (0.99-1.46) ng/mL, P < 0.001, respectively]. Furthermore, compared with the low SIRT7 group, there were lower levels of systolic blood pressure, hyperlipidemia, and the ultrasonic electrocardiogram parameters left ventricular end-diastolic diameter and left atrial in diastole in the high SIRT7 group (P < 0.05, respectively). More importantly, multivariate logistic regression analyses indicated that plasma SIRT7 was a predictor of hypertension [OR: 0.06, 95 % CI (0.02-0.19), P < 0.001]. Receiver operating characteristics curve analysis revealed that the optimal cutoff value for plasma SIRT7 levels in detecting hypertension was determined as 0.85 ng/mL with a sensitivity of 73.6 % and a specificity of 89.0 %. The area under the curve for SIRT7 was 0.821 (95 % CI, 0.751-0.878; P < 0.001). CONCLUSION Plasma levels of SIRT7 are decreased in patients with essential hypertension, implying its potential as a biomarker for diagnosing essential hypertension..
Collapse
Affiliation(s)
- Ying Liu
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Ying Guo
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China; Department of Geriatrics, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Ning Wang
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China; Department of Geriatrics, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Zhaojie Dong
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China; Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University. Beijing, China
| | - Xiaoyan Yang
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China; Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University. Beijing, China
| | - Qian Liu
- Department of Geriatrics, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Guohong Wang
- Department of Geriatrics, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Mingzhao Qin
- Department of Geriatrics, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jiawei Song
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China; Department of Geriatrics, Beijing Tongren Hospital, Capital Medical University, Beijing, China; Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University. Beijing, China
| | - Jing Li
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China; Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University. Beijing, China
| | - Li Xu
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China; Department of Geriatrics, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Ying Dong
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China; Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University. Beijing, China.
| | - Jiuchang Zhong
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China; Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University. Beijing, China; Medical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
9
|
Zahedian S, Hadizadeh M, Farazi MM, Jafarinejad-Farsangi S. MiRNA-miRNA interaction network in peripheral blood of patients with myocardial infarction: a gene expression meta-analysis. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2024; 43:998-1015. [PMID: 38497563 DOI: 10.1080/15257770.2024.2330597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 03/05/2024] [Accepted: 03/07/2024] [Indexed: 03/19/2024]
Abstract
In recent years, investigations have revealed that microRNAs (miRNAs) can bind together and form a miRNA-miRNA-mRNA regulatory network that alters the consequence of miRNA-mRNA interaction. If we consider the miRNA that binds to mRNA as the primary miRNA and the miRNA that binds to the primary miRNA as the secondary one, secondry miRNAs can act as master regulators upstream of primary miRNAs and their target mRNAs. One of the distinguishing characteristics of secondary miRNAs as master regulators within a diverse set of differentially expressed genes is the absence of direct target mRNA for them. Instead, these master regulators exclusively govern the regulation of miRNAs that target specific mRNAs. Through in silico analysis, we identified 18 miRNAs among 385 differentially expressed miRNAs (DEmiRNAs) with no direct target mRNAs among 58 differentially expressed mRNAs (DEmRNAs) in peripheral blood of patients with myocardial infarction (MI). Instead, these secondary miRNAs targeted 9 primary miRNAs that had 36 direct targets among 58 DEmRNAs. We found that one primary miRNA might be regulated by more than one secondary miRNAs and each secondary miRNA can target more than one primary miRNAs. Among identified miRNA-miRNA-mRNA networks miR-188-5p/miR-299-3p/natural killer cell granule protein (NKG7), miR-200a-3p/miR-199b-5p/granzyme B (GZMB), and miR-377-3p/miR-581/oviductal glycoprotein 1 (OVGP1) exhibited higher scors in terms of expression levels (>2-fold increase or decrease) and strengh of interactions (ΔG < -5). Given the extensive network of miRNA interactions, focusing on master regulators opens up avenues for identifying key regulatory nodes for more effective therapeutic strategies.
