1
|
Zhang J. Non-coding RNAs and angiogenesis in cardiovascular diseases: a comprehensive review. Mol Cell Biochem 2024; 479:2921-2953. [PMID: 38306012 DOI: 10.1007/s11010-023-04919-5] [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: 10/27/2023] [Accepted: 12/18/2023] [Indexed: 02/03/2024]
Abstract
Non-coding RNAs (ncRNAs) have key roles in the etiology of many illnesses, including heart failure, myocardial infarction, stroke, and in physiological processes like angiogenesis. In transcriptional regulatory circuits that control heart growth, signaling, and stress response, as well as remodeling in cardiac disease, ncRNAs have become important players. Studies on ncRNAs and cardiovascular disease have made great progress recently. Here, we go through the functions of non-coding RNAs (ncRNAs) like circular RNAs (circRNAs), and microRNAs (miRNAs) as well as long non-coding RNAs (lncRNAs) in modulating cardiovascular disorders.
Collapse
Affiliation(s)
- Jie Zhang
- Medical School, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| |
Collapse
|
2
|
Zhou HM, Yue SJ, Wang WX, Zhang Q, Xu DQ, Li JJ, Tang YP, Yang XY. Exploring the effective compounds and potential mechanisms of Shengxian Decoction against coronary heart disease by UPLC-Q-TOF/MS and network pharmacology analysis. Heliyon 2024; 10:e29558. [PMID: 38681620 PMCID: PMC11046127 DOI: 10.1016/j.heliyon.2024.e29558] [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: 09/21/2023] [Revised: 04/08/2024] [Accepted: 04/10/2024] [Indexed: 05/01/2024] Open
Abstract
As a well-known classical Chinese medicine prescription, Shengxian Decoction (SXD) has been applied for a century to treat cardiovascular diseases, especially coronary heart disease (CHD), but the potentially effective compounds and underlying mechanisms remain unclear. With ultra-performance liquid chromatography-quadrupole-time of flight-mass spectrometry (UPLC-Q-TOF/MS) and network pharmacology analysis, the potential effective compounds of SXD and their pharmacological mechanisms against CHD were identified and revealed. 57 effective compounds with favorable pharmacokinetic characteristics and biological activities were screened through UPLC-Q-TOF/MS analysis, database and literature mining, interacting with 96 CHD-related targets to support potential synergistic therapeutic actions. Systematic analysis of the PPI network and microarray data further revealed six core targets, including TNF, IL-1β, IL-6, TP53, VEGFA and PTGS2, which were mainly involved in fluid shear stress and atherosclerosis, lipid and atherosclerosis, PI3K-Akt signaling pathway et al. Moreover, the proposed contribution indexes of effective compounds indicated these compounds, including isoferulic acid, quercetin, calycosin, ferulic acid, kaempferol, calycosin 7-O-glycoside, formononetin, astragaloside IV and saikosaponin D, as the core compounds of SXD. The molecular docking results confirmed that those core compound-target pairs exhibited strong binding energy. Furthermore, we validated that SXD significantly alleviated myocardial tissue injury in CHD rats and reversed H/R-induced decreases in H9c2 cell viability by attenuating the production of TNF, IL-6 and IL-1β, and reducing cardiomyocyte apoptosis via down-regulating the TP53, caspase3 and cytochrome C mRNA expression levels as well as caspase3, caspase9 and cytochrome C protein expression levels according to RT-qPCR and Western blot results. Our findings explained the pharmacological mechanisms underlying the effectiveness of SXD in the treatment of CHD, and laid a foundation for future basic and clinical research of SXD.
Collapse
Affiliation(s)
- Hao-ming Zhou
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, and State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, 712046, Shaanxi Province, China
| | - Shi-jun Yue
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, and State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, 712046, Shaanxi Province, China
- International Joint Research Center on Resource Utilization and Quality Evaluation of Traditional Chinese Medicine of Hebei Province, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Wen-xiao Wang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, and State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, 712046, Shaanxi Province, China
- International Joint Research Center on Resource Utilization and Quality Evaluation of Traditional Chinese Medicine of Hebei Province, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Qiao Zhang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, and State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, 712046, Shaanxi Province, China
| | - Ding-qiao Xu
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, and State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, 712046, Shaanxi Province, China
| | - Jia-jia Li
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, and State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, 712046, Shaanxi Province, China
| | - Yu-ping Tang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, and State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, 712046, Shaanxi Province, China
| | - Xin-yu Yang
- Department of Pharmacy, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
- Beijing Key Laboratory of Bio-characteristic Profiling for Evaluation of Rational Drug Use, Beijing, 100038, China
| |
Collapse
|
3
|
Chen L, Yang X, Wang K, Guo L, Zou C. Humanin inhibits lymphatic endothelial cells dysfunction to alleviate myocardial infarction-reperfusion injury via BNIP3-mediated mitophagy. Free Radic Res 2024; 58:180-193. [PMID: 38535980 DOI: 10.1080/10715762.2024.2333074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 02/20/2024] [Indexed: 05/02/2024]
Abstract
OBJECTIVE Acute myocardial infarction (AMI) ranks among the top contributors to sudden death and disability worldwide. It should be noted that current therapies always cause increased reperfusion damage. Evidence suggests that humanin (HN) reduces mitochondrial dysfunction to have cardio-protective effects against MI-reperfusion injury. In this context, we hypothesized that HN may attenuate MI-reperfusion injury by alleviating lymphatic endothelial cells dysfunction through the regulation of mitophagy. MATERIALS AND METHODS In this study, primary lymphatic endothelial cells were selected as the experimental model. Cells were maintained under 1% O2 to induce a hypoxic phenotype. For in vivo experiments, the left coronary arteries of C57/BL6 mice were clamped for 45 min followed by 24 h reperfusion to develop MI-reperfusion injury. The volume of infarcted myocardium in MI-reperfusion injury mouse models were TTC staining. PCR and western blot were used to quantify the expression of autophagy-, mitophagy- and mitochondria-related markers. The fibrosis and apoptosis in the ischemic area were evaluated for Masson staining and TUNEL respectively. We also used western blot to analyze the expression of VE-Cadherin in lymphatic endothelial cells. RESULTS We firstly exhibited a specific mechanism by which HN mitigates MI-reperfusion injury. We demonstrated that HN effectively reduces such injury in vivo and also inhibits dysfunction in lymphatic endothelial cells in vitro. Importantly, this inhibitory effect is mediated through BNIP3-associated mitophagy. CONCLUSIONS In conclusion, HN alleviates myocardial infarction-reperfusion injury by inhibiting lymphatic endothelial cells dysfunction, primarily through BNIP3-mediated mitophagy.
Collapse
Affiliation(s)
- Lu Chen
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Center for Cardiovascular Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaohua Yang
- Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Kai Wang
- Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Lina Guo
- Center for Cardiovascular Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Cao Zou
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
4
|
Chen C, Wang J, Liu C, Hu J, Liu L. Pioneering therapies for post-infarction angiogenesis: Insight into molecular mechanisms and preclinical studies. Biomed Pharmacother 2023; 166:115306. [PMID: 37572633 DOI: 10.1016/j.biopha.2023.115306] [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: 06/16/2023] [Revised: 08/01/2023] [Accepted: 08/07/2023] [Indexed: 08/14/2023] Open
Abstract
Acute myocardial infarction (MI), despite significant progress in its treatment, remains a leading cause of chronic heart failure and cardiovascular events such as cardiac arrest. Promoting angiogenesis in the myocardial tissue after MI to restore blood flow in the ischemic and hypoxic tissue is considered an effective treatment strategy. The repair of the myocardial tissue post-MI involves a robust angiogenic response, with mechanisms involved including endothelial cell proliferation and migration, capillary growth, changes in the extracellular matrix, and stabilization of pericytes for neovascularization. In this review, we provide a detailed overview of six key pathways in angiogenesis post-MI: the PI3K/Akt/mTOR signaling pathway, the Notch signaling pathway, the Wnt/β-catenin signaling pathway, the Hippo signaling pathway, the Sonic Hedgehog signaling pathway, and the JAK/STAT signaling pathway. We also discuss novel therapeutic approaches targeting these pathways, including drug therapy, gene therapy, protein therapy, cell therapy, and extracellular vesicle therapy. A comprehensive understanding of these key pathways and their targeted therapies will aid in our understanding of the pathological and physiological mechanisms of angiogenesis after MI and the development and application of new treatment strategies.
Collapse
Affiliation(s)
- Cong Chen
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| | - Jie Wang
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China.
| | - Chao Liu
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| | - Jun Hu
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| | - Lanchun Liu
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| |
Collapse
|
5
|
Salvatori F, D’Aversa E, Serino ML, Singh AV, Secchiero P, Zauli G, Tisato V, Gemmati D. miRNAs Epigenetic Tuning of Wall Remodeling in the Early Phase after Myocardial Infarction: A Novel Epidrug Approach. Int J Mol Sci 2023; 24:13268. [PMID: 37686073 PMCID: PMC10487654 DOI: 10.3390/ijms241713268] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
Myocardial infarction (MI) is one of the leading causes of death in Western countries. An early diagnosis decreases subsequent severe complications such as wall remodeling or heart failure and improves treatments and interventions. Novel therapeutic targets have been recognized and, together with the development of direct and indirect epidrugs, the role of non-coding RNAs (ncRNAs) yields great expectancy. ncRNAs are a group of RNAs not translated into a product and, among them, microRNAs (miRNAs) are the most investigated subgroup since they are involved in several pathological processes related to MI and post-MI phases such as inflammation, apoptosis, angiogenesis, and fibrosis. These processes and pathways are finely tuned by miRNAs via complex mechanisms. We are at the beginning of the investigation and the main paths are still underexplored. In this review, we provide a comprehensive discussion of the recent findings on epigenetic changes involved in the first phases after MI as well as on the role of the several miRNAs. We focused on miRNAs function and on their relationship with key molecules and cells involved in healing processes after an ischemic accident, while also giving insight into the discrepancy between males and females in the prognosis of cardiovascular diseases.
Collapse
Affiliation(s)
- Francesca Salvatori
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.S.)
| | - Elisabetta D’Aversa
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.S.)
| | - Maria Luisa Serino
- Centre Haemostasis & Thrombosis, University of Ferrara, 44121 Ferrara, Italy
| | - Ajay Vikram Singh
- Department of Chemical and Product Safety, German Federal Institute for Risk Assessment (BfR), 10589 Berlin, Germany
| | - Paola Secchiero
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.S.)
| | - Giorgio Zauli
- Department of Environmental Science and Prevention, University of Ferrara, 44121 Ferrara, Italy
| | - Veronica Tisato
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.S.)
- LTTA Centre, University of Ferrara, 44121 Ferrara, Italy
- University Centre for Studies on Gender Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Donato Gemmati
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.S.)
- Centre Haemostasis & Thrombosis, University of Ferrara, 44121 Ferrara, Italy
- University Centre for Studies on Gender Medicine, University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
6
|
Martín-Bórnez M, Falcón D, Morrugares R, Siegfried G, Khatib AM, Rosado JA, Galeano-Otero I, Smani T. New Insights into the Reparative Angiogenesis after Myocardial Infarction. Int J Mol Sci 2023; 24:12298. [PMID: 37569674 PMCID: PMC10418963 DOI: 10.3390/ijms241512298] [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: 07/03/2023] [Revised: 07/23/2023] [Accepted: 07/29/2023] [Indexed: 08/13/2023] Open
Abstract
Myocardial infarction (MI) causes massive loss of cardiac myocytes and injury to the coronary microcirculation, overwhelming the limited capacity of cardiac regeneration. Cardiac repair after MI is finely organized by complex series of procedures involving a robust angiogenic response that begins in the peri-infarcted border area of the infarcted heart, concluding with fibroblast proliferation and scar formation. Efficient neovascularization after MI limits hypertrophied myocytes and scar extent by the reduction in collagen deposition and sustains the improvement in cardiac function. Compelling evidence from animal models and classical in vitro angiogenic approaches demonstrate that a plethora of well-orchestrated signaling pathways involving Notch, Wnt, PI3K, and the modulation of intracellular Ca2+ concentration through ion channels, regulate angiogenesis from existing endothelial cells (ECs) and endothelial progenitor cells (EPCs) in the infarcted heart. Moreover, cardiac repair after MI involves cell-to-cell communication by paracrine/autocrine signals, mainly through the delivery of extracellular vesicles hosting pro-angiogenic proteins and non-coding RNAs, as microRNAs (miRNAs). This review highlights some general insights into signaling pathways activated under MI, focusing on the role of Ca2+ influx, Notch activated pathway, and miRNAs in EC activation and angiogenesis after MI.
Collapse
Affiliation(s)
- Marta Martín-Bórnez
- Group of Cardiovascular Pathophysiology, Institute of Biomedicine of Seville, University Hospital of Virgen del Rocío/University of Seville/CSIC, Avenida Manuel Siurot s/n, 41013 Seville, Spain; (M.M.-B.); (D.F.); (R.M.)
- Department of Medical Physiology and Biophysics, Faculty of Medicine, University of Seville, 41009 Seville, Spain
| | - Débora Falcón
- Group of Cardiovascular Pathophysiology, Institute of Biomedicine of Seville, University Hospital of Virgen del Rocío/University of Seville/CSIC, Avenida Manuel Siurot s/n, 41013 Seville, Spain; (M.M.-B.); (D.F.); (R.M.)
- Department of Medical Physiology and Biophysics, Faculty of Medicine, University of Seville, 41009 Seville, Spain
| | - Rosario Morrugares
- Group of Cardiovascular Pathophysiology, Institute of Biomedicine of Seville, University Hospital of Virgen del Rocío/University of Seville/CSIC, Avenida Manuel Siurot s/n, 41013 Seville, Spain; (M.M.-B.); (D.F.); (R.M.)
- Department of Medical Physiology and Biophysics, Faculty of Medicine, University of Seville, 41009 Seville, Spain
- Department of Cell Biology, Physiology and Immunology, Universidad de Córdoba, 14071 Córdoba, Spain
| | - Geraldine Siegfried
- RyTME, Bordeaux Institute of Oncology (BRIC)-UMR1312 Inserm, B2 Ouest, Allée Geoffroy St Hilaire CS50023, 33615 Pessac, France (A.-M.K.)
| | - Abdel-Majid Khatib
- RyTME, Bordeaux Institute of Oncology (BRIC)-UMR1312 Inserm, B2 Ouest, Allée Geoffroy St Hilaire CS50023, 33615 Pessac, France (A.-M.K.)
| | - Juan A. Rosado
- Cellular Physiology Research Group, Department of Physiology, Institute of Molecular Pathology Biomarkers (IMPB), University of Extremadura, 10003 Caceres, Spain;
| | - Isabel Galeano-Otero
- Group of Cardiovascular Pathophysiology, Institute of Biomedicine of Seville, University Hospital of Virgen del Rocío/University of Seville/CSIC, Avenida Manuel Siurot s/n, 41013 Seville, Spain; (M.M.-B.); (D.F.); (R.M.)
- Department of Medical Physiology and Biophysics, Faculty of Medicine, University of Seville, 41009 Seville, Spain
| | - Tarik Smani
- Group of Cardiovascular Pathophysiology, Institute of Biomedicine of Seville, University Hospital of Virgen del Rocío/University of Seville/CSIC, Avenida Manuel Siurot s/n, 41013 Seville, Spain; (M.M.-B.); (D.F.); (R.M.)
- Department of Medical Physiology and Biophysics, Faculty of Medicine, University of Seville, 41009 Seville, Spain
| |
Collapse
|
7
|
Liu H, Zhou Z, Deng H, Tian Z, Wu Z, Liu X, Ren Z, Jiang Z. Trim65 attenuates isoproterenol-induced cardiac hypertrophy by promoting autophagy and ameliorating mitochondrial dysfunction via the Jak1/Stat1 signaling pathway. Eur J Pharmacol 2023; 949:175735. [PMID: 37080331 DOI: 10.1016/j.ejphar.2023.175735] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 04/10/2023] [Accepted: 04/17/2023] [Indexed: 04/22/2023]
Abstract
Pathological cardiac hypertrophy is a major cause of heart failure, and there is no effective approach for its prevention or treatment. The Trim family is a recently identified family of E3 ubiquitin ligases that regulate cardiac hypertrophy. Trim65, which is a memberof the Trim family, previous studies have not determined whether Trim65 affects cardiac hypertrophy. In this study, the effects of Trim65 on isoproterenol (ISO)-induced cardiac hypertrophy and the underlying mechanisms were investigated. In contrast to C57BL/6 mice, Trim65-knockout (Trim65-KO) mice developed more severe myocardial hypertrophy, fibrosis and cardiac dysfunction after being intraperitoneally injected with ISO for 2 weeks. Transmission electron microscopy (TEM) revealed that the autophagic flux was inhibited, mitochondria were swollen, and mitochondrial cristae were lost or decreased in the myocardium of Trim65-KO mice. In vitro studies demonstrated that overexpression of Trim65 inhibited ISO-induced cardiomyocyte hypertrophy by increasing mitochondrial density and membrane potential, and the Stat1 inhibitor fludarabine attenuated the effect of Trim65 knockdown on ISO-induced cardiomyocyte hypertrophy by reducing Reactive oxygen species (ROS) production and increasing the mitochondrial density and membrane potential. Our findings provide the first link between Trim65 and mitochondria, and we found for the first time that Trim65 inhibits mitochondria-dependent apoptosis and autophagy via the Jak1/Stat1 signalling pathway, ultimately attenuating ISO-induced cardiac hypertrophy; this effect of Trim65 might be mediated via the regulation of Jak1 ubiquitination. Taking these findings together, we suggest that genes that are related to mitochondria-dependent apoptosis and that are associated with Trim65 could be promising therapeutic targets for cardiac hypertrophy.
Collapse
Affiliation(s)
- HuiTing Liu
- Hengyang Medical College, University of South China, Hengyang City, Hunan Province, 421001, PR China
| | - ZhiXiang Zhou
- Hengyang Medical College, University of South China, Hengyang City, Hunan Province, 421001, PR China
| | - HuaNian Deng
- Hengyang Medical College, University of South China, Hengyang City, Hunan Province, 421001, PR China
| | - Zhen Tian
- Hengyang Medical College, University of South China, Hengyang City, Hunan Province, 421001, PR China
| | - ZeFan Wu
- Hengyang Medical College, University of South China, Hengyang City, Hunan Province, 421001, PR China
| | - XiYan Liu
- Hengyang Medical College, University of South China, Hengyang City, Hunan Province, 421001, PR China
| | - Zhong Ren
- Hengyang Medical College, University of South China, Hengyang City, Hunan Province, 421001, PR China
| | - ZhiSheng Jiang
- Hengyang Medical College, University of South China, Hengyang City, Hunan Province, 421001, PR China.
| |
Collapse
|
8
|
Fan M, Yang K, Wang X, Chen L, Gill PS, Ha T, Liu L, Lewis NH, Williams DL, Li C. Lactate promotes endothelial-to-mesenchymal transition via Snail1 lactylation after myocardial infarction. SCIENCE ADVANCES 2023; 9:eadc9465. [PMID: 36735787 PMCID: PMC9897666 DOI: 10.1126/sciadv.adc9465] [Citation(s) in RCA: 100] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 01/03/2023] [Indexed: 06/01/2023]
Abstract
High levels of lactate are positively associated with the prognosis and mortality in patients with heart attack. Endothelial-to-mesenchymal transition (EndoMT) plays an important role in cardiac fibrosis. Here, we report that lactate exerts a previously unknown function that increases cardiac fibrosis and exacerbates cardiac dysfunction by promoting EndoMT following myocardial infarction (MI). Treatment of endothelial cells with lactate disrupts endothelial cell function and induces mesenchymal-like function following hypoxia by activating the TGF-β/Smad2 pathway. Mechanistically, lactate induces an association between CBP/p300 and Snail1, leading to lactylation of Snail1, a TGF-β transcription factor, through lactate transporter monocarboxylate transporter (MCT)-dependent signaling. Inhibiting Snail1 diminishes lactate-induced EndoMT and TGF-β/Smad2 activation after hypoxia/MI. The MCT inhibitor CHC mitigates lactate-induced EndoMT and Snail1 lactylation. Silence of MCT1 compromises lactate-promoted cardiac dysfunction and EndoMT after MI. We conclude that lactate acts as an important molecule that up-regulates cardiac EndoMT after MI via induction of Snail1 lactylation.
Collapse
Affiliation(s)
- Min Fan
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
- The Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Kun Yang
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
- The Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Xiaohui Wang
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
- The Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Linjian Chen
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, China
| | - P. Spencer Gill
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
- The Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Tuanzhu Ha
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
- The Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Li Liu
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Nicole H. Lewis
- Department of Medical Education, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - David L. Williams
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
- The Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Chuanfu Li
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
- The Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| |
Collapse
|
9
|
Xin C, Zhang J, Hao N, Wang J, Liu H, Wei H, Wang Y, Wang C, Wang S, Zheng C, Zhang Z, Jin Z. Irisin inhibits NLRP3 inflammasome activation in HG/HF incubated cardiac microvascular endothelial cells with H/R injury. Microcirculation 2022; 29:e12786. [PMID: 36151930 DOI: 10.1111/micc.12786] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 09/13/2022] [Accepted: 09/16/2022] [Indexed: 12/30/2022]
Abstract
PURPOSE NLRP3 inflammasome mediates myocardial ischemia/reperfusion (MI/R) injury and diabetic vascular endothelia dysfunction. However, the role of NLRP3 inflammasome in MI/R injury with diabetes has not been fully described. Irisin plays an important role in anti-inflammation and improves endothelial function in type 2 diabetes. The current study aimed to investigate the effect of irisin on regulating NLRP3 inflammasome activation in diabetic vascular endothelia dysfunction. METHODS Cardiac microvascular endothelial cells (CMECs) were cultured and subjected to high glucose/high fat (HG/HF) receiving hypoxia/reoxygenation (H/R) with irisin incubation or not. Then, apoptosis, viability, migration, NO secretion, and inflammasome activation were examined. RESULTS The hypoxic CMECs exhibited increased apoptosis, impaired viability, and migration, even decreased NO secretion and enhanced inflammasome activation. Moreover, irisin incubation decreased NLRP3 activation and attenuated cell injury in HG/HF cultured CMECs subjected to H/R injury, which was abolished by NLRP3 inflammasome activation. Meanwhile, NLRP3 inflammasome siRNA also attenuated H/R injury in CMECs under HG/HF condition. CONCLUSION The current study demonstrated for the first time that irisin inhibits NLRP3 inflammasome activation in CMECs as a novel mechanism in myocardial ischemia/reperfusion injury in diabetes.
Collapse
Affiliation(s)
- Chao Xin
- PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Jinglong Zhang
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Ningbo Hao
- PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Jianan Wang
- PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Hui Liu
- PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Hanwen Wei
- PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Yong Wang
- The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Chengzhu Wang
- PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Shuo Wang
- PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Chengrong Zheng
- PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Zheng Zhang
- PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Zhitao Jin
- PLA Rocket Force Characteristic Medical Center, Beijing, China
| |
Collapse
|
10
|
Csöbönyeiová M, Beerová N, Klein M, Debreová-Čeháková M, Danišovič Ľ. Cell-Based and Selected Cell-Free Therapies for Myocardial Infarction: How Do They Compare to the Current Treatment Options? Int J Mol Sci 2022; 23:10314. [PMID: 36142245 PMCID: PMC9499607 DOI: 10.3390/ijms231810314] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/03/2022] [Accepted: 09/05/2022] [Indexed: 11/17/2022] Open
Abstract
Because of cardiomyocyte death or dysfunction frequently caused by myocardial infarction (MI), heart failure is a leading cause of morbidity and mortality in modern society. Paradoxically, only limited and non-curative therapies for heart failure or MI are currently available. As a result, over the past two decades research has focused on developing cell-based approaches promoting the regeneration of infarcted tissue. Cell-based therapies for myocardial regeneration include powerful candidates, such as multipotent stem cells (mesenchymal stem cells (MSCs), bone-marrow-derived stem cells, endothelial progenitor cells, and hematopoietic stem cells) and induced pluripotent stem cells (iPSCs). These possess unique properties, such as potency to differentiate into desired cell types, proliferation capacity, and patient specificity. Preclinical and clinical studies have demonstrated modest improvement in the myocardial regeneration and reduced infarcted areas upon transplantation of pluripotent or multipotent stem cells. Another cell population that need to be considered as a potential source for cardiac regeneration are telocytes found in different organs, including the heart. Their therapeutic effect has been studied in various heart pathologies, such as MI, arrhythmias, or atrial amyloidosis. The most recent cell-free therapeutic tool relies on the cardioprotective effect of complex cargo carried by small membrane-bound vesicles-exosomes-released from stem cells via exocytosis. The MSC/iPSC-derived exosomes could be considered a novel exosome-based therapy for cardiovascular diseases thanks to their unique content. There are also other cell-free approaches, e.g., gene therapy, or acellular cardiac patches. Therefore, our review provides the most recent insights into the novel strategies for myocardial repair based on the regenerative potential of different cell types and cell-free approaches.
Collapse
Affiliation(s)
- Mária Csöbönyeiová
- National Institute of Rheumatic Diseases, Nábrežie I. Krasku 4, 921 12 Piešťany, Slovakia
- Institute of Histology and Embryology, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Nikoleta Beerová
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Martin Klein
- National Institute of Rheumatic Diseases, Nábrežie I. Krasku 4, 921 12 Piešťany, Slovakia
- Institute of Histology and Embryology, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Michaela Debreová-Čeháková
- National Institute of Rheumatic Diseases, Nábrežie I. Krasku 4, 921 12 Piešťany, Slovakia
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Ľuboš Danišovič
- National Institute of Rheumatic Diseases, Nábrežie I. Krasku 4, 921 12 Piešťany, Slovakia
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| |
Collapse
|
11
|
Xin H, Li C, Cai T, Cao J, Wang M. LncRNA KCNQ1OT1 contributes to hydrogen peroxide-induced apoptosis, inflammation, and oxidative stress of cardiomyocytes via miR-130a-3p/ZNF791 axis. Cell Biol Int 2022; 46:2018-2027. [PMID: 35989482 DOI: 10.1002/cbin.11873] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 11/08/2022]
Abstract
It has been reported that long noncoding RNA (lncRNA) KCNQ1 opposite strand/antisense transcript 1 (KCNQ1OT1) played an important role in myocardial infarction (MI). However, the regulatory network behind KCNQ1OT1 in MI is largely unknown. Quantitative real time polymerase chain reaction (qRT-PCR) was applied to detect the enrichment of KCNQ1OT1, microRNA-130a-3p (miR-130a-3p) and zinc finger 791 (ZNF791). The viability and apoptosis of AC16 cells were measured by (4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and flow cytometry. Enzyme-linked immunosorbent assay (ELISA) was conducted to assess the inflammation and oxidative stress status of AC16 cells. The targeted relationship between miR-130a-3p and KCNQ1OT1 or ZNF791 was predicted by StarBase bioinformatic database, and dual-luciferase reporter assay and RNA immunoprecipitation (RIP) assay were carried out to verify these predictions. Hydrogen peroxide (H2 O2 ) stimulation caused a significant upregulation in the expression of KCNQ1OT1, while the level of miR-130a-3p showed an opposite phenomenon. KCNQ1OT1 was a crucial downstream component in H2 O2 -mediated toxic effects, and KCNQ1OT1 accelerated H2 O2 -induced toxic effects in AC16 cells. KCNQ1OT1 could sponge miR-130a-3p and down-regulate its expression. MiR-130a-3p exerted opposite effects to KCNQ1OT1, and the depletion of miR-130a-3p attenuated the protective effects of KCNQ1OT1 intervention on AC16 cells exposed to H2 O2 . MiR-130a-3p could bind to ZNF791, and ZNF791 served as the target of miR-130a-3p to promote H2 O2 -induced injury of AC16 cells. ZNF791 was modulated by KCNQ1OT1/miR-130a-3p signaling in H2 O2 -treated AC16 cells. In all, lncRNA KCNQ1OT1 deteriorated H2 O2 -mediated injury in cardiomyocytes through upregulating ZNF791 via serving as a molecular sponge for miR-130a-3p.
Collapse
Affiliation(s)
- Hong Xin
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi 'an Medical University, Xi 'an, Shaanxi, China
| | - Chengliang Li
- Department of General Practice, The First Affiliated Hospital of Xi 'an Medical University, Xi 'an, Shaanxi, China
| | - Tianzhi Cai
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi 'an Medical University, Xi 'an, Shaanxi, China
| | - Jinlong Cao
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi 'an Medical University, Xi 'an, Shaanxi, China
| | - Meixue Wang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi 'an Medical University, Xi 'an, Shaanxi, China
| |
Collapse
|
12
|
Walkowski B, Kleibert M, Majka M, Wojciechowska M. Insight into the Role of the PI3K/Akt Pathway in Ischemic Injury and Post-Infarct Left Ventricular Remodeling in Normal and Diabetic Heart. Cells 2022; 11:cells11091553. [PMID: 35563860 PMCID: PMC9105930 DOI: 10.3390/cells11091553] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/28/2022] [Accepted: 05/03/2022] [Indexed: 02/07/2023] Open
Abstract
Despite the significant decline in mortality, cardiovascular diseases are still the leading cause of death worldwide. Among them, myocardial infarction (MI) seems to be the most important. A further decline in the death rate may be achieved by the introduction of molecularly targeted drugs. It seems that the components of the PI3K/Akt signaling pathway are good candidates for this. The PI3K/Akt pathway plays a key role in the regulation of the growth and survival of cells, such as cardiomyocytes. In addition, it has been shown that the activation of the PI3K/Akt pathway results in the alleviation of the negative post-infarct changes in the myocardium and is impaired in the state of diabetes. In this article, the role of this pathway was described in each step of ischemia and subsequent left ventricular remodeling. In addition, we point out the most promising substances which need more investigation before introduction into clinical practice. Moreover, we present the impact of diabetes and widely used cardiac and antidiabetic drugs on the PI3K/Akt pathway and discuss the molecular mechanism of its effects on myocardial ischemia and left ventricular remodeling.
Collapse
Affiliation(s)
- Bartosz Walkowski
- Laboratory of Centre for Preclinical Research, Department of Experimental and Clinical Physiology, Medical University of Warsaw, Banacha 1b, 02-097 Warsaw, Poland; (B.W.); (M.W.)
| | - Marcin Kleibert
- Laboratory of Centre for Preclinical Research, Department of Experimental and Clinical Physiology, Medical University of Warsaw, Banacha 1b, 02-097 Warsaw, Poland; (B.W.); (M.W.)