Collapse
Affiliation(s)
- Setareh Zahedian
- Student Research Committee, Kerman University of Medical Science, Kerman, Iran
| | - Morteza Hadizadeh
- Cardiovascular Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Mojtaba Farazi
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | | |
Collapse
|
10
|
Hoque P, Romero B, Akins RE, Batish M. Exploring the Multifaceted Biologically Relevant Roles of circRNAs: From Regulation, Translation to Biomarkers. Cells 2023; 12:2813. [PMID: 38132133 PMCID: PMC10741722 DOI: 10.3390/cells12242813] [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: 11/15/2023] [Revised: 12/02/2023] [Accepted: 12/05/2023] [Indexed: 12/23/2023] Open
Abstract
CircRNAs are a category of regulatory RNAs that have garnered significant attention in the field of regulatory RNA research due to their structural stability and tissue-specific expression. Their circular configuration, formed via back-splicing, results in a covalently closed structure that exhibits greater resistance to exonucleases compared to linear RNAs. The distinctive regulation of circRNAs is closely associated with several physiological processes, as well as the advancement of pathophysiological processes in several human diseases. Despite a good understanding of the biogenesis of circular RNA, details of their biological roles are still being explored. With the steady rise in the number of investigations being carried out regarding the involvement of circRNAs in various regulatory pathways, understanding the biological and clinical relevance of circRNA-mediated regulation has become challenging. Given the vast landscape of circRNA research in the development of the heart and vasculature, we evaluated cardiovascular system research as a model to critically review the state-of-the-art understanding of the biologically relevant functions of circRNAs. We conclude the review with a discussion of the limitations of current functional studies and provide potential solutions by which these limitations can be addressed to identify and validate the meaningful and impactful functions of circRNAs in different physiological processes and diseases.
Collapse
Affiliation(s)
- Parsa Hoque
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA;
| | - Brigette Romero
- Department of Medical and Molecular Sciences, University of Delaware, Newark, DE 19716, USA;
| | - Robert E Akins
- Nemours Children’s Research, Nemours Children’s Health System, Wilmington, DE 19803, USA;
| | - Mona Batish
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA;
- Department of Medical and Molecular Sciences, University of Delaware, Newark, DE 19716, USA;
| |
Collapse
|
11
|
Ghafouri-Fard S, Shoorei H, Sabernia T, Hussen BM, Taheri M, Pourmoshtagh H. Circular RNAs and inflammation: Epigenetic regulators with diagnostic role. Pathol Res Pract 2023; 251:154912. [PMID: 38238072 DOI: 10.1016/j.prp.2023.154912] [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: 09/12/2023] [Revised: 10/18/2023] [Accepted: 10/26/2023] [Indexed: 01/23/2024]
Abstract
Circular RNAs (circRNAs) are a group of transcripts generally known to be non-coding transcripts, but occasionally producing short peptides. Circ_Ttc3/miR-148a, circ_TLK1/miR-106a-5p, circ_VMA21/miR-9-3p, circ_0068,888/miR-21-5p, circ_VMA21/miR-199a-5p, circ_AFF2/miR-375, circ_0008360/miR-135b-5p and circ-FBXW7/miR-216a-3p are examples of circRNA/miRNA pairs that contribute in the pathogenesis of immune-related conditions. CircRNAs have been found to regulate function of immune system and participate in the pathophysiology of immune-related disorders. In the current study, we searched PubMed and Google Scholar databases until July 2022 with the key words "circRNA" OR "circular RNA" AND "inflammation". Then, we assessed the abstract of retrieved articles to include original articles that assessed contribution of circRNAs in the pathoetiology of inflammation and related disorders. Finally, we went through the main texts of the articles and tabulated the available information. Therefore, the current study summarizes the role of circRNAs in the pathoetiology of sepsis, atherosclerosis, rheumatoid arthritis and osteoarthritis, immune-related cardiovascular, pulmonary, gastrointestinal and nervous system disorders.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamed Shoorei
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran; Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Toofan Sabernia
- Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Bashdar Mahmud Hussen
- Department of Biomedical Sciences, College of Science, Cihan University-Erbil, Kurdistan Region, Iraq; Department of Clinical Analysis, College of Pharmacy, Hawler Medical University, Kurdistan Region, Iraq
| | - Mohammad Taheri
- Institute of Human Genetics, Jena University Hospital, Jena, Germany; Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Hasan Pourmoshtagh
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
12
|
Long Q, Lv B, Jiang S, Lin J. The Landscape of Circular RNAs in Cardiovascular Diseases. Int J Mol Sci 2023; 24:ijms24054571. [PMID: 36902000 PMCID: PMC10003248 DOI: 10.3390/ijms24054571] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 02/18/2023] [Accepted: 02/20/2023] [Indexed: 03/03/2023] Open
Abstract
Cardiovascular disease (CVD) remains the leading cause of mortality globally. Circular RNAs (circRNAs) have attracted extensive attention for their roles in the physiological and pathological processes of various cardiovascular diseases (CVDs). In this review, we briefly describe the current understanding of circRNA biogenesis and functions and summarize recent significant findings regarding the roles of circRNAs in CVDs. These results provide a new theoretical basis for diagnosing and treating CVDs.