- Correspondence: (M.K.); (M.M.)
| | - Miłosz Majka
- Laboratory of Centre for Preclinical Research, Department of Experimental and Clinical Physiology, Medical University of Warsaw, Banacha 1b, 02-097 Warsaw, Poland; (B.W.); (M.W.)
- Correspondence: (M.K.); (M.M.)
| | - Małgorzata Wojciechowska
- Laboratory of Centre for Preclinical Research, Department of Experimental and Clinical Physiology, Medical University of Warsaw, Banacha 1b, 02-097 Warsaw, Poland; (B.W.); (M.W.)
- Invasive Cardiology Unit, Independent Public Specialist Western Hospital John Paul II, Daleka 11, 05-825 Grodzisk Mazowiecki, Poland
| |
Collapse
|
13
|
Plasma-derived extracellular vesicles transfer microRNA-130a-3p to alleviate myocardial ischemia/reperfusion injury by targeting ATG16L1. Cell Tissue Res 2022; 389:99-114. [PMID: 35503135 DOI: 10.1007/s00441-022-03605-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 02/24/2022] [Indexed: 11/02/2022]
Abstract
Extracellular vesicles (EVs) are implicated in myocardial ischemia/reperfusion (I/R) injury as modulators by shuttling diverse cargoes, including microRNAs (miRNAs). The current study was initiated to unravel the potential involvement of plasma-derived EVs carrying miR-130a-3p on myocardial I/R injury. Rats were induced with moderate endoplasmic reticulum stress, followed by isolation of plasma-derived EVs. Then, an I/R rat model and hypoxia/reoxygenation (H/R) cardiomyoblast model were established to simulate a myocardial I/R injury environment where miR-130a-3p was found to be abundantly expressed. miR-130a-3p was confirmed to target and negatively regulate autophagy-related 16-like 1 (ATG16L1) in cardiomyoblasts. Based on a co-culture system, miR-130a-3p delivered by EVs derived from plasma protected H/R-exposed cardiomyoblasts against H/R-induced excessive cardiomyoblast autophagy, inflammation, and damage, improving cardiac dysfunction as well as myocardial I/R-induced cardiac dysfunction and tissue injury. The mechanism underlying the functional role of EVs-loaded miR-130a-3p was found to be dependent on its targeting relation with ATG16L1. The protective action of EV-carried miR-130a-3p was further re-produced in a rat model serving as in vivo validation as evidenced by improved cardiac function, tissue injury, myocardial fibrosis, and myocardial infarction. Collectively, miR-130a-3p shuttled by plasma-derived EVs was demonstrated to alleviate excessive cardiomyoblast autophagy and improve myocardial I/R injury.
Collapse
|
14
|
Varzideh F, Kansakar U, Donkor K, Wilson S, Jankauskas SS, Mone P, Wang X, Lombardi A, Santulli G. Cardiac Remodeling After Myocardial Infarction: Functional Contribution of microRNAs to Inflammation and Fibrosis. Front Cardiovasc Med 2022; 9:863238. [PMID: 35498051 PMCID: PMC9043126 DOI: 10.3389/fcvm.2022.863238] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 03/08/2022] [Indexed: 01/12/2023] Open
Abstract
After an ischemic injury, the heart undergoes a complex process of structural and functional remodeling that involves several steps, including inflammatory and fibrotic responses. In this review, we are focusing on the contribution of microRNAs in the regulation of inflammation and fibrosis after myocardial infarction. We summarize the most updated studies exploring the interactions between microRNAs and key regulators of inflammation and fibroblast activation and we discuss the recent discoveries, including clinical applications, in these rapidly advancing fields.
Collapse
Affiliation(s)
- Fahimeh Varzideh
- Department of Medicine, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Aging Research, New York, NY, United States
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Wilf Family Cardiovascular Research Institute, Institute for Neuroimmunology and Inflammation (INI), New York, NY, United States
| | - Urna Kansakar
- Department of Medicine, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Aging Research, New York, NY, United States
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Wilf Family Cardiovascular Research Institute, Institute for Neuroimmunology and Inflammation (INI), New York, NY, United States
| | - Kwame Donkor
- Department of Medicine, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Aging Research, New York, NY, United States
| | - Scott Wilson
- Department of Medicine, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Aging Research, New York, NY, United States
| | - Stanislovas S. Jankauskas
- Department of Medicine, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Aging Research, New York, NY, United States
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Wilf Family Cardiovascular Research Institute, Institute for Neuroimmunology and Inflammation (INI), New York, NY, United States
| | - Pasquale Mone
- Department of Medicine, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Aging Research, New York, NY, United States
| | - Xujun Wang
- Department of Medicine, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Aging Research, New York, NY, United States
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Wilf Family Cardiovascular Research Institute, Institute for Neuroimmunology and Inflammation (INI), New York, NY, United States
| | - Angela Lombardi
- Department of Medicine, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Aging Research, New York, NY, United States
| | - Gaetano Santulli
- Department of Medicine, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Aging Research, New York, NY, United States
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Wilf Family Cardiovascular Research Institute, Institute for Neuroimmunology and Inflammation (INI), New York, NY, United States
- *Correspondence: Gaetano Santulli,
| |
Collapse
|
15
|
Ai X, Yu P, Peng L, Luo L, Liu J, Li S, Lai X, Luan F, Meng X. Berberine: A Review of its Pharmacokinetics Properties and Therapeutic Potentials in Diverse Vascular Diseases. Front Pharmacol 2022; 12:762654. [PMID: 35370628 PMCID: PMC8964367 DOI: 10.3389/fphar.2021.762654] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/20/2021] [Indexed: 12/11/2022] Open
Abstract
Traditional Chinese medicine plays a significant role in the treatment of various diseases and has attracted increasing attention for clinical applications. Vascular diseases affecting vasculature in the heart, cerebrovascular disease, atherosclerosis, and diabetic complications have compromised quality of life for affected individuals and increase the burden on health care services. Berberine, a naturally occurring isoquinoline alkaloid form Rhizoma coptidis, is widely used in China as a folk medicine for its antibacterial and anti-inflammatory properties. Promisingly, an increasing number of studies have identified several cellular and molecular targets for berberine, indicating its potential as an alternative therapeutic strategy for vascular diseases, as well as providing novel evidence that supports the therapeutic potential of berberine to combat vascular diseases. The purpose of this review is to comprehensively and systematically describe the evidence for berberine as a therapeutic agent in vascular diseases, including its pharmacological effects, molecular mechanisms, and pharmacokinetics. According to data published so far, berberine shows remarkable anti-inflammatory, antioxidant, antiapoptotic, and antiautophagic activity via the regulation of multiple signaling pathways, including AMP-activated protein kinase (AMPK), nuclear factor κB (NF-κB), mitogen-activated protein kinase silent information regulator 1 (SIRT-1), hypoxia-inducible factor 1α (HIF-1α), vascular endothelial growth factor phosphoinositide 3-kinase (PI3K), protein kinase B (Akt), janus kinase 2 (JAK-2), Ca2+ channels, and endoplasmic reticulum stress. Moreover, we discuss the existing limitations of berberine in the treatment of vascular diseases, and give corresponding measures. In addition, we propose some research perspectives and challenges, and provide a solid evidence base from which further studies can excavate novel effective drugs from Chinese medicine monomers.
Collapse
Affiliation(s)
- Xiaopeng Ai
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Peiling Yu
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Lixia Peng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Liuling Luo
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jia Liu
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shengqian Li
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xianrong Lai
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fei Luan
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xianli Meng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
16
|
Nikolajevic J, Ariaee N, Liew A, Abbasnia S, Fazeli B, Sabovic M. The Role of MicroRNAs in Endothelial Cell Senescence. Cells 2022; 11:cells11071185. [PMID: 35406749 PMCID: PMC8997793 DOI: 10.3390/cells11071185] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/24/2022] [Accepted: 03/30/2022] [Indexed: 12/05/2022] Open
Abstract
Cellular senescence is a complex, dynamic process consisting of the irreversible arrest of growth and gradual deterioration of cellular function. Endothelial senescence affects the cell’s ability to repair itself, which is essential for maintaining vascular integrity and leads to the development of endothelial dysfunction, which has an important role in the pathogenesis of cardiovascular diseases. Senescent endothelial cells develop a particular, senescence-associated secretory phenotype (SASP) that detrimentally affects both surrounding and distant endothelial cells, thereby facilitating the ageing process and development of age-related disorders. Recent studies highlight the role of endothelial senescence and its dysfunction in the pathophysiology of several age-related diseases. MicroRNAs are small noncoding RNAs that have an important role in the regulation of gene expression at the posttranscriptional level. Recently, it has been discovered that miRNAs could importantly contribute to endothelial cell senescence. Overall, the research focus has been shifting to new potential mechanisms and targets to understand and prevent the structural and functional changes in ageing senescent endothelial cells in order to prevent the development and limit the progression of the wide spectrum of age-related diseases. The aim of this review is to provide some insight into the most important pathways involved in the modulation of endothelial senescence and to reveal the specific roles of several miRNAs involved in this complex process. Better understanding of miRNA’s role in endothelial senescence could lead to new approaches for prevention and possibly also for the treatment of endothelial cells ageing and associated age-related diseases.
Collapse
Affiliation(s)
- Jovana Nikolajevic
- Department of Vascular Diseases, University Medical Center, 1000 Ljubljana, Slovenia;
- Correspondence:
| | - Nazila Ariaee
- Allergy Research Center, Mashhad University of Medical Sciences, Mashhad 1696700, Iran;
| | - Aaron Liew
- Department of Medicine, National University of Galway, H91 CF50 Galway, Ireland;
| | - Shadi Abbasnia
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad 1696700, Iran;
| | - Bahare Fazeli
- Vascular Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad 1696700, Iran;
| | - Miso Sabovic
- Department of Vascular Diseases, University Medical Center, 1000 Ljubljana, Slovenia;
- Medical Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia
| |
Collapse
|
17
|
Signaling pathways and targeted therapy for myocardial infarction. Signal Transduct Target Ther 2022; 7:78. [PMID: 35273164 PMCID: PMC8913803 DOI: 10.1038/s41392-022-00925-z] [Citation(s) in RCA: 285] [Impact Index Per Article: 95.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 01/28/2022] [Accepted: 02/08/2022] [Indexed: 02/07/2023] Open
Abstract
Although the treatment of myocardial infarction (MI) has improved considerably, it is still a worldwide disease with high morbidity and high mortality. Whilst there is still a long way to go for discovering ideal treatments, therapeutic strategies committed to cardioprotection and cardiac repair following cardiac ischemia are emerging. Evidence of pathological characteristics in MI illustrates cell signaling pathways that participate in the survival, proliferation, apoptosis, autophagy of cardiomyocytes, endothelial cells, fibroblasts, monocytes, and stem cells. These signaling pathways include the key players in inflammation response, e.g., NLRP3/caspase-1 and TLR4/MyD88/NF-κB; the crucial mediators in oxidative stress and apoptosis, for instance, Notch, Hippo/YAP, RhoA/ROCK, Nrf2/HO-1, and Sonic hedgehog; the controller of myocardial fibrosis such as TGF-β/SMADs and Wnt/β-catenin; and the main regulator of angiogenesis, PI3K/Akt, MAPK, JAK/STAT, Sonic hedgehog, etc. Since signaling pathways play an important role in administering the process of MI, aiming at targeting these aberrant signaling pathways and improving the pathological manifestations in MI is indispensable and promising. Hence, drug therapy, gene therapy, protein therapy, cell therapy, and exosome therapy have been emerging and are known as novel therapies. In this review, we summarize the therapeutic strategies for MI by regulating these associated pathways, which contribute to inhibiting cardiomyocytes death, attenuating inflammation, enhancing angiogenesis, etc. so as to repair and re-functionalize damaged hearts.
Collapse
|
18
|
G protein-coupled receptor Mas induces an inhibitory effect on myocardial infarction-induced myocardial injury. Int J Biol Macromol 2022; 207:72-80. [PMID: 35247425 DOI: 10.1016/j.ijbiomac.2022.02.163] [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: 09/27/2021] [Revised: 09/28/2021] [Accepted: 02/25/2022] [Indexed: 11/22/2022]
Abstract
Myocardial infarction (MI) is the most prevalent disease with high mortality, leading to devastating heart injury. Here, we aimed to explore the effect of MAS1 on the MI-induced myocardial injury. Significantly, we identified that the expression of MAS1 was decreased in the MI rat model and hypoxia and reoxygenation (H/R)-treated H9C2 cells. Hematoxylin & Eosin (H&E) staining revealed that the overexpression of MAS1 notably attenuated MI-related myocardium injury in the MI rat model. Echocardiography analysis revealed that MI inhibited left ventricular ejection fraction (LVEF) and left ventricular fraction shortening (LVFS), whereas the MAS1 overexpression could increase LVEF and LVFS in the MI rats. Meanwhile, the expression of collagen I, collagen III, α-SMA, ANP, and BNP was decreased by MAS1 overexpression in the MI rats. MAS1 attenuated cardiomyocyte apoptosis in vivo and in vitro. Mechanically, the overexpression of MAS1 decreased the expression of PTEN and enhanced the phosphorylation of PI3K and AKT in vivo and in vitro. The overexpression of PTEN and the PI3k inhibitor LY294002 could reverse the MAS1-mediated MI injury. Thus, we conclude that MAS1 inhibits MI-induced myocardial injury by modulating PTEN/PI3K/AKT signaling. Our finding provides new insight into the mechanism by which MAS1 attenuates MI-related cardiac dysfunction.
Collapse
|
19
|
Bei Y, Lu D, Bär C, Chatterjee S, Costa A, Riedel I, Mooren FC, Zhu Y, Huang Z, Wei M, Hu M, Liu S, Yu P, Wang K, Thum T, Xiao J. MiR-486 attenuates cardiac ischemia/reperfusion injury and mediates the beneficial effect of exercise for myocardial protection. Mol Ther 2022; 30:1675-1691. [PMID: 35077859 PMCID: PMC9077322 DOI: 10.1016/j.ymthe.2022.01.031] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 10/23/2021] [Accepted: 01/20/2022] [Indexed: 10/19/2022] Open
Abstract
Exercise and its regulated molecules have myocardial protective effects against cardiac ischemia/reperfusion (I/R) injury. The muscle-enriched miR-486 was previously identified to be upregulated in exercised heart, which prompted us to investigate the functional roles of miR-486 in cardiac I/R injury and to further explore its potential in contributing to exercise-induced protection against I/R injury. Our data showed that miR-486 was significantly downregulated in the heart upon cardiac I/R injury. Both preventive and therapeutic interventions of adeno-associated virus 9 (AAV9)-mediated miR-486 overexpression could reduce cardiac I/R injury. Using AAV9 expressing miR-486 with cTnT promoter, we further demonstrated that cardiac muscle cell-targeted miR-486 overexpression was also sufficient to protect against cardiac I/R injury. Consistently, miR-486 was downregulated in oxygen glucose deprivation/reperfusion (OGDR)-stressed cardiomyocytes, while upregulating miR-486 inhibited cardiomyocyte apoptosis through PTEN and FoxO1 inhibition and AKT/mTOR activation. Finally, we observed that miR-486 was necessary for exercise-induced protection against cardiac I/R injury. In conclusion, miR-486 is protective against cardiac I/R injury and myocardial apoptosis through targeting PTEN and FoxO1 and activation of the AKT/mTOR pathway, and mediates the beneficial effect of exercise for myocardial protection. Increasing miR-486 might be a promising therapeutic strategy for myocardial protection.