Collapse
Affiliation(s)
- Qi Long
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Bingjie Lv
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shijiu Jiang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jibin Lin
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Correspondence:
| |
Collapse
|
13
|
Circular RNAs: Biogenesis, Biological Functions, and Roles in Myocardial Infarction. Int J Mol Sci 2023; 24:ijms24044233. [PMID: 36835653 PMCID: PMC9963350 DOI: 10.3390/ijms24044233] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 02/08/2023] [Accepted: 02/14/2023] [Indexed: 02/25/2023] Open
Abstract
Non-coding RNAs have been excavated as important cardiac function modulators and linked to heart diseases. Significant advances have been obtained in illuminating the effects of microRNAs and long non-coding RNAs. Nevertheless, the characteristics of circular RNAs are rarely mined. Circular RNAs (circRNAs) are widely believed to participate in cardiac pathologic processes, especially in myocardial infarction. In this review, we round up the biogenesis of circRNAs, briefly describe their biological functions, and summarize the latest literature on multifarious circRNAs related to new therapies and biomarkers for myocardial infarction.
Collapse
|
14
|
Fadaei S, Zarepour F, Parvaresh M, Motamedzadeh A, Tamehri Zadeh SS, Sheida A, Shabani M, Hamblin MR, Rezaee M, Zarei M, Mirzaei H. Epigenetic regulation in myocardial infarction: Non-coding RNAs and exosomal non-coding RNAs. Front Cardiovasc Med 2022; 9:1014961. [PMID: 36440025 PMCID: PMC9685618 DOI: 10.3389/fcvm.2022.1014961] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 10/17/2022] [Indexed: 08/13/2023] Open
Abstract
Myocardial infarction (MI) is one of the leading causes of deaths globally. The early diagnosis of MI lowers the rate of subsequent complications and maximizes the benefits of cardiovascular interventions. Many efforts have been made to explore new therapeutic targets for MI, and the therapeutic potential of non-coding RNAs (ncRNAs) is one good example. NcRNAs are a group of RNAs with many different subgroups, but they are not translated into proteins. MicroRNAs (miRNAs) are the most studied type of ncRNAs, and have been found to regulate several pathological processes in MI, including cardiomyocyte inflammation, apoptosis, angiogenesis, and fibrosis. These processes can also be modulated by circular RNAs and long ncRNAs via different mechanisms. However, the regulatory role of ncRNAs and their underlying mechanisms in MI are underexplored. Exosomes play a crucial role in communication between cells, and can affect both homeostasis and disease conditions. Exosomal ncRNAs have been shown to affect many biological functions. Tissue-specific changes in exosomal ncRNAs contribute to aging, tissue dysfunction, and human diseases. Here we provide a comprehensive review of recent findings on epigenetic changes in cardiovascular diseases as well as the role of ncRNAs and exosomal ncRNAs in MI, focusing on their function, diagnostic and prognostic significance.
Collapse
Affiliation(s)
- Sara Fadaei
- Department of Internal Medicine and Endocrinology, Shohadae Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Zarepour
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Mehrnoosh Parvaresh
- Department of Physical Medicine and Rehabilitation, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| | - Alireza Motamedzadeh
- Department of Internal Medicine, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | | | - Amirhossein Sheida
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Shabani
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- Department of Anesthesiology, School of Allied Medical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Michael R. Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, South Africa
| | - Mehdi Rezaee
- Department of Anesthesiology, School of Medicine, Shahid Madani Hospital, Alborz University of Medical Sciences, Karaj, Iran
| | - Maryam Zarei
- Tehran Heart Center, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
15
|
Integrated Bioinformatics and Validation of lncRNA-Mediated ceRNA Network in Myocardial Ischemia/Reperfusion Injury. J Immunol Res 2022; 2022:7260801. [PMID: 36189147 PMCID: PMC9519285 DOI: 10.1155/2022/7260801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/05/2022] [Accepted: 08/01/2022] [Indexed: 11/26/2022] Open
Abstract
Background Myocardial ischemia/reperfusion (MI/R) injury is a common pathology in ischemia heart disease. Long noncoding RNAs (lncRNAs) are significant regulators related to many ischemia/reperfusion conditions. This study is aimed at exploring the molecule mechanism of lncRNA-mediated competing endogenous RNA (ceRNA) network in MI/R. Methods The dataset profiles of MI/R and normal tissues (GSE130217 and GSE124176) were obtained from the GEO database. Integrated bioinformatics were performed to screen out differentially expressed genes (DEGs). Thereafter, an lncRNA-mediated ceRNA network was constructed by the starBase database. The GO annotations and KEGG pathway analysis were conducted to study action mechanism and related pathways of DEGs in MI/R. A model of hypoxia/reoxygenation- (H/R-) treated HL-1 cell was performed to verify the expression of lncRNAs through qRT-PCR. Results 2406 differentially expressed- (DE-) mRNAs, 70 DE-lncRNAs, and 156 DE-miRNAs were acquired. These DEGs were conducted to construct an lncRNA-mediated ceRNA network, and a subnetwork including lncRNA Xist/miRNA-133c/mRNA (Slc30a9) was screen out. The functional enrichment analyses revealed that the lncRNAs involved in the ceRNA network might functions in oxidative stress and calcium signaling pathway. The lncRNA Xist expression is reduced under H/R conditions, followed by the increased level of miRNA-133c, thus downregulating the expression of Slc30a9. Conclusion In sum, the identified ceRNA network which included the lncRNA Xist/miR-133c/Slc30a9 axis might contribute a better understanding to the pathogenesis and development of MI/R injury and offer a novel targeted therapy way.
Collapse
|
16
|
Xiong J, Hu Y, Liu Y, Zeng X. CircRNA mmu_circ_0000021 regulates microvascular function via the miR-143-3p/NPY axis and intracellular calcium following ischemia/reperfusion injury. Cell Death Dis 2022; 8:315. [PMID: 35821018 PMCID: PMC9276824 DOI: 10.1038/s41420-022-01108-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 06/10/2022] [Accepted: 06/28/2022] [Indexed: 11/18/2022]
Abstract
Cardiac ischemia-reperfusion (I/R) is associated with a high rate of complications. Restoring microvascular function is crucial for cardiac repair. However, the molecular mechanisms by which the circRNAs repairs microvascular dysfunction are unknown. High-throughput RNA sequencing and quantitative real-time PCR (qRT-PCR) were used to measures circRNA levels in cardiac tissue samples. We found a total of 80 up-regulated and 54 down-regulated differentially expressed circRNAs, of which mmu_circ_0000021 were consistent with bioinformatics predictions. Next, mmu_circ_0000021 knockdown and overexpression were performed to indicate the functional role of mmu_circ_0000021. The interaction of mmu_circ_0000021, miR-143-3p and NPY were evaluated using dual-luciferase assays, RNA pull-down assays and RNA immunoprecipitation (RIP). Immunohistochemistry, transmission electron microscopy, and immunofluorescence were used to determine the presence of leukocytes and changes in microvascular morphology and function. Mechanistically, mmu_circ_0000021 involved in regulating microvascular dysfunction via miR-143-3p by targeting NPY. However, the contraction of microvascular spasm caused by NPY is related to calmodulin. By regulating NPY, Circular RNA (circRNA) further affects microvascular spasm, regulates microcirculation disorders, and restores cardiac function. Our findings highlight a novel role for mmu_circ_0000021 by regulating microvascular function following I/R injury.