Collapse
Affiliation(s)
- Yihua Bei
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China; Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Dongchao Lu
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover 30625, Germany; REBIRTH Center for Translational Regenerative Medicine, Hannover Medical School, Hannover 30625, Germany
| | - Christian Bär
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover 30625, Germany; REBIRTH Center for Translational Regenerative Medicine, Hannover Medical School, Hannover 30625, Germany
| | - Shambhabi Chatterjee
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover 30625, Germany
| | - Alessia Costa
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover 30625, Germany; REBIRTH Center for Translational Regenerative Medicine, Hannover Medical School, Hannover 30625, Germany
| | - Isabelle Riedel
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover 30625, Germany
| | - Frank C Mooren
- Witten/Herdecke University, Faculty of Health/School of Medicine, Witten 58448, Germany
| | - Yujiao Zhu
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China; Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Zhenzhen Huang
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China; Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Meng Wei
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China; Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Meiyu Hu
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China; Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Sunyi Liu
- Department of Cardiology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Pujiao Yu
- Department of Cardiology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Kun Wang
- Department of Cardio-thoracic Surgery, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover 30625, Germany; REBIRTH Center for Translational Regenerative Medicine, Hannover Medical School, Hannover 30625, Germany; Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover 30625, Germany.
| | - Junjie Xiao
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China; Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
20
|
Wu C, Liu B, Wang R, Li G. The Regulation Mechanisms and Clinical Application of MicroRNAs in Myocardial Infarction: A Review of the Recent 5 Years. Front Cardiovasc Med 2022; 8:809580. [PMID: 35111829 PMCID: PMC8801508 DOI: 10.3389/fcvm.2021.809580] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 12/24/2021] [Indexed: 12/21/2022] Open
Abstract
Myocardial infarction (MI) is the most frequent end-point of cardiovascular pathology, leading to higher mortality worldwide. Due to the particularity of the heart tissue, patients who experience ischemic infarction of the heart, still suffered irreversible damage to the heart even if the vascular reflow by treatment, and severe ones can lead to heart failure or even death. In recent years, several studies have shown that microRNAs (miRNAs), playing a regulatory role in damaged hearts, bring light for patients to alleviate MI. In this review, we summarized the effect of miRNAs on MI with some mechanisms, such as apoptosis, autophagy, proliferation, inflammatory; the regulation of miRNAs on cardiac structural changes after MI, including angiogenesis, myocardial remodeling, fibrosis; the application of miRNAs in stem cell therapy and clinical diagnosis; other non-coding RNAs related to miRNAs in MI during the past 5 years.
Collapse
|
21
|
Wang H, Xu J, Wu X, Zheng S, Han Y, Ding G. Longitudinal change in microRNA-130a expression and its correlation with the risk of developing major adverse cardiovascular and cerebral events in patients undergoing continuous ambulatory peritoneal dialysis. J Clin Lab Anal 2021; 35:e24039. [PMID: 34708454 DOI: 10.1002/jcla.24039] [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: 07/28/2021] [Revised: 09/22/2021] [Accepted: 09/23/2021] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND MicroRNA-130a (miR-130a) regulates angio-cellular dysregulation, atherosclerosis, and cardiocerebral injuries, serving as a biomarker for major adverse cardiovascular and cerebral events (MACCE) in several chronic diseases. However, its clinical application in patients with end-stage renal disease (ESRD) undergoing continuous ambulatory peritoneal dialysis (CAPD), who are at a high risk of developing MACCE, has not been reported. Therefore, this study aimed to explore this aspect. METHODS miR-130a expression in peripheral blood mononuclear cells obtained from 50 healthy controls (HCs) at recruitment and 257 ESRD patients undergoing CAPD at month (M)0, M12, M24, and M36 was determined by reverse transcription-quantitative polymerase chain reaction. ESRD patients undergoing CAPD were followed up until MACCE occurred or M36. Then, MACCE were recorded, and MACCE-free survival was calculated. RESULTS miR-130a expression was significantly lower in ESRD patients undergoing CAPD than in HCs (p < 0.001). In addition, miR-130a expression significantly decreased from M0 to M36 in ESRD patients undergoing CAPD (p < 0.001). Moreover, miR-130a expression at M0, M12, and M24 was significantly lower in patients with MACCE than in those without MACCE (all p < 0.05). Furthermore, high miR-130a expression at M0, M12, and M36 was significantly correlated with prolonged MACCE-free survival in ESRD patients undergoing CAPD (all p < 0.05), and high miR-130a expression at M0 was an independent factor for improved MACCE-free survival (p = 0.015; hazard ratio (HR) (95% confidential interval): 0.456 (0.243-0.857)). CONCLUSION miR-130a expression decreases continuously with disease progression in patients with ESRD undergoing CAPD. Additionally, this expression is negatively correlated with MACCE risk in these patients.
Collapse
Affiliation(s)
- Hong Wang
- Department of Nephrology, Taizhou First People's Hospital, Taizhou, Zhejiang, China
| | - Jinglin Xu
- Department of Nephrology, Taizhou First People's Hospital, Taizhou, Zhejiang, China
| | - Xinhong Wu
- Department of Nephrology, Taizhou First People's Hospital, Taizhou, Zhejiang, China
| | - Shouhao Zheng
- Department of Nephrology, Taizhou First People's Hospital, Taizhou, Zhejiang, China
| | - Yingmin Han
- Department of Nephrology, Taizhou First People's Hospital, Taizhou, Zhejiang, China
| | - Guoming Ding
- Department of Nephrology, Taizhou First People's Hospital, Taizhou, Zhejiang, China
| |
Collapse
|
22
|
Wang X, Zhang X, Chen Y, Zhao C, Zhou W, Chen W, Zhang C, Ding K, Li W, Xu H, Lou L, Chu Z, Hu S, Yang J. Cardiac-specific deletion of FDPS induces cardiac remodeling and dysfunction by enhancing the activity of small GTP-binding proteins. J Pathol 2021; 255:438-450. [PMID: 34467534 DOI: 10.1002/path.5789] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 08/04/2021] [Accepted: 08/26/2021] [Indexed: 12/14/2022]
Abstract
The mevalonate pathway is essential for cholesterol biosynthesis. Previous studies have suggested that the key enzyme in this pathway, farnesyl diphosphate synthase (FDPS), regulates the cardiovascular system. We used human samples and mice that were deficient in cardiac FDPS (c-Fdps-/- mice) to investigate the role of FDPS in cardiac homeostasis. Cardiac function was assessed using echocardiography. Left ventricles were examined and tested for histological and molecular markers of cardiac remodeling. Our results showed that FDPS levels were downregulated in samples from patients with cardiomyopathy. Furthermore, c-Fdps-/- mice exhibited cardiac remodeling and dysfunction. This dysfunction was associated with abnormal activation of Ras and Rheb, which may be due to the accumulation of geranyl pyrophosphate. Activation of Ras and Rheb stimulated downstream mTOR and ERK pathways. Moreover, administration of farnesyltransferase inhibitors attenuated cardiac remodeling and dysfunction in c-Fdps-/- mice. These results indicate that FDPS plays an important role in cardiac homeostasis. Deletion of FDPS stimulates the downstream mTOR and ERK signaling pathways, resulting in cardiac remodeling and dysfunction. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Xiying Wang
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China
| | - Xuan Zhang
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China
| | - Yuxiao Chen
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China
| | - Chenze Zhao
- Department of Cardiology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, PR China
| | - Weier Zhou
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China
| | - Wanwan Chen
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China
| | - Chi Zhang
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China
| | - Kejun Ding
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China
| | - Weidong Li
- Department of Cardiothoracic Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China
| | - Hongfei Xu
- Department of Cardiothoracic Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China
| | - Lian Lou
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China
| | - Zhenliang Chu
- Department of Cardiology, The Second Hospital of Jiaxing, Jiaxing, PR China
| | - ShenJiang Hu
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China
| | - Jian Yang
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China
| |
Collapse
|
23
|
Liang T, Gao F, Chen J. Role of PTEN-less in cardiac injury, hypertrophy and regeneration. CELL REGENERATION (LONDON, ENGLAND) 2021; 10:25. [PMID: 34337686 PMCID: PMC8326232 DOI: 10.1186/s13619-021-00087-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 06/18/2021] [Indexed: 12/20/2022]
Abstract
Cardiovascular diseases are the leading cause of death worldwide. Cardiomyocytes are capable of coordinated contractions, which are mainly responsible for pumping blood. When cardiac stress occurs, cardiomyocytes undergo transition from physiological homeostasis to hypertrophic growth, proliferation, or apoptosis. During these processes, many cellular factors and signaling pathways participate. PTEN is a ubiquitous dual-specificity phosphatase and functions by dephosphorylating target proteins or lipids, such as PIP3, a second messenger in the PI3K/AKT signaling pathway. Downregulation of PTEN expression or inhibiting its biologic activity improves heart function, promotes cardiomyocytes proliferation, reduces cardiac fibrosis as well as dilation, and inhibits apoptosis following ischemic stress such as myocardial infarction. Inactivation of PTEN exhibits a potentially beneficial therapeutic effects against cardiac diseases. In this review, we summarize various strategies for PTEN inactivation and highlight the roles of PTEN-less in regulating cardiomyocytes during cardiac development and stress responses.
Collapse
Affiliation(s)
- Tian Liang
- Department of Cardiology, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China
| | - Feng Gao
- Department of Cardiology, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China
| | - Jinghai Chen
- Department of Cardiology, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China. .,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China.
| |
Collapse
|
24
|
Cardiac Mitochondrial PTEN-L determines cell fate between apoptosis and survival during chronic alcohol consumption. Apoptosis 2021; 25:590-604. [PMID: 32591959 DOI: 10.1007/s10495-020-01616-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Chronic alcohol consumption induces myocardial damage and a type of non-ischemic cardiomyopathy termed alcoholic cardiomyopathy, where mitochondrial ultrastructural damages and suppressed fusion activity promote cardiomyocyte apoptosis. The aim of the present study is to determine the role of mitochondrial fission proteins and/or other proteins that localise on cardiac mitochondria for apoptosis upon ethanol consumption. In vivo and in vitro chronic alcohol exposure increased mitochondrial Drp1 levels but knockdown of the same did not confer cardioprotection in H9c2 cells. These cells displayed downregulated expression of MFN2 and OPA1 for Bak-mediated cytochrome c release and apoptosis. Dysregulated PTEN/AKT cell survival signal in both ethanol treated and Drp1 knockdown cells augmented oxidative stress by promoting mitochondrial PTEN-L and MFN1 interaction. Inhibiting this interaction with VO-OHpic, a reversible PTEN inhibitor, prevented Bak insertion into the mitochondria and release of cytochrome c to cytoplasm. Thus, our study provides evidence that Drp1-mediated mitochondrial fission is dispensable for ethanol-induced cardiotoxicity and that stress signals induce mitochondrial PTEN-L accumulation for structural and functional dyshomeostasis. Our in vivo results also demonstrates the therapeutic potential of VO-OHpic for habitual alcoholics developing myocardial dysfunction.
Collapse
|
25
|
Alimoradi N, Firouzabadi N, Fatehi R. Metformin and insulin-resistant related diseases: Emphasis on the role of microRNAs. Biomed Pharmacother 2021; 139:111662. [PMID: 34243629 DOI: 10.1016/j.biopha.2021.111662] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/16/2021] [Accepted: 04/22/2021] [Indexed: 12/14/2022] Open
Abstract
Metformin is one of the most prescribed drugs in type II diabetes (T2DM) which has recently found new applications in the prevention and treatment of various illnesses, from metabolic disorders to cardiovascular and age-related diseases. Metformin improves insulin resistance (IR) by modulating metabolic mechanisms and mitochondrial biogenesis. Alternation of microRNAs (miRs) in the treatment of IR-related illnesses has been observed by metformin therapy. MiRs are small non-coding RNAs that play important roles in RNA silencing, targeting the 3'untranslated region (3'UTR) of most mRNAs and inhibiting the translation of related proteins. As a result, their dysregulation is associated with many diseases. Metformin may alter miRs levels in the treatment of various diseases by AMPK-dependent or AMPK-independent mechanisms. Here, we summarized the therapeutic role of metformin by modifying the aberrant expression of miRs as potential biomarkers or therapeutic targets in diseases in which IR plays a key role.
Collapse
Affiliation(s)
- Nahid Alimoradi
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negar Firouzabadi
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Reihaneh Fatehi
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
26
|
Shen K, Liu X, Chen D, Chang J, Zhang Y, Kou X. Voluntary wheel-running exercise attenuates brain aging of rats through activating miR-130a-mediated autophagy. Brain Res Bull 2021; 172:203-211. [PMID: 33964346 DOI: 10.1016/j.brainresbull.2021.04.027] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 03/09/2021] [Accepted: 04/20/2021] [Indexed: 01/03/2023]
Abstract
Autophagy is a highly regulated intracellular process for the degradation of protein aggregates and damaged organelles. Recently, autophagy has been implicated in Alzheimer's disease (AD) and aging. Autophagy process is regulated by the recruitment and assembly of several autophagy-related genes (Atgs) such as, Atg7 and LC3, as the highly conserved and important markers involved in the regulation of autophagy. We recently reported the reduced LC3-II/LC3-I ratio, down-regulated ATG7, and increased p62 protein levels in hippocampal tissues of aging rats. MicroRNA-130a (miR-130a) plays a crucial role in physiological and pathological processes, but whether miR-130a affects the autophagy of brain is unknown. We aim to explore the regulatory role of miR-130a on the autophagy and cell senescence of SH-SY5Y, as well as LC3-II/LC3-I ratio, and the expression of p62, ATG7, Ac-p53 and p21 during exercise intervention of aging rats. In this study, miR-130a expression was markedly down-regulated in the hippocampal of aged rats companying with up-regulated expression of Ac-p53 and p21 when compared with young rats. In contrast, voluntary wheel running could up-regulate miR-130a expression; decrease the expression of Ac-p53 and p21 in aging rats. Interestingly, exercise reversed the impaired autophagy resulted from aging possibly by activating AMPK signaling. Moreover, overexpression of miR-130a in d-galactose (D-gal)-induced SH-SY5Y cell senescence model attenuated d-gal-induced impaired autophagy and cell senescence, demonstrated by decreased levels of LC3, Ac-p53, p21 and increased p62, suggesting that voluntary wheel running can alleviate brain aging in natural aging rats by up-regulating miR-130a-mediated autophagy.