Collapse
Affiliation(s)
- Jingjie Xiong
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Yisen Hu
- Guangxi Key Laboratory Base of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention & Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, Nanning, Guangxi, China
| | - Yi Liu
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Xiaocong Zeng
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China. .,Guangxi Key Laboratory Base of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention & Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, Nanning, Guangxi, China. .,School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China.
| |
Collapse
|
17
|
Wu F, Zhu Y, Zhou C, Gui W, Li H, Lin X. Regulation mechanism and pathogenic role of lncRNA plasmacytoma variant translocation 1 (PVT1) in human diseases. Genes Dis 2022. [DOI: 10.1016/j.gendis.2022.05.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
|
18
|
Li XT, Zhang YP, Zhang MW, Zhang ZZ, Zhong JC. Sirtuin 7 serves as a promising therapeutic target for cardiorenal diseases. Eur J Pharmacol 2022; 925:174977. [PMID: 35513019 DOI: 10.1016/j.ejphar.2022.174977] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 04/06/2022] [Accepted: 04/22/2022] [Indexed: 12/25/2022]
Abstract
Cardiovascular disorders and associated renal diseases account for the main cause of morbidity and mortality worldwide, necessitating the development of novel effective approaches for the prevention and treatment of cardiorenal diseases. Mammalian sirtuins (SIRTs) function as nicotinamide adenine dinucleotide (NAD+)-dependent protein/histone deacetylases. Seven members of SIRTs share a highly invariant catalytic core domain responsible for the specific enzymatic activity. Intriguingly, the broad distribution of SIRTs and alternative isoforms implicate its distinct functions in diverse cardiac and renal cells and tissue types. Notably, SIRT7 has been shown to exert beneficial effects in cardiorenal physiology and pathophysiology via modulation of senescence, DNA damage repair, ribosomal RNA synthesis, protein biosynthesis, angiogenesis, apoptosis, superoxide generation, cardiorenal metabolism, and dysfunction. Furthermore, SIRT7 has emerged as a critical modulator of a broad range of cellular activities including oxidative stress, inflammation response, endoplasmic reticulum stress, and mitochondrial homeostasis, which are all of great significance in postponing the progression of cardiorenal diseases. More importantly, SIRT7 has been implicated in cardiorenal hypertrophy, fibrosis, remodeling, heart failure, atherosclerosis as well as renal acid-base and electrolyte homeostasis as an essential regulator. In this review, we focus on the involvement in cardiorenal physiology and pathophysiology, diverse actions and underlying mechanisms of the SIRT7 signaling, highlighting its updated research progress in heart failure, atherosclerosis, diabetic nephropathy and other cardiorenal diseases. Targeting SIRT7 signaling could be potentially exploited as a therapeutic strategy aiming to prevent and treat cardiorenal diseases.