Collapse
Affiliation(s)
- Keyin Shen
- Wuhan Sports University, Wuhan, 430079, China
| | - Xingran Liu
- Hubei Key Laboratory of Sport Training and Monitoring, College of Health Science, Wuhan Sports University, Wuhan, 430079, China
| | - Dandan Chen
- Hubei Key Laboratory of Sport Training and Monitoring, College of Health Science, Wuhan Sports University, Wuhan, 430079, China
| | - Jingru Chang
- Hubei Key Laboratory of Sport Training and Monitoring, College of Health Science, Wuhan Sports University, Wuhan, 430079, China
| | - Ying Zhang
- Hubei Key Laboratory of Sport Training and Monitoring, College of Health Science, Wuhan Sports University, Wuhan, 430079, China
| | - Xianjuan Kou
- Hubei Key Laboratory of Sport Training and Monitoring, College of Health Science, Wuhan Sports University, Wuhan, 430079, China.
| |
Collapse
|
27
|
Rian/miR-210-3p/Nfkb1 Feedback Loop Promotes Hypoxia-Induced Cell Apoptosis in Myocardial Infarction Through Deactivating the PI3K/Akt Signaling Pathway. J Cardiovasc Pharmacol 2021; 76:207-215. [PMID: 32187165 DOI: 10.1097/fjc.0000000000000824] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Myocardial infarction (MI) is a severe disease that could lead to reversible or irreversible ischemic heart damage. A previous study has revealed that microRNA mmu-miR-210-3p expression is downregulated in fat-1 transgenic mice post-MI. Nevertheless, the specific mechanism of miR-210-3p in MI remains obscure. In this study, we observed that miR-210-3p expression was downregulated in the mice's left ventricle post-MI, and miR-210-3p expression was suppressed while cell apoptosis was promoted in H9c2 cells under hypoxia condition. Besides, miR-210-3p overexpression could enhance cell proliferation and inhibit cell apoptosis in hypoxia-treated H9c2 cells. Then, molecular mechanism assays revealed that miR-210-3p overexpression could activate the PI3K/Akt pathway, and nuclear factor of kappa light polypeptide gene enhancer in B cells 1 (Nfkb1) was the target of miR-210-3p. In addition, RNA imprinted and accumulated in nucleus (Rian), a long noncoding RNA, could sponge miR-210-3p to upregulate Nfkb1 expression. Besides, Nfkb1 was verified to facilitate the transcription of Rian by binding with a Rian promoter. Furthermore, rescue assays revealed that both Nfkb1 and PI3K/Akt pathway are engaged in the Rian-mediated cell proliferation and apoptosis in hypoxia-treated H9c2 cells. In conclusion, a Rian/miR-210-3p/Nfkb1 feedback loop enhances hypoxia-induced cell apoptosis in MI through deactivating the PI3K/Akt pathway.
Collapse
|
28
|
Chang C, Yan J, Yao Z, Zhang C, Li X, Mao H. Effects of Mesenchymal Stem Cell-Derived Paracrine Signals and Their Delivery Strategies. Adv Healthc Mater 2021; 10:e2001689. [PMID: 33433956 PMCID: PMC7995150 DOI: 10.1002/adhm.202001689] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 12/13/2020] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) have been widely studied as a versatile cell source for tissue regeneration and remodeling due to their potent bioactivity, which includes modulation of inflammation response, macrophage polarization toward proregenerative lineage, promotion of angiogenesis, and reduction in fibrosis. This review focuses on profiling the effects of paracrine signals of MSCs, commonly referred to as the secretome, and highlighting the various engineering approaches to tune the MSC secretome. Recent advances in biomaterials‐based therapeutic strategies for delivery of MSCs and MSC‐derived secretome in the form of extracellular vesicles are discussed, along with their advantages and challenges.
Collapse
Affiliation(s)
- Calvin Chang
- Department of Biomedical Engineering, School of Medicine Johns Hopkins University Baltimore MD 21205 USA
- Translational Tissue Engineering Center Johns Hopkins School of Medicine Baltimore MD 21287 USA
- Institute for NanoBioTechnology Johns Hopkins University Baltimore MD 21218 USA
| | - Jerry Yan
- Department of Biomedical Engineering, School of Medicine Johns Hopkins University Baltimore MD 21205 USA
- Translational Tissue Engineering Center Johns Hopkins School of Medicine Baltimore MD 21287 USA
- Institute for NanoBioTechnology Johns Hopkins University Baltimore MD 21218 USA
| | - Zhicheng Yao
- Translational Tissue Engineering Center Johns Hopkins School of Medicine Baltimore MD 21287 USA
- Institute for NanoBioTechnology Johns Hopkins University Baltimore MD 21218 USA
- Department of Materials Science and Engineering, Whiting School of Engineering Johns Hopkins University Baltimore MD 21218 USA
| | - Chi Zhang
- Translational Tissue Engineering Center Johns Hopkins School of Medicine Baltimore MD 21287 USA
- Institute for NanoBioTechnology Johns Hopkins University Baltimore MD 21218 USA
- Department of Materials Science and Engineering, Whiting School of Engineering Johns Hopkins University Baltimore MD 21218 USA
| | - Xiaowei Li
- Mary and Dick Holland Regenerative Medicine Program and Department of Neurological Sciences University of Nebraska Medical Center Omaha NE 68198 USA
| | - Hai‐Quan Mao
- Department of Biomedical Engineering, School of Medicine Johns Hopkins University Baltimore MD 21205 USA
- Translational Tissue Engineering Center Johns Hopkins School of Medicine Baltimore MD 21287 USA
- Institute for NanoBioTechnology Johns Hopkins University Baltimore MD 21218 USA
- Department of Materials Science and Engineering, Whiting School of Engineering Johns Hopkins University Baltimore MD 21218 USA
| |
Collapse
|
29
|
PTEN mediates serum deprivation-induced cytotoxicity in H9c2 cells via the PI3K/AKT signaling pathway. Toxicol In Vitro 2021; 73:105131. [PMID: 33652126 DOI: 10.1016/j.tiv.2021.105131] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 02/09/2021] [Accepted: 02/25/2021] [Indexed: 12/22/2022]
Abstract
The pathogenesis of acute myocardial infarction (AMI) is associated with cardiomyocyte necrosis and apoptosis. Numerous studies have determined the regulatory effects of Phosphatase and tensin homolog (PTEN) cell proliferation and apoptosis in other cell types. However, the potential role of PTEN in cardiomyocyte is unclear. In this study, we used H9c2 cells cultured under serum deprivation to simulate the apoptosis process of myocardial infarction. Small interference RNA (siRNA) of PTEN was used to knock down the expression of PTEN. Cell viability was determined by CCK-8. Cell proliferation was examined by Edu staining, and the protein expression was analyzed by Western blot. We also evaluated the generation of ROS, the degree of DNA damage, and cell apoptosis using immunofluorescence assay. As a result, we observed that serum deprivation in H9c2 cells increased PTEN expression. Functionally, the PTEN knockdown experiment using siRNA inhibited serum deprivation-induced cell apoptosis, ROS production, and DNA damage, whereas increased cell proliferation. All these effects could be reversed by phosphatidylinositol 3-kinase (PI3K) inhibitor, which indicated the PI3K/protein kinase B (AKT) might be the critical component of the PTEN effects during serum deficiency. In conclusion, our study indicated the role of the PTEN/PI3K/AKT pathway in serum deprivation-induced cytotoxicity in H9c2 cells.
Collapse
|
30
|
Li Y, Zhang H, Li Z, Yan X, Li Y, Liu S. microRNA-130a-5p suppresses myocardial ischemia reperfusion injury by downregulating the HMGB2/NF-κB axis. BMC Cardiovasc Disord 2021; 21:121. [PMID: 33658008 PMCID: PMC7931544 DOI: 10.1186/s12872-020-01742-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 10/13/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Myocardial ischemia reperfusion injury (MIRI) is defined as tissue injury in the pathological process of progressive aggravation in ischemic myocardium after the occurrence of acute coronary artery occlusion. Research has documented the involvement of microRNAs (miRs) in MIRI. However, there is obscure information about the role of miR-130a-5p in MIRI. Herein, this study aims to investigate the effect of miR-130a-5p on MIRI. METHODS MIRI mouse models were established. Then, the cardiac function and hemodynamics were detected using ultrasonography and multiconductive physiological recorder. Functional assays in miR-130a-5p were adopted to test the degrees of oxidative stress, mitochondrial functions, inflammation and apoptosis. Hematoxylin and eosin (HE) staining was performed to validate the myocardial injury in mice. Reverse transcription quantitative polymerase chain reaction (RT-qPCR) was employed to assess the expression patterns of miR-130a-5p, high mobility group box (HMGB)2 and NF-κB. Then, dual-luciferase reporter gene assay was performed to elucidate the targeting relation between miR-130a-5p and HMGB2. RESULTS Disrupted structural arrangement in MIRI mouse models was evident from HE staining. RT-qPCR revealed that overexpressed miR-130a-5p alleviated MIRI, MIRI-induced oxidative stress and mitochondrial disorder in the mice. Next, the targeting relation between miR-130a-5p and HMGB2 was ascertained. Overexpressed HMGB2 annulled the protective effects of miR-130a-5p in MIRI mice. Additionally, miR-130a-5p targets HMGB2 to downregulate the nuclear factor kappa-B (NF-κB) axis, mitigating the inflammatory injury induced by MIRI. CONCLUSION Our study demonstrated that miR-130a-5p suppresses MIRI by down-regulating the HMGB2/NF-κB axis. This investigation may provide novel insights for development of MIRI treatments.
Collapse
Affiliation(s)
- Yong Li
- Department of Cardiology, Harrision International Peace Hospital, No. 180 Renmin East Road, Hengshui, 053000, Hebei, People's Republic of China.
| | - Hongbo Zhang
- Department of Cardiology, Harrision International Peace Hospital, No. 180 Renmin East Road, Hengshui, 053000, Hebei, People's Republic of China
| | - Zhanhu Li
- Department of Cardiology, Harrision International Peace Hospital, No. 180 Renmin East Road, Hengshui, 053000, Hebei, People's Republic of China
| | - Xiaoju Yan
- Department of Cardiology, Harrision International Peace Hospital, No. 180 Renmin East Road, Hengshui, 053000, Hebei, People's Republic of China
| | - Yuan Li
- Department of Cardiology, Harrision International Peace Hospital, No. 180 Renmin East Road, Hengshui, 053000, Hebei, People's Republic of China
| | - Shuai Liu
- Department of Cardiology, Harrision International Peace Hospital, No. 180 Renmin East Road, Hengshui, 053000, Hebei, People's Republic of China
| |
Collapse
|
31
|
Feng Q, Ren Y, Hou A, Guo J, Mao Z, Liu S, Wang B, Bai Z, Hou X. MicroRNA-130a Increases and Predicts Cardiotoxicity during Adjuvant Chemotherapy in Human Epidermal Growth Factor Receptor-2-Positive Breast Cancer. J Breast Cancer 2021; 24:153-163. [PMID: 33818020 PMCID: PMC8090801 DOI: 10.4048/jbc.2021.24.e15] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 12/25/2020] [Accepted: 01/18/2021] [Indexed: 12/21/2022] Open
Abstract
PURPOSE This study aimed to investigate the changes in microRNA-130a (miR-130a) and its correlation with cardiotoxicity during epirubicin/cyclophosphamide followed by docetaxel plus trastuzumab (EC-D+T) adjuvant chemotherapy in human epidermal growth factor receptor-2-positive (HER2⁺) breast cancer patients. METHODS A total of 72 HER2⁺ breast cancer patients who underwent resection and were scheduled to receive EC-D+T adjuvant therapy were consecutively enrolled. The expression of miR-130a and cardiotoxicity (defined as any of the following situations: 1) absolute decline of left ventricular ejection fraction (LVEF) ≥ 10% and LVEF < 53%; 2) heart failure; 3) acute coronary artery syndromes; and 4) fatal arrhythmia) were assessed every 3 months throughout the 15-month EC-D+T treatment. RESULTS The accumulating cardiotoxicity rate was 12 (16.7%), of which the incidence of heart failure, acute coronary syndrome, life-threatening arrhythmias, ΔLVEF ≥ 10%, and LVEF < 53% was 0 (0.0%), 1 (1.4%), 0 (0.0%), and 12 (16.7%), respectively. Baseline miR-130a expression was negatively correlated with LVEF (%) and positively correlated with cardiac troponin I. The expression of miR-130a gradually increased in both cardiotoxicity and non-cardiotoxicity patients during EC-D+T treatment, while the increment of miR-130a was more obvious in cardiotoxicity patients compared with non-cardiotoxicity patients. Further logistic regression and receiver operating characteristic curve analysis indicated that miR-130a was an independent predictive factor for increased cardiotoxicity risk. CONCLUSION MiR-130a increases constantly and predicts high cardiotoxicity risk during EC-D+T adjuvant chemotherapy in HER2⁺ breast cancer patients.
Collapse
Affiliation(s)
- Qiang Feng
- Department of Cardiology, HanDan Central Hospital, Handan, China
| | - Yanbin Ren
- Department of Ultrasound Medicine, HanDan Central Hospital, Handan, China.
| | - Aijun Hou
- Department of Cardiology, HanDan Central Hospital, Handan, China
| | - Jing Guo
- Department of Cardiology, HanDan Central Hospital, Handan, China
| | - Zhezhe Mao
- Second Ward of Respiratory Medicine, HanDan Central Hospital, Handan, China
| | - Shaojun Liu
- Office of Quality Management, HanDan Central Hospital, Handan, China
| | - Boya Wang
- Department of Cardiology, HanDan Central Hospital, Handan, China
| | - Zhichao Bai
- Department of Cardiology, HanDan Central Hospital, Handan, China
| | - Xiaoying Hou
- Department of Ultrasound Medicine, HanDan Central Hospital, Handan, China
| |
Collapse
|
32
|
Blawas AM, Ware KE, Schmaltz E, Zheng L, Spruance J, Allen AS, West N, Devos N, Corcoran DL, Nowacek DP, Eward WC, Fahlman A, Somarelli JA. An integrated comparative physiology and molecular approach pinpoints mediators of breath-hold capacity in dolphins. Evol Med Public Health 2021; 9:420-430. [PMID: 35169481 PMCID: PMC8833867 DOI: 10.1093/emph/eoab036] [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: 07/16/2021] [Accepted: 10/17/2021] [Indexed: 11/14/2022] Open
Abstract
Abstract
Background and objectives
Ischemic events, such as ischemic heart disease and stroke, are the number one cause of death globally. Ischemia prevents blood, carrying essential nutrients and oxygen, from reaching tissues, leading to cell and tissue death, and eventual organ failure. While humans are relatively intolerant to ischemic events, other species, such as marine mammals, have evolved a unique tolerance to chronic ischemia/reperfusion during apneic diving. To identify possible molecular features of an increased tolerance for apnea, we examined changes in gene expression in breath-holding dolphins.
Methodology
Here, we capitalized on the adaptations possesed by bottlenose dolphins (Tursiops truncatus) for diving as a comparative model of ischemic stress and hypoxia tolerance to identify molecular features associated with breath holding. Given that signals in the blood may influence physiological changes during diving, we used RNA-Seq and enzyme assays to examine time-dependent changes in gene expression in the blood of breath-holding dolphins.
Results
We observed time-dependent upregulation of the arachidonate 5-lipoxygenase (ALOX5) gene and increased lipoxygenase activity during breath holding. ALOX5 has been shown to be activated during hypoxia in rodent models, and its metabolites, leukotrienes, induce vasoconstriction.
Conclusions and implications
The upregulation of ALOX5 mRNA occurred within the calculated aerobic dive limit of the species, suggesting that ALOX5 may play a role in the dolphin’s physiological response to diving, particularly in a pro-inflammatory response to ischemia and in promoting vasoconstriction. These observations pinpoint a potential molecular mechanism by which dolphins, and perhaps other marine mammals, respond to the prolonged breath holds associated with diving.
Collapse
Affiliation(s)
- Ashley M Blawas
- Nicholas School of the Environment, Duke University Marine Laboratory, Beaufort, NC, USA
| | - Kathryn E Ware
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Emma Schmaltz
- Nicholas School of the Environment, Duke University Marine Laboratory, Beaufort, NC, USA
| | - Larry Zheng
- Nicholas School of the Environment, Duke University Marine Laboratory, Beaufort, NC, USA
| | - Jacob Spruance
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Austin S Allen
- Nicholas School of the Environment, Duke University Marine Laboratory, Beaufort, NC, USA
| | | | - Nicolas Devos
- Duke Center for Genomic and Computational Biology, Duke University, Durham, NC, USA
| | - David L Corcoran
- Duke Center for Genomic and Computational Biology, Duke University, Durham, NC, USA
| | - Douglas P Nowacek
- Nicholas School of the Environment, Duke University Marine Laboratory, Beaufort, NC, USA
- Pratt School of Engineering, Duke University, Durham, NC, USA
| | - William C Eward
- Department of Orthopaedic Surgery, Duke University Medical Center, Durham, NC, USA
- Duke University Medical Center, Duke Cancer Institute, Durham, NC, USA
| | - Andreas Fahlman
- Global Diving Research, Inc., Ottawa, ON, Canada
- Research Department, Fundación Oceanogrāfic de la Comunitat Valenciana, Valencia, Spain
| | - Jason A Somarelli
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
- Duke University Medical Center, Duke Cancer Institute, Durham, NC, USA
| |
Collapse
|
33
|
Cheng N, Wang MY, Wu YB, Cui HM, Wei SX, Liu B, Wang R. Circular RNA POSTN Promotes Myocardial Infarction-Induced Myocardial Injury and Cardiac Remodeling by Regulating miR-96-5p/BNIP3 Axis. Front Cell Dev Biol 2021; 8:618574. [PMID: 33681183 PMCID: PMC7930329 DOI: 10.3389/fcell.2020.618574] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 12/30/2020] [Indexed: 12/23/2022] Open
Abstract
Myocardial infarction (MI) is the most prevalent cardiac disease with high mortality, leading to severe heart injury. Circular RNAs (circRNAs) are a new type of regulatory RNAs and participate in multiple pathological cardiac progressions. However, the role of circRNAs Postn (circPostn) in MI modulation remains unclear. Here, we aimed to explore the effect of circPostn on MI-induced myocardial injury and cardiac remodeling. We identified that the expression of circPostn was elevated in the plasma of MI patients, MI mouse model, and hypoxia and reoxygenation (H/R)-treated human cardiomyocytes. The depletion of circPostn significantly attenuated MI-related myocardium injury and reduced the infarct size in MI mouse model. The circPostn knockdown obviously enhanced left ventricular ejection fraction (LVEF) and left ventricular fraction shortening (LVFS) and inhibited left ventricular anterior wall thickness at diastole (LVAWd) and left ventricular posterior wall thickness at diastole (LVPWd). The depletion of circPostn was able to decrease MI-induced expression of collagen 1α1 and collagen 3α1 in the ventricular tissues of mice. The protein expression of collagen and α-smooth muscle actin (SMA) was up-regulated in MI mice and was inhibited by circPostn knockdown. Meanwhile, the expression of atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP) was repressed by circPostn depletion in the ventricular tissues of MI mice. Besides, the circPostn depletion attenuated cardiomyocyte apoptosis in mice. Mechanically, circPostn served as a miR-96-5p sponge and miR-96-5p-targeted BNIP3 in human cardiomyocytes, in which circPostn up-regulated BNIP3 expression by targeting miR-96-5p. circPostn promoted H/R-induced cardiomyocyte injury by modulating miR-96-5p/BNIP3 axis. Thus, we conclude that circPostn contributes to MI-induced myocardial injury and cardiac remodeling by regulating miR-96-5p/BNIP3 axis. Our finding provides new insight into the mechanism by which circPostn regulates MI-related cardiac dysfunction. circPostn, miR-96-5p, and BNIP3 are potential targets for the treatment of MI-caused heart injury.
Collapse
Affiliation(s)
- Nan Cheng
- Department of Cardiovascular Surgery, PLA General Hospital, Beijing, China
| | - Ming-Yan Wang
- Department of Cardiovascular Surgery, PLA General Hospital, Beijing, China
| | - Yuan-Bin Wu
- Department of Cardiovascular Surgery, PLA General Hospital, Beijing, China
| | - Hui-Min Cui
- Department of Cardiovascular Surgery, PLA General Hospital, Beijing, China
| | - Shi-Xiong Wei
- Department of Cardiovascular Surgery, PLA General Hospital, Beijing, China
| | - Bing Liu
- Department of Cardiovascular Surgery, PLA General Hospital, Beijing, China
| | - Rong Wang
- Department of Cardiovascular Surgery, PLA General Hospital, Beijing, China
| |
Collapse
|
34
|
Wei X, Zheng Y, Zhang W, Tan J, Zheng H. Ultrasound‑targeted microbubble destruction‑mediated Galectin‑7‑siRNA promotes the homing of bone marrow mesenchymal stem cells to alleviate acute myocardial infarction in rats. Int J Mol Med 2020; 47:677-687. [PMID: 33416139 PMCID: PMC7797467 DOI: 10.3892/ijmm.2020.4830] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 10/19/2020] [Indexed: 12/13/2022] Open
Abstract
Bone marrow mesenchymal stem cells (BMSCs) are accepted as a form of cellular therapy to improve cardiac function following acute myocardial infarction (AMI). The present study was performed to investigate the synergistic effect of ultrasound-targeted microbubble destruction (UTMD)-mediated Galectin-7-small interfering (si)RNA with the homing of BMSCs for AMI. The rat model of AMI was established, followed by identification of BMSCs. Rats with AMI received BMSC transplantation, BMSC transplantation + UTMD + siRNA negative control, or BMSC transplantation + UTMD + Galectin-7-siRNA. The cardiac function, hemodynamics indexes, degree of myocardial fiber injury and expression of apoptosis-related proteins in myocardial tissues of rats were detected. The homing of BMSCs was observed, and the indexes of myocardial microenvironment and the TGF-β/Smads pathway-related proteins in myocardial tissues were determined. AMI rats treated with UTMD-mediated Galectin-7-siRNA exhibited improved cardiac function and hemodynamics-related indices, decreased myocardial fiber injury and apoptotic cells, as well as enhanced homing ability of BMSCs, improved myocardial microenvironment, and suppressed TGF-β1/Smads pathway activation. In conclusion, the present study demonstrated that UTMD-mediated Galectin-7-siRNA treatment could enhance the homing ability of BMSCs, thus alleviating AMI in rats.
Collapse
Affiliation(s)
- Xin Wei
- Department of Ultrasound, People's Hospital of Deyang City, Deyang, Sichuan 618000, P.R. China
| | - Yan Zheng
- Department of Ultrasound, People's Hospital of Deyang City, Deyang, Sichuan 618000, P.R. China
| | - Weilin Zhang
- Department of Ultrasound, People's Hospital of Deyang City, Deyang, Sichuan 618000, P.R. China
| | - Jing Tan
- Department of Cardiology, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Hong Zheng
- Department of Ultrasound, People's Hospital of Deyang City, Deyang, Sichuan 618000, P.R. China
| |
Collapse
|
35
|
SET8 suppression mediates high glucose-induced vascular endothelial inflammation via the upregulation of PTEN. Exp Mol Med 2020; 52:1715-1729. [PMID: 33028948 PMCID: PMC8080680 DOI: 10.1038/s12276-020-00509-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 07/27/2020] [Accepted: 08/10/2020] [Indexed: 11/19/2022] Open
Abstract
Hyperglycemia-mediated endothelial inflammation participates in the pathogenesis of cardiovascular complications in subjects with diabetes. Previous studies reported that phosphatase and tensin homolog deleted on chromosome ten (PTEN) and SET8 participate in high glucose-mediated endothelial inflammation. In this study, we hypothesize that SET8 regulates PTEN expression, thus contributing to high glucose-mediated vascular endothelial inflammation. Our data indicated that plasma soluble intercellular adhesion molecule-1 (sICAM-1) and endothelial selectin (e-selectin) were increased in patients with diabetes and diabetic rats. PTEN expression was augmented in the peripheral blood mononuclear cells of patients with diabetes and in the aortic tissues of diabetic rats. Our in vitro study indicated that high glucose increased monocyte/endothelial adhesion, endothelial adhesion molecule expression and p65 phosphorylation in human umbilical vein endothelial cells (HUVECs). Moreover, high glucose led to endothelial inflammation via upregulation of PTEN. Furthermore, high glucose inhibited SET8 expression and histone H4 lysine 20 methylation (H4K20me1), a downstream target of SET8. SET8 overexpression reversed the effects of high-glucose treatment. shSET8-mediated endothelial inflammation was counteracted by siPTEN. Furthermore, SET8 was found to interact with FOXO1. siFOXO1 attenuated high glucose-mediated endothelial inflammation. FOXO1 overexpression-mediated endothelial inflammation was counteracted by siPTEN. H4K20me1 and FOXO1 were enriched in the PTEN promoter region. shSET8 increased PTEN promoter activity and augmented the positive effect of FOXO1 overexpression on PTEN promoter activity. Our in vivo study indicated that SET8 was downregulated and FOXO1 was upregulated in the peripheral blood mononuclear cells of patients with diabetes and the aortic tissues of diabetic rats. In conclusion, SET8 interacted with FOXO1 to modulate PTEN expression in vascular endothelial cells, thus contributing to hyperglycemia-mediated endothelial inflammation. High glucose levels in patients with diabetes trigger vascular inflammation by affecting the expression of key proteins in blood vessel linings. Elevated glucose causes inflammation of the endothelium, a thin layer of cells that lines blood and lymph vessels, leading to cardiovascular complications. The phosphatase and tensin homolog protein (PTEN) contributes to endothelial inflammation, but the precise mechanisms are unclear. Xuefang Shen at Fudan University in Shanghai, China, and co-workers demonstrated that elevated glucose increases PTEN expression, with increased levels of the protein found in peripheral blood cells of diabetic patients and aortic tissues of diabetic rats. In further experiments on rats, the researchers found that glucose also suppressed another protein called SET8, which contributed to increased PTEN levels. This suggests that SET8 is involved in PTEN modulation, and that both proteins influence vascular inflammation.
Collapse
|
36
|
Fan M, Yang K, Wang X, Wang Y, Tu F, Ha T, Liu L, Williams DL, Li C. Endothelial cell HSPA12B and yes-associated protein cooperatively regulate angiogenesis following myocardial infarction. JCI Insight 2020; 5:139640. [PMID: 32790647 PMCID: PMC7526558 DOI: 10.1172/jci.insight.139640] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 08/05/2020] [Indexed: 12/19/2022] Open
Abstract
Angiogenesis is essential for cardiac functional recovery after myocardial infarction (MI). HSPA12B is predominately expressed in endothelial cells and required for angiogenesis. Yes-associated protein (YAP) plays an important role in tumor angiogenesis. This study investigated the cooperative role of HSPA12B and YAP in angiogenesis after MI. Silencing of either HSPA12B or YAP impaired hypoxia-promoted endothelial cell proliferation and angiogenesis. Deficiency of HSPA12B suppressed YAP expression and nuclear translocation after hypoxia. Knockdown of YAP attenuated hypoxia-stimulated HSPA12B nuclear translocation and abrogated HSPA12B-promoted endothelial cell angiogenesis. Mechanistically, hypoxia induced an interaction between endothelial HSPA12B and YAP. ChIP assay showed that HSPA12B is a target gene of YAP/transcriptional enhanced associated domain 4 (TEAD4) and a coactivator in YAP-associated angiogenesis. In vivo studies using the MI model showed that endothelial cell-specific deficiency of HSPA12B (eHspa12b-/-) or YAP (eYap-/-) impaired angiogenesis and exacerbated cardiac dysfunction compared with WT mice. MI increased YAP expression and nuclear translocation in WT hearts but not eHspa12b-/- hearts. HSPA12B expression and nuclear translocation were upregulated in WT MI hearts but not eYap-/- MI myocardium. Our data demonstrate that endothelial HSPA12B is a target and coactivator for YAP/TEAD4 and cooperates with YAP to regulate endothelial angiogenesis after MI.
Collapse
Affiliation(s)
- Min Fan
- Department of Surgery and
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University (ETSU), Johnson City, Tennessee, USA
| | - Kun Yang
- Department of Surgery and
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University (ETSU), Johnson City, Tennessee, USA
| | - Xiaohui Wang
- Department of Surgery and
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University (ETSU), Johnson City, Tennessee, USA
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, Massachusetts, USA
| | | | - Fei Tu
- Department of Surgery and
| | - Tuanzhu Ha
- Department of Surgery and
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University (ETSU), Johnson City, Tennessee, USA
| | - Li Liu
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - David L. Williams
- Department of Surgery and
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University (ETSU), Johnson City, Tennessee, USA
| | - Chuanfu Li
- Department of Surgery and
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University (ETSU), Johnson City, Tennessee, USA
| |
Collapse
|
37
|
Zhu Y, Zou C, Jia Y, Zhang H, Ma X, Zhang J. Knockdown of circular RNA circMAT2B reduces oxygen-glucose deprivation-induced inflammatory injury in H9c2 cells through up-regulating miR-133. Cell Cycle 2020; 19:2622-2630. [PMID: 32897801 DOI: 10.1080/15384101.2020.1814025] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Myocardial infarction (MI) is the main cause of morbidity and mortality. Reperfusion ways can cause damage to cardiomyocytes. CircMAT2B, a novel circRNA, takes positive roles in regulating glucose metabolism under hypoxia. Therefore, we aimed to explore the effects of circMAT2B on MI. Oxygen-glucose deprivation (OGD)-induced H9c2 cell model was employed to stimulate MI. Ex-circMAT2B, si-circMAT2B, miR-133 inhibitor and relative control were transfected into H9c2 cells. qRT-PCR was employed to examine levels of circMAT2B and miR-133. Cell activity, apoptosis, ROS generation and release of inflammatory factors were assessed by CCK-8, flow cytometry, ROS species assay kit and ELISA, respectively. Moreover, the expression of apoptosis-related and pathway-related factors was detected through western blot analysis. The results showed that circMAT2B expression was notably up-regulated by OGD treatment. Moreover, circMAT2B knockdown could effectively decrease OGD-induced the increasing of apoptosis, ROS generation and the expression of IL-1β, IL-6 and TNF-α. Besides, miR-133 was positively regulated by si-circMAT2B. CircMAT2B knockdown attenuated OGD-induced H9c2 cell damage and alleviated OGD-induced the inhibition of PI3K/AKT and Raf/MEK/ERK pathways through up-regulating miR-133. In brief, circMAT2B knockdown works as an inflammatory inhibitor in OGD-induced H9c2 cells inflammatory injury through up-regulating miR-133.