Collapse
Affiliation(s)
- Xue-Ting Li
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China; Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Ye-Ping Zhang
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China; Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Mi-Wen Zhang
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China; Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Zhen-Zhou Zhang
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China; Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China; Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Jiu-Chang Zhong
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China; Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China; Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China.
| |
Collapse
|
19
|
Xu H, Liu YY, Li LS, Liu YS. Sirtuins at the Crossroads between Mitochondrial Quality Control and Neurodegenerative Diseases: Structure, Regulation, Modifications, and Modulators. Aging Dis 2022; 14:794-824. [PMID: 37191431 DOI: 10.14336/ad.2022.1123] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 11/23/2022] [Indexed: 04/03/2023] Open
Abstract
Sirtuins (SIRT1-SIRT7), a family of nicotinamide adenine dinucleotide (NAD+)-dependent enzymes, are key regulators of life span and metabolism. In addition to acting as deacetylates, some sirtuins have the properties of deacylase, decrotonylase, adenosine diphosphate (ADP)-ribosyltransferase, lipoamidase, desuccinylase, demalonylase, deglutarylase, and demyristolyase. Mitochondrial dysfunction occurs early on and acts causally in the pathogenesis of neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington's disease (HD). Sirtuins are implicated in the regulation of mitochondrial quality control, which is highly associated with the pathogenesis of neurodegenerative diseases. There is growing evidence indicating that sirtuins are promising and well-documented molecular targets for the treatment of mitochondrial dysfunction and neurodegenerative disorders by regulating mitochondrial quality control, including mitochondrial biogenesis, mitophagy, mitochondrial fission/fusion dynamics, and mitochondrial unfolded protein responses (mtUPR). Therefore, elucidation of the molecular etiology of sirtuin-mediated mitochondrial quality control points to new prospects for the treatment of neurodegenerative diseases. However, the mechanisms underlying sirtuin-mediated mitochondrial quality control remain obscure. In this review, we update and summarize the current understanding of the structure, function, and regulation of sirtuins with an emphasis on the cumulative and putative effects of sirtuins on mitochondrial biology and neurodegenerative diseases, particularly their roles in mitochondrial quality control. In addition, we outline the potential therapeutic applications for neurodegenerative diseases of targeting sirtuin-mediated mitochondrial quality control through exercise training, calorie restriction, and sirtuin modulators in neurodegenerative diseases.
Collapse
|
20
|
Wang Y, Tan J, Wang L, Pei G, Cheng H, Zhang Q, Wang S, He C, Fu C, Wei Q. MiR-125 Family in Cardiovascular and Cerebrovascular Diseases. Front Cell Dev Biol 2021; 9:799049. [PMID: 34926475 PMCID: PMC8674784 DOI: 10.3389/fcell.2021.799049] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 11/11/2021] [Indexed: 02/05/2023] Open
Abstract
Cardiovascular and cerebrovascular diseases are a serious threaten to the health of modern people. Understanding the mechanism of occurrence and development of cardiovascular and cerebrovascular diseases, as well as reasonable prevention and treatment of them, is a huge challenge that we are currently facing. The miR-125 family consists of hsa-miR-125a, hsa-miR-125b-1 and hsa-miR-125b-2. It is a kind of miRNA family that is highly conserved among different species. A large amount of literature shows that the lack of miR-125 can cause abnormal development of the cardiovascular system in the embryonic period. At the same time, the miR-125 family participates in the occurrence and development of a variety of cardiovascular and cerebrovascular diseases, including myocardial ischemia, atherosclerosis, ischemia-reperfusion injury, ischemic stroke, and heart failure directly or indirectly. In this article, we summarized the role of the miR-125 family in the development and maturation of cardiovascular system, the occurrence and development of cardiovascular and cerebrovascular diseases, and its important value in the current fiery stem cell therapy. In addition, we presented this in the form of table and diagrams. We also discussed the difficulties and challenges faced by the miR-125 family in clinical applications.