Collapse
Affiliation(s)
- Yanhui Zhu
- Department of Cardiac Surgery, Shandong Provincial Hospital Affiliated to Shandong University , Jinan, Shandong, China
| | - Chengwei Zou
- Department of Cardiac Surgery, Shandong Provincial Hospital Affiliated to Shandong University , Jinan, Shandong, China
| | - Yanting Jia
- Department of Cardiac Surgery, Shandong Provincial Hospital Affiliated to Shandong University , Jinan, Shandong, China
| | - Haizhou Zhang
- Department of Cardiac Surgery, Shandong Provincial Hospital Affiliated to Shandong University , Jinan, Shandong, China
| | - Xiaochun Ma
- Department of Cardiac Surgery, Shandong Provincial Hospital Affiliated to Shandong University , Jinan, Shandong, China
| | - Jun Zhang
- Department of Cardiac Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University , Jinan, Shandong, China
| |
Collapse
|
38
|
Upregulating MicroRNA-203 Alleviates Myocardial Remodeling and Cell Apoptosis Through Downregulating Protein Tyrosine Phosphatase 1B in Rats With Myocardial Infarction. J Cardiovasc Pharmacol 2020; 74:474-481. [PMID: 31725080 DOI: 10.1097/fjc.0000000000000733] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Myocardial infarction (MI) is one of cardiovascular diseases with high incidence and mortality. MicroRNAs, as posttranscriptional regulators of genes, are involved in many diseases, including cardiovascular diseases. The aim of the present study was to determine whether miR-203 was functional in MI therapy and how it worked. Left anterior descending artery ligation and hypoxia/reoxygenation (H/R) treatment were, respectively, performed to obtain MI rats and hypoxia-injured H9c2 cells. Western blot and quantitative real-time polymerase chain reaction were used to determine protein levels and messenger RNA of relevant genes, respectively. Lentivirus-mediated overexpression of miR-203 was performed to study the miR-203 functions on left ventricular remodeling, infarct size, and cardiomyocyte apoptosis. Compared with the sham group, miR-203 levels were significantly decreased in MI and H/R groups. However, overexpressing miR-203 greatly improved the cardiac function, reduced infarct size in rats after MI and weakened infarction-induced apoptosis by increasing Bcl-2 and reducing decreasing Bax, cleaved caspase-3, and cleaved caspase-9. In addition, Protein tyrosine phosphatase 1B (PTP1B) was proved as a target of miR-203 in cardiomyocytes, and it was negatively regulated by miR-203. Further experiments indicated that PTP1B overexpression could remarkably inhibit miR-203-mediated antiapoptosis of cardiomyocytes and alleviate protective effects of miR-203 on mitochondria after H/R treatment. Altogether, miR-203 prevented infarction-induced apoptosis by regulating PTP1B, including reducing proapoptosis proteins, inactivating caspase pathway, and protecting mitochondria. In conclusion, miR-203 had abilities to alleviate MI-caused injury on myocardium tissues and reduce mitochondria-mediated apoptosis, which might be a potential target used for MI therapy.
Collapse
|
39
|
Reduced expression of microRNA-130a promotes endothelial cell senescence and age-dependent impairment of neovascularization. Aging (Albany NY) 2020; 12:10180-10193. [PMID: 32457253 PMCID: PMC7346016 DOI: 10.18632/aging.103340] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Accepted: 05/18/2020] [Indexed: 12/30/2022]
Abstract
Aging is associated with impaired neovascularization in response to ischemia. MicroRNAs are small noncoding RNAs emerging as key regulators of physiological and pathological processes. Here we investigated the potential role of microRNAs in endothelial cell senescence and age-dependent impairment of neovascularization. Next generation sequencing and qRT-PCR analyses identified miR-130a as a pro-angiogenic microRNA which expression is significantly reduced in old mouse aortic endothelial cells (ECs). Transfection of young ECs with a miR-130a inhibitor leads to accelerated senescence and reduced angiogenic functions. Conversely, forced expression of miR-130a in old ECs reduces senescence and improves angiogenesis. In a mouse model of hindlimb ischemia, intramuscular injection of miR-130a mimic in older mice restores blood flow recovery and vascular densities in ischemic muscles, improves mobility and reduces tissue damage. miR-130a directly targets antiangiogenic homeobox genes MEOX2 and HOXA5. MEOX2 and HOXA5 are significantly increased in the ischemic muscles of aging mice, but forced expression of miR-130a reduces the expression of these factors. miR-130a treatment after ischemia is also associated with increased number and improved functional activities of pro-angiogenic cells (PACs). Forced expression of miR-130a could constitute a novel strategy to improve blood flow recovery and reduce ischemia in older patients with ischemic vascular diseases.
Collapse
|
40
|
Abstract
MicroRNAs (miRNA) are non-coding RNAs that regulate gene expression in up to 90% of the human genome through interactions with messenger RNA (mRNA). The expression of miRNAs varies and changes in diseased and healthy states, including all stages of myocardial ischemia-reperfusion and subsequent ischemia-reperfusion injury (IRI). These changes in expression make miRNAs an attractive potential therapeutic target. Herein, we review the differences in miRNA expression prior to ischemia (including remote ischemic conditioning and ischemic pre-conditioning), the changes during ischemia-reperfusion, and the changes in miRNA expression after IRI, with an emphasis on inflammatory and fibrotic pathways. Additionally, we review the effects of manipulating the levels of certain miRNAs on changes in infarct size, inflammation, remodeling, angiogenesis, and cardiac function after either ischemia-reperfusion or permanent coronary ligation. Levels of target miRNA can be increased using molecular mimics ("agomirs"), or can be decreased by using "antagomirs" which are antisense molecules that act to bind and thus inactivate the target miRNA sequence. Other non-coding RNAs, including long non-coding RNAs and circular RNAs, also regulate gene expression and have a role in the regulation of IRI pathways. We review the mechanisms and downstream effects of the miRNAs that have been studied as therapy in both permanent coronary ligation and ischemia-reperfusion models.
Collapse
|
41
|
Tu F, Wang X, Zhang X, Ha T, Wang Y, Fan M, Yang K, Gill PS, Ozment TR, Dai Y, Liu L, Williams DL, Li C. Novel Role of Endothelial Derived Exosomal HSPA12B in Regulating Macrophage Inflammatory Responses in Polymicrobial Sepsis. Front Immunol 2020; 11:825. [PMID: 32457753 PMCID: PMC7221167 DOI: 10.3389/fimmu.2020.00825] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 04/14/2020] [Indexed: 12/20/2022] Open
Abstract
Endothelial cell dysfunction contributes to sepsis induced initiate immune response and the infiltration of immune cells into organs, resulting in organ injury. Heat shock protein A12B (HSPA12B) is predominantly expressed in endothelial cells. The present study investigated whether endothelial HSPA12B could regulate macrophage pro-inflammatory response during sepsis. Wild type (WT) and endothelial cell-specific HSPA12B deficient (HSPA12B-/-) mice were subjected to CLP sepsis. Mortality and cardiac function were monitored. Higher mortality, worsened cardiac dysfunction, and greater infiltrated macrophages in the myocardium and spleen were observed in HSPA12B-/- septic mice compared with the WT septic mice. The serum levels of TNF-α and IL-1β were higher and the levels of IL-10 were lower in HSPA12B-/- septic mice than in WT septic mice. Importantly, endothelial exosomes contain HSPA12B which can be uptaken by macrophages. Interestingly, endothelial exosomal HSPA12B significantly increases IL-10 levels and decreases TNF-α and IL-1β production in LPS-stimulated macrophages. Mechanistic studies show that endothelial exosomal HSPA12B downregulates NF-κB activation and nuclear translocation in LPS stimulated macrophages. These data suggest that endothelial HSPA12B plays a novel role in the regulation of macrophage pro-inflammatory response via exosomes during sepsis and that sepsis induced cardiomyopathy and mortality are associated with endothelial cell deficiency of HSPA12B.
Collapse
Affiliation(s)
- Fei Tu
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States.,The Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Xiaohui Wang
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States.,The Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Xia Zhang
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States.,The Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Tuanzhu Ha
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States.,The Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Yana Wang
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Min Fan
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States.,The Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Kun Yang
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - P Spencer Gill
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States.,The Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Tammy R Ozment
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States.,The Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Yuan Dai
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Li Liu
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - David L Williams
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States.,The Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Chuanfu Li
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States.,The Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| |
Collapse
|
42
|
Ma C, Peng P, Zhou Y, Liu T, Wang L, Lu C. MicroRNA‑93 promotes angiogenesis and attenuates remodeling via inactivation of the Hippo/Yap pathway by targeting Lats2 after myocardial infarctionω. Mol Med Rep 2020; 22:483-493. [PMID: 32319642 PMCID: PMC7248469 DOI: 10.3892/mmr.2020.11085] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 04/01/2020] [Indexed: 01/10/2023] Open
Abstract
Inactivation of the Hippo pathway protects the myocardium from cardiac ischemic injury. MicroRNAs (miRs) have been reported to play pivotal roles in the progression of myocardial infarction (MI). The present study examined whether miR-93 could promote angiogenesis and attenuate remodeling after MI via inactivation of the Hippo/Yes-associated protein (Yap) pathway, by targeting large tumor suppressor kinase 2 (Lats2). It was identified that transfection of human umbilical vein endothelial cells with miR-93 mimic significantly decreased Lats2 expression and Yap phosphorylation, increased cell viability and migration, and attenuated cell apoptosis following hypoxia/reoxygenation injury. Moreover, increased expression of miR-93 resulted in an improvement of cardiac function, promotion of angiogenesis and attenuation of remodeling after MI. Additionally, miR-93 overexpression significantly decreased intracellular adhesion molecule 1 and vascular cell adhesion protein 1 expression levels, as well as attenuated the infiltration of neutrophils and macrophages into the myocardium after MI. Furthermore, it was found that miR-93 overexpression significantly suppressed Lats2 expression and decreased the levels of phosphorylated Yap in the myocardium after MI. Collectively, the present results suggested that miR-93 may exert a protective effect against MI via inactivation of the Hippo/Yap pathway by targeting Lats2.
Collapse
Affiliation(s)
- Chengjie Ma
- Clinical and Research Center of Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, P.R. China
| | - Peipei Peng
- Department of Anesthesiology, First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Yan Zhou
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Tianya Liu
- Jiangsu Province Key Laboratory of Anesthesiology, School of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Lijuan Wang
- Jiangsu Province Key Laboratory of Anesthesiology, School of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Chen Lu
- Jiangsu Province Key Laboratory of Anesthesiology, School of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| |
Collapse
|
43
|
Li Y, Zhou J, Zhang O, Wu X, Guan X, Xue Y, Li S, Zhuang X, Zhou B, Miao G, Zhang L. RETRACTED: Bone marrow mesenchymal stem cells-derived exosomal microRNA-185 represses ventricular remolding of mice with myocardial infarction by inhibiting SOCS2. Int Immunopharmacol 2020; 80:106156. [PMID: 31945609 DOI: 10.1016/j.intimp.2019.106156] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 12/09/2019] [Accepted: 12/25/2019] [Indexed: 02/06/2023]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). This article has been retracted at the request of the Editor-in-Chief. Concern was raised about the integrity of the images in Figures 5B and 7C, which appear to contain suspected duplications, as detailed here: https://pubpeer.com/publications/C968FDCECE2069D7FF43B346B261ED and here: https://docs.google.com/spreadsheets/d/1r0MyIYpagBc58BRF9c3luWNlCX8VUvUuPyYYXzxWvgY/edit#gid=262337249. Numerous additional suspected image duplications were detected within Figures 5 and 6. Most of these image duplications involve either pasting portions of one image into another, or rotating/flipping the image. The journal requested the corresponding author comment on these concerns and provide the raw data. The authors did not respond to this request and therefore the Editor-in-Chief decided to retract the article.
Collapse
Affiliation(s)
- Yanbing Li
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100043 China
| | - Jie Zhou
- Department of Cardiology, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China
| | - Ou Zhang
- Department of Cardiology, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China
| | - Xuejiao Wu
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100043 China
| | - Xiaonan Guan
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100043 China
| | - Yajun Xue
- Department of Cardiology, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China
| | - Siyuan Li
- School of Clinical Medicine, Tsinghua University, China
| | | | - Boda Zhou
- Department of Cardiology, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China
| | - Guobin Miao
- Department of Cardiology, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China.
| | - Lin Zhang
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100043 China.
| |
Collapse
|
44
|
Chen Q, Huang M, Wu J, Jiang Q, Zheng X. Exosomes isolated from the plasma of remote ischemic conditioning rats improved cardiac function and angiogenesis after myocardial infarction through targeting Hsp70. Aging (Albany NY) 2020; 12:3682-3693. [PMID: 32074081 PMCID: PMC7066898 DOI: 10.18632/aging.102837] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 01/27/2020] [Indexed: 12/15/2022]
Abstract
Remote ischemic conditioning (RIC) is a promising therapeutic strategy to protect heart against ischemic-reperfusion injury. Exosomes have been proved to be an important regulator in many pathological processes. Whether the exosomes derived from RIC could improve cardiac remodeling and function after myocardial infarction (MI) has not been reported. MI animal model was established by ligating the left coronary artery. The bilateral hindlimbs of rats were subjected to RIC treatment using tourniquets. Exosomes were isolated from the plasma of RIC rats and identified by transmission electron microscope. The proliferation, migration, and apoptosis of endothelial cells were measured by CCK8, traswell, and flow cytometry. Western blotting, and qRT-PCR were applied to measure the expression of angiogenesis-related molecules, and immunohistochemistry staining was used to observe the expression of vWF. RIC and RIC exosomes remarkably facilitated cardiac function, cardiac cell remodeling, and angiogenesis. RIC exosomes markedly increased the cell ratio in the G1 phase, cell migration, cell proliferation, tube formation, and inhibited cell apoptosis through Hsp70. The expression of eNOS, iNOS, HIF-1α, Ang-1, and VEGF was markedly increased by RIC exosomes. RIC exosomes significantly improved cardiac function, cardiac remodeling, and angiogenesis after MI, and they accelerated angiogenesis through increasing the levels of angiogenesis-related molecules.
Collapse
Affiliation(s)
- Qin Chen
- Department of Cardiology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China.,Fujian Institute of Coronary Artery Disease, Fuzhou, Fujian 350001, P.R. China.,Fujian Heart Medical Center, Fuzhou, Fujian 350001, P.R. China
| | - Minghan Huang
- The Second Affiliated Hospital of Fujian Traditional Chinese Medical University, Fuzhou, Fujian 350003, P.R. China
| | - Jiayi Wu
- Department of Cardiology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China.,Fujian Institute of Coronary Artery Disease, Fuzhou, Fujian 350001, P.R. China.,Fujian Heart Medical Center, Fuzhou, Fujian 350001, P.R. China
| | - Qiong Jiang
- Department of Cardiology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China.,Fujian Institute of Coronary Artery Disease, Fuzhou, Fujian 350001, P.R. China.,Fujian Heart Medical Center, Fuzhou, Fujian 350001, P.R. China
| | - Xingchun Zheng
- Department of Cardiology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China.,Fujian Institute of Coronary Artery Disease, Fuzhou, Fujian 350001, P.R. China.,Fujian Heart Medical Center, Fuzhou, Fujian 350001, P.R. China
| |
Collapse
|
45
|
Tian J, Popal MS, Zhao Y, Liu Y, Chen K, Liu Y. Interplay between Exosomes and Autophagy in Cardiovascular Diseases: Novel Promising Target for Diagnostic and Therapeutic Application. Aging Dis 2019; 10:1302-1310. [PMID: 31788341 PMCID: PMC6844582 DOI: 10.14336/ad.2018.1020] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 10/20/2018] [Indexed: 12/12/2022] Open
Abstract
Exosome, is identified as a nature nanocarrier and intercellular messenger that regulates cell to cell communication. Autophagy is critical in maintenance of protein homeostasis by degradation of damaged proteins and organelles. Autophagy and exosomes take pivotal roles in cellular homeostasis and cardiovascular disease. Currently, the coordinated mechanisms for exosomes and autophagy in the maintenance of cellular fitness are now garnering much attention. In the present review, we discussed the interplay of exosomes and autophagy in the context of physiology and pathology of the heart, which might provide novel insights for diagnostic and therapeutic application of cardiovascular diseases.