Collapse
Affiliation(s)
- Yang Wang
- Department of Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, China
| | - Jing Tan
- Department of Ultrasound Medicine, Binzhou People's Hospital, Binzhou, China
| | - Lu Wang
- Department of Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, China
| | - Gaiqin Pei
- Department of Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, China
| | - Hongxin Cheng
- Department of Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, China
| | - Qing Zhang
- Department of Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, China
| | - Shiqi Wang
- Department of Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, China
| | - Chengqi He
- Department of Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, China
| | - Chenying Fu
- National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China.,Aging and Geriatric Mechanism Laboratory, West China Hospital, Sichuan University, Chengdu, China
| | - Quan Wei
- Department of Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, China
| |
Collapse
|
21
|
Wen ZJ, Xin H, Wang YC, Liu HW, Gao YY, Zhang YF. Emerging roles of circRNAs in the pathological process of myocardial infarction. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 26:828-848. [PMID: 34729251 PMCID: PMC8536508 DOI: 10.1016/j.omtn.2021.10.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Myocardial infarction (MI) is defined as cardiomyocyte death in a clinical context consistent with ischemic insult. MI remains one of the leading causes of morbidity and mortality worldwide. Although there are a number of effective clinical methods for the diagnosis and treatment of MI, further investigation of novel biomarkers and molecular therapeutic targets is required. Circular RNAs (circRNAs), novel non-coding RNAs, have been reported to function mainly by acting as microRNA (miRNA) sponges or binding to RNA-binding proteins (RBPs). The circRNA-miRNA-mRNA (protein) regulatory pathway regulates gene expression and affects the pathological mechanisms of various diseases. Undoubtedly, a more comprehensive understanding of the relationship between MI and circRNA will lay the foundation for the development of circRNA-based diagnostic and therapeutic strategies for MI. Therefore, this review summarizes the pathophysiological process of MI and various approaches to measure circRNA levels in MI patients, tissues, and cells; highlights the significance of circRNAs in the regulation MI pathogenesis and development; and provides potential clinical insight for the diagnosis, prognosis, and treatment of MI.
Collapse
Affiliation(s)
- Zeng-Jin Wen
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Hui Xin
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yong-Chen Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Hao-Wen Liu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Yan-Yan Gao
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Yin-Feng Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| |
Collapse
|
22
|
Ma J, Huang W, Zhu C, Sun X, Zhang Q, Zhang L, Qi Q, Bai X, Feng Y, Wang C. miR-423-3p activates FAK signaling pathway to drive EMT process and tumor growth in lung adenocarcinoma through targeting CYBRD1. J Clin Lab Anal 2021; 35:e24044. [PMID: 34714955 PMCID: PMC8649330 DOI: 10.1002/jcla.24044] [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] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/24/2021] [Accepted: 09/25/2021] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) is a malignant tumor with a high fatality rate and poor overall survival, while molecular targets diagnosing and alleviating lung cancer remain inadequate. METHODS In this article, we highlighted the upregulation of microRNA-423-3p (miR-423-3p) in LUAD, especially in smokers aged over 40, and revealed that the high expression of miR-423-3p was significantly associated with smoker, age, and pathologic stage of LUAD patients. RESULTS Moreover, overexpressing miR-423-3p could facilitate LUAD cell proliferation, invasion, adhesion, and epithelial-mesenchymal transition (EMT) process, while depleted miR-423-3p caused repressive influence upon it. Mechanically, we identified that miR-423-3p could activate FAK signaling pathway through binding to the 3'-UTR of cytochrome B reductase 1 (CYBRD1). Furthermore, we demonstrated that CYBRD1 was lowly expressed in LUAD, and miR-423-3p overexpression could rescue the impairment of LUAD cell proliferation, invasion, adhesion, and EMT caused by CYBRD1 depletion. Noticeably, miR-423-3p depletion efficiently hindered LUAD tumor growth in vivo. CONCLUSION Collectively, our findings demonstrated that miR-423-3p/CYBRD1 axis could be regarded as a promising biomarker to alleviate the poor LUAD prognosis.