Collapse
Affiliation(s)
- Jinfan Tian
- 1Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,2Cardiovascular disease center, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Mohammad Sharif Popal
- 1Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yingke Zhao
- 3Li Ka Shing Faculty of Medicine, The University of HongKong, Pokfulam, Hong Kong
| | - Yanfei Liu
- 4Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Keji Chen
- 2Cardiovascular disease center, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yue Liu
- 2Cardiovascular disease center, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
46
|
Li X, Zhang S, Wa M, Liu Z, Hu S. MicroRNA-101 Protects Against Cardiac Remodeling Following Myocardial Infarction via Downregulation of Runt-Related Transcription Factor 1. J Am Heart Assoc 2019; 8:e013112. [PMID: 31766975 PMCID: PMC6912979 DOI: 10.1161/jaha.119.013112] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background Myocardial infarction (MI) generally leads to heart failure and sudden death. The hearts of people with MI undergo remodeling with the features of expanded myocardial infarct size and dilated left ventricle. Many microRNAs (miRs) have been revealed to be involved in the remodeling process; however, the participation of miR‐101 remains unknown. Therefore, this study aims to find out the regulatory mechanism of miR‐101 in MI‐induced cardiac remodeling. Methods and Results Microarray data analysis was conducted to screen differentially expressed genes in MI. The rat model of MI was established by left coronary artery ligation. In addition, the relationship between miR‐101 and runt‐related transcription factor 1 (RUNX1) was identified using dual luciferase reporter assay. After that, the rats injected with lentiviral vector expressing miR‐101 mimic, inhibitor, or small interfering RNA against RUNX1 were used to examine the effects of miR‐101 and RUNX1 on transforming growth factor β signaling pathway, cardiac function, infarct size, myocardial fibrosis, and cardiomyocyte apoptosis. RUNX1 was highly expressed, while miR‐101 was poorly expressed in MI. miR‐101 was identified to target RUNX1. Following that, it was found that overexpression of miR‐101 or silencing of RUNX1 improved the cardiac function and elevated left ventricular end‐diastolic and end‐systolic diameters. Also, miR‐101 elevation or RUNX1 depletion decreased infarct size, myocardial fibrosis, and cardiomyocyte apoptosis. Moreover, miR‐101 could negatively regulate RUNX1 to inactivate the transforming growth factor β1/Smad family member 2 signaling pathway. Conclusions Taken together, miR‐101 plays a protective role against cardiac remodeling following MI via inactivation of the RUNX1‐dependent transforming growth factor β1/Smad family member 2 signaling pathway, proposing miR‐101 and RUNX1 as potential therapeutic targets for MI.
Collapse
Affiliation(s)
- Xidong Li
- Department of Cardiology Linyi People's Hospital Linyi China
| | - Shouwen Zhang
- Department of Cardiology Linyi People's Hospital Linyi China
| | | | - Zhonghua Liu
- Department of Endocrinology Linyi People's Hospital Linyi China
| | - Shunpeng Hu
- Department of Cardiology Linyi People's Hospital Linyi China
| |
Collapse
|
47
|
The role of toll-like receptors in myocardial toxicity induced by doxorubicin. Immunol Lett 2019; 217:56-64. [PMID: 31707054 DOI: 10.1016/j.imlet.2019.11.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 10/21/2019] [Accepted: 11/06/2019] [Indexed: 02/06/2023]
Abstract
Doxorubicin is an effective antitumor drug commonly used in the treatment of a wide variety of cancers. However, doxorubicin may cause cardiac toxicity, which can cause congestive heart failure in severe cases, and this seriously limits its clinical application. It is believed that doxorubicin promotes the formation of reactive oxygen species, inducing oxidative stress, and at the same time, reduces the content of antioxidant substances in cardiac tissues, causing adverse effects. Toll-like receptors (TLRs) are biomolecules expressed on the surfaces of macrophages, dendritic cells, and epithelial cells that can recognize various types of pathogen-related or damage-related molecular patterns. In recent years, a large number of studies have confirmed that TLRs play important roles in the cardiac toxicity induced by doxorubicin. This review aimed to explore the role of TLRs in the cardiac toxicity induced by doxorubicin and provide possible solutions.
Collapse
|
48
|
Zhang L, Zhang J, Tong Q, Wang G, Dong H, Wang Z, Sun Q, Wu H. Reduction of miR-29a-3p induced cardiac ischemia reperfusion injury in mice via targeting Bax. Exp Ther Med 2019; 18:1729-1737. [PMID: 31410131 PMCID: PMC6676207 DOI: 10.3892/etm.2019.7722] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 04/11/2019] [Indexed: 12/14/2022] Open
Abstract
The current study mainly aimed to evaluate the expression and the potential mechanism of miR-29a-3p in the hearts of mice after cardiac ischemia reperfusion (CIR) injury. Quantitative PCR was carried out to assess the relative levels of miR-29a-3p in the hearts of a CIR injury mouse model. To the best of our knowledge, the current study is the first to show that the level of miR-29a-3p was significantly decreased in the hearts of CIR injury mouse models compared with that of sham controls. Moreover, the authors found that decreased miR-29a-3p levels enhanced the production of reactive oxygen species in cardiomyocytes. Meanwhile, the inhibition of miR-29a-3p induced substantial cardiomyocyte apoptosis. Further study showed that the inhibition of miR-29a-3p decreased the activation of Akt and p38, suggesting a stress-induced self-regulatory mechanism after CIR injury in primary cardiomyocytes. A dual luciferase assay and western blot analysis showed that Bax was a target gene of miR-29a-3p. The authors also measured the level of miR-29a-3p in the plasma of 100 acute myocardial infarction (AMI) patients and found that circulating miR-29a-3p was significantly decreased in AMI patients. Receiver operating characteristic curve analysis showed that miR-29a-3p could be used to screen AMI patients from healthy controls. Hence, the authors of the current study propose that reduced miR-29a-3p levels in primary cardiomyocytes contribute to CIR injury-related apoptosis mainly by targeting Bax.
Collapse
Affiliation(s)
- Liang Zhang
- Heart Center, Beijing Chest Hospital, Capital Medical University, Beijing 101149, P.R. China
| | - Jian Zhang
- Heart Center, Beijing Chest Hospital, Capital Medical University, Beijing 101149, P.R. China
| | - Qiguang Tong
- Heart Center, Beijing Chest Hospital, Capital Medical University, Beijing 101149, P.R. China
| | - Guannan Wang
- Heart Center, Beijing Chest Hospital, Capital Medical University, Beijing 101149, P.R. China
| | - Hongling Dong
- Heart Center, Beijing Chest Hospital, Capital Medical University, Beijing 101149, P.R. China
| | - Zhonglu Wang
- Heart Center, Beijing Chest Hospital, Capital Medical University, Beijing 101149, P.R. China
| | - Qi Sun
- Heart Center, Beijing Chest Hospital, Capital Medical University, Beijing 101149, P.R. China
| | - Hangyu Wu
- Heart Center, Beijing Chest Hospital, Capital Medical University, Beijing 101149, P.R. China
| |
Collapse
|
49
|
Chouvarine P, Legchenko E, Geldner J, Riehle C, Hansmann G. Hypoxia drives cardiac miRNAs and inflammation in the right and left ventricle. J Mol Med (Berl) 2019; 97:1427-1438. [PMID: 31338525 DOI: 10.1007/s00109-019-01817-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 06/20/2019] [Accepted: 07/02/2019] [Indexed: 12/16/2022]
Abstract
Alveolar and myocardial hypoxia may be causes or sequelae of pulmonary hypertension (PH) and heart failure. We hypothesized that hypoxia initiates specific epigenetic and transcriptional, pro-inflammatory programs in the right ventricle (RV) and left ventricle (LV). We performed an expression screen of 750 miRNAs by qPCR arrays in the murine RV and LV in normoxia (Nx) and hypoxia (Hx; 10% O2 for 18 h, 48 h, and 5d). Additional validation included single qPCR analysis of miRNA and pro-inflammatory transcripts in murine and human RV/LV, and neonatal rat cardiomyocytes (NRCMs). Differential qPCR-analysis (Hx vs. Nx in RV, Hx vs. Nx in LV, and RV vs. LV in Hx) identified nine hypoxia-regulated miRNAs: let-7e-5p, miR-29c-3p, miR-127-3p, miR-130a-3p, miR-146b-5p, miR-197-3p, miR-214-3p, miR-223-3p, and miR-451. Hypoxia downregulated miR-146b in the RV (p < 0.01) and, less so, in the LV (trend; p = 0.28). In silico alignment showed significant binding affinity of miR-146b-5p sequence with the 3'UTR of TRAF6 known to be upstream of pro-inflammatory NF-kB. Consistently, hypoxia induced TRAF6, IL-6, CCL2(MCP-1) in the mouse RV and LV. Incubating neonatal rat cardiomyocytes with pre-miR-146b led to a downregulation of TRAF6, IL-6, and CCL2(MCP-1). TRAF6 mRNA expression was also increased by 3-fold in the RV and LV of end-stage idiopathic pulmonary arterial hypertension (PAH) patients vs. non-PAH controls. We identified hypoxia-regulated, ventricle-specific miRNA expression profiles in the adult mouse heart in vivo. Hypoxia suppresses miR-146b, thus de-repressing TRAF6, and inducing pro-inflammatory IL-6 and CCL2(MCP-1). This novel hypoxia-induced miR-146b-TRAF6-IL-6/CCL2(MCP-1) axis likely drives cardiac fibrosis and dysfunction, and may lead to heart failure. KEY MESSAGES: Chouvarine P, Legchenko E, Geldner J, Riehle C, Hansmann G. Hypoxia drives cardiac miRNAs and inflammation in the right and left ventricle. • Hypoxia drives ventricle-specific miRNA profiles, regulating cardiac inflammation. • miR-146b-5p downregulates TRAF6, known to act upstream of pro-inflammatory NF-κB. • Hypoxia downregulates miR-146b and induces TRAF6, IL-6, CCL2 (MCP-1) in the murine RV and LV. • The inhibitory regulatory effects of miR-146b are confirmed in primary rat cardiomyocytes (pre-miR, anti-miR) and human explant heart tissue (endstage pulmonary arterial hypertension). • A novel miR-146b-TRAF6-IL-6/CCL2(MCP-1) axis likely drives cardiac inflammation, fibrosis and ventricular dysfunction.
Collapse
Affiliation(s)
- Philippe Chouvarine
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Ekaterina Legchenko
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Jonas Geldner
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Christian Riehle
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Georg Hansmann
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| |
Collapse
|
50
|
Hardy SA, Mabotuwana NS, Murtha LA, Coulter B, Sanchez-Bezanilla S, Al-Omary MS, Senanayake T, Loering S, Starkey M, Lee RJ, Rainer PP, Hansbro PM, Boyle AJ. Novel role of extracellular matrix protein 1 (ECM1) in cardiac aging and myocardial infarction. PLoS One 2019; 14:e0212230. [PMID: 30789914 PMCID: PMC6383988 DOI: 10.1371/journal.pone.0212230] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 01/29/2019] [Indexed: 12/28/2022] Open
Abstract
INTRODUCTION The prevalence of heart failure increases in the aging population and following myocardial infarction (MI), yet the extracellular matrix (ECM) remodeling underpinning the development of aging- and MI-associated cardiac fibrosis remains poorly understood. A link between inflammation and fibrosis in the heart has long been appreciated, but has mechanistically remained undefined. We investigated the expression of a novel protein, extracellular matrix protein 1 (ECM1) in the aging and infarcted heart. METHODS Young adult (3-month old) and aging (18-month old) C57BL/6 mice were assessed. Young mice were subjected to left anterior descending artery-ligation to induce MI, or transverse aortic constriction (TAC) surgery to induce pressure-overload cardiomyopathy. Left ventricle (LV) tissue was collected early and late post-MI/TAC. Bone marrow cells (BMCs) were isolated from young healthy mice, and subject to flow cytometry. Human cardiac fibroblast (CFb), myocyte, and coronary artery endothelial & smooth muscle cell lines were cultured; human CFbs were treated with recombinant ECM1. Primary mouse CFbs were cultured and treated with recombinant angiotensin-II or TGF-β1. Immunoblotting, qPCR and mRNA fluorescent in-situ hybridization (mRNA-FISH) were conducted on LV tissue and cells. RESULTS ECM1 expression was upregulated in the aging LV, and in the infarct zone of the LV early post-MI. No significant differences in ECM1 expression were found late post-MI or at any time-point post-TAC. ECM1 was not expressed in any resident cardiac cells, but ECM1 was highly expressed in BMCs, with high ECM1 expression in granulocytes. Flow cytometry of bone marrow revealed ECM1 expression in large granular leucocytes. mRNA-FISH revealed that ECM1 was indeed expressed by inflammatory cells in the infarct zone at day-3 post-MI. ECM1 stimulation of CFbs induced ERK1/2 and AKT activation and collagen-I expression, suggesting a pro-fibrotic role. CONCLUSIONS ECM1 expression is increased in ageing and infarcted hearts but is not expressed by resident cardiac cells. Instead it is expressed by bone marrow-derived granulocytes. ECM1 is sufficient to induce cardiac fibroblast stimulation in vitro. Our findings suggest ECM1 is released from infiltrating inflammatory cells, which leads to cardiac fibroblast stimulation and fibrosis in aging and MI. ECM1 may be a novel intermediary between inflammation and fibrosis.
Collapse
Affiliation(s)
- Sean A. Hardy
- School of Medicine and Public Health, The University of Newcastle, Callaghan, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Nishani S. Mabotuwana
- School of Medicine and Public Health, The University of Newcastle, Callaghan, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Lucy A. Murtha
- School of Medicine and Public Health, The University of Newcastle, Callaghan, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Brianna Coulter
- School of Medicine and Public Health, The University of Newcastle, Callaghan, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Sonia Sanchez-Bezanilla
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia
- Priority Research Centre’s for Healthy Lungs and GrowUpWell, School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia
| | - Mohammed S. Al-Omary
- School of Medicine and Public Health, The University of Newcastle, Callaghan, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- Department of Cardiovascular Medicine, John Hunter Hospital, New Lambton Heights, NSW, Australia
| | - Tharindu Senanayake
- School of Medicine and Public Health, The University of Newcastle, Callaghan, NSW, Australia
| | - Svenja Loering
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia
- Priority Research Centre’s for Healthy Lungs and GrowUpWell, School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia
| | - Malcolm Starkey
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia
- Priority Research Centre’s for Healthy Lungs and GrowUpWell, School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia
| | - Randall J. Lee
- Department of Medicine, Division of Cardiology, University of California San Francisco, San Francisco, CA, United States of America
- Edyth and Eli Broad Center for Regenerative Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, United States of America
| | - Peter P. Rainer
- Division of Cardiology, Medical University of Graz, Graz, Austria
| | - Philip M. Hansbro
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia
- Priority Research Centre’s for Healthy Lungs and GrowUpWell, School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia
- Centre for inflammation, Centenary Institute, Sydney, NSW, Australia
- University of Technology, Faculty of Science, Ultimo, NSW, Australia
| | - Andrew J. Boyle
- School of Medicine and Public Health, The University of Newcastle, Callaghan, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- Department of Cardiovascular Medicine, John Hunter Hospital, New Lambton Heights, NSW, Australia
- * E-mail:
| |
Collapse
|