Collapse
Affiliation(s)
- Jun Ma
- Department of Lung CancerTianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin’s Clinical Research Center for CancerTianjinChina
- Department of Thoracic SurgeryShanxi Provincial People’s HospitalTaiyuanChina
| | - Wuhao Huang
- Department of Lung CancerTianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin’s Clinical Research Center for CancerTianjinChina
| | - Chaonan Zhu
- Department of Thoracic SurgeryNorth China University of Science and Technology Affiliated HospitalTangshanChina
| | - Xiaoyan Sun
- Department of Lung CancerTianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin’s Clinical Research Center for CancerTianjinChina
| | - Qiang Zhang
- Department of Lung CancerTianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin’s Clinical Research Center for CancerTianjinChina
| | - Lianmin Zhang
- Department of Lung CancerTianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin’s Clinical Research Center for CancerTianjinChina
| | - Qi Qi
- Department of Lung CancerTianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin’s Clinical Research Center for CancerTianjinChina
| | - Xiaoming Bai
- Department of Thoracic SurgeryShanxi Provincial People’s HospitalTaiyuanChina
| | - Yun Feng
- Department of Thoracic SurgeryShanxi Provincial People’s HospitalTaiyuanChina
| | - Changli Wang
- Department of Lung CancerTianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin’s Clinical Research Center for CancerTianjinChina
| |
Collapse
|
23
|
Zhang H, Liu Y, Cao X, Wang W, Cui X, Yang X, Wang Y, Shi J. Nrf2 Promotes Inflammation in Early Myocardial Ischemia-Reperfusion via Recruitment and Activation of Macrophages. Front Immunol 2021; 12:763760. [PMID: 34917083 PMCID: PMC8669137 DOI: 10.3389/fimmu.2021.763760] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 11/10/2021] [Indexed: 12/20/2022] Open
Abstract
Cardiomyocyte apoptosis in response to inflammation is a primary cause of myocardial ischemia-reperfusion injury (IRI). Nuclear factor erythroid 2 like 2 (Nrf2) reportedly plays an important role in myocardial IRI, but the underlying mechanism remains obscure. Expression data from the normal heart tissues of mice or heart tissues treated with reperfusion for 6 h after ischemia (IR6h) were acquired from the GEO database; changes in biological function and infiltrating immune cells were analyzed. The binding between the molecules was verified by chromatin immunoprecipitation sequencing. Based on confirmation that early myocardial ischemia-reperfusion (myocardial ischemia/reperfusion for 6 hours, IR6h) promoted myocardial apoptosis and inflammatory response, we found that Nrf2, cooperating with Programmed Cell Death 4, promoted transcription initiation of C-C Motif Chemokine Ligand 3 (Ccl3) in myocardial tissues of mice treated with IR6h. Moreover, Ccl3 contributed to the high signature score of C-C motif chemokine receptor 1 (Ccr1)-positive macrophages. The high signature score of Ccr1-positive macrophages leads to the release of pro-inflammatory factors interleukin 1 beta and interleukin 6. This study is the first to elucidate the damaging effect of Nrf2 via remodeling of the immune microenvironment in early myocardial ischemia-reperfusion, which provides us with new perspectives and treatment strategies for myocardial ischemia-reperfusion.
Collapse
Affiliation(s)
- Haijian Zhang
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
- Department of Thoracic Surgery, Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, and Research Institution of Translational Medicine in Cardiothoracic Diseases in Affiliated Hospital of Nantong University, Nantong, China
- *Correspondence: Haijian Zhang, ; Jiahai Shi,
| | - Yifei Liu
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, China
| | - Xiaoqing Cao
- Department of Thoracic Surgery, Beijing Chest Hospital, Capital Medical University (Beijing Tuberculosis and Thoracic Tumor Research Institute), Beijing, China
| | - Wenmiao Wang
- Graduate School, Dalian Medical University, Dalian, China
| | - Xiaohong Cui
- Department of General Surgery, Shanghai Electric Power Hospital, Shanghai, China
| | - Xuechao Yang
- Department of Thoracic Surgery, Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, and Research Institution of Translational Medicine in Cardiothoracic Diseases in Affiliated Hospital of Nantong University, Nantong, China
| | - Yan Wang
- Department of Emergency, Affiliated Hospital of Nantong University, Nantong, China
| | - Jiahai Shi
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
- Department of Thoracic Surgery, Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, and Research Institution of Translational Medicine in Cardiothoracic Diseases in Affiliated Hospital of Nantong University, Nantong, China
- *Correspondence: Haijian Zhang, ; Jiahai Shi,
| |
Collapse
|