1
|
Wójtowicz A, Molcan T, Lukasik K, Żebrowska E, Pawlina-Tyszko K, Gurgul A, Szmatoła T, Bugno-Poniewierska M, Ferreira-Dias G, Skarzynski DJ, Szóstek-Mioduchowska A. The potential role of miRNAs and regulation of their expression in the development of mare endometrial fibrosis. Sci Rep 2023; 13:15938. [PMID: 37743390 PMCID: PMC10518347 DOI: 10.1038/s41598-023-42149-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 09/06/2023] [Indexed: 09/26/2023] Open
Abstract
Mare endometrial fibrosis (endometrosis), is one of the main causes of equine infertility. Despite the high prevalence, both ethology, pathogenesis and the nature of its progression remain poorly understood. Recent studies have shown that microRNAs (miRNAs) are important regulators in multiple cellular processes and functions under physiological and pathological circumstances. In this article, we reported changes in miRNA expression at different stages of endometrosis and the effect of transforming growth factor (TGF)-β1 on the expression of the most dysregulated miRNAs. We identified 1, 26, and 5 differentially expressed miRNAs (DEmiRs), in categories IIA (mild fibrosis), IIB (moderate fibrosis), and III (severe fibrosis) groups compared to category I (no fibrosis) endometria group, respectively (Padjusted < 0.05, log2FC ≥ 1.0/log2FC ≤ - 1.0). This study indicated the potential involvement of miRNAs in the regulation of the process associated to the development and progression of endometrosis. The functional enrichment analysis revealed, that DEmiRs target genes involved in the mitogen-activated protein kinases, Hippo, and phosphoinositide-3-kinase (PI3K)-Akt signalling pathways, focal adhesion, and extracellular matrix-receptor interaction. Moreover, we demonstrated that the most potent profibrotic cytokine-TGF-β1-downregulated novel-eca-miR-42 (P < 0.05) expression in fibroblasts derived from endometria at early-stage endometrosis (category IIA).
Collapse
Affiliation(s)
- Anna Wójtowicz
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland
| | - Tomasz Molcan
- Molecular Biology Laboratory, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland
| | - Karolina Lukasik
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland
| | - Ewelina Żebrowska
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland
| | - Klaudia Pawlina-Tyszko
- Department of Animal Molecular Biology, National Research Institute of Animal Production, Cracow, Poland
| | - Artur Gurgul
- Department of Animal Reproduction, Anatomy and Genomics, The University of Agriculture in Krakow, Cracow, Poland
| | - Tomasz Szmatoła
- Department of Animal Molecular Biology, National Research Institute of Animal Production, Cracow, Poland
- Center for Experimental and Innovative Medicine, University of Agriculture in Krakow, Cracow, Poland
| | - Monika Bugno-Poniewierska
- Department of Animal Reproduction, Anatomy and Genomics, The University of Agriculture in Krakow, Cracow, Poland
| | - Graca Ferreira-Dias
- Faculty of Veterinary Medicine, CIISA - Center for Interdisciplinary Research in Animal Health, University of Lisbon, Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Lisbon, Portugal
| | - Dariusz J Skarzynski
- Department of Reproduction and Clinic of Farm Animals, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, Wroclaw, Poland
| | - Anna Szóstek-Mioduchowska
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland.
| |
Collapse
|
2
|
Chen J, Liu Z, Ma L, Gao S, Fu H, Wang C, Lu A, Wang B, Gu X. Targeting Epigenetics and Non-coding RNAs in Myocardial Infarction: From Mechanisms to Therapeutics. Front Genet 2022; 12:780649. [PMID: 34987550 PMCID: PMC8721121 DOI: 10.3389/fgene.2021.780649] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 11/30/2021] [Indexed: 12/12/2022] Open
Abstract
Myocardial infarction (MI) is a complicated pathology triggered by numerous environmental and genetic factors. Understanding the effect of epigenetic regulation mechanisms on the cardiovascular disease would advance the field and promote prophylactic methods targeting epigenetic mechanisms. Genetic screening guides individualised MI therapies and surveillance. The present review reported the latest development on the epigenetic regulation of MI in terms of DNA methylation, histone modifications, and microRNA-dependent MI mechanisms and the novel therapies based on epigenetics.
Collapse
Affiliation(s)
- Jinhong Chen
- Department of TCM, Tianjin University of TCM, Tianjin, China
| | - Zhichao Liu
- Department of TCM, Tianjin University of TCM, Tianjin, China
| | - Li Ma
- Department of TCM, Tianjin University of TCM, Tianjin, China
| | - Shengwei Gao
- Department of TCM, Tianjin University of TCM, Tianjin, China
| | - Huanjie Fu
- Department of TCM, Tianjin University of TCM, Tianjin, China
| | - Can Wang
- Acupuncture Department, The First Affiliated Hospital of Tianjin University of TCM, Tianjin, China
| | - Anmin Lu
- Department of TCM, Tianjin University of TCM, Tianjin, China
| | - Baohe Wang
- Department of Cardiology, The Second Affiliated Hospital of Tianjin University of TCM, Tianjin, China
| | - Xufang Gu
- Department of Cardiology, The Second Affiliated Hospital of Tianjin University of TCM, Tianjin, China
| |
Collapse
|
3
|
MicroRNAs-The Heart of Post-Myocardial Infarction Remodeling. Diagnostics (Basel) 2021; 11:diagnostics11091675. [PMID: 34574016 PMCID: PMC8469128 DOI: 10.3390/diagnostics11091675] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 09/02/2021] [Accepted: 09/09/2021] [Indexed: 12/20/2022] Open
Abstract
Myocardial infarction (MI) is one of the most frequent cardiac emergencies, with significant potential for mortality. One of the major challenges of the post-MI healing response is that replacement fibrosis could lead to left ventricular remodeling (LVR) and heart failure (HF). This process involves canonical and non-canonical transforming growth factor-beta (TGF-β) signaling pathways translating into an intricate activation of cardiac fibroblasts and disproportionate collagen synthesis. Accumulating evidence has indicated that microRNAs (miRNAs) significantly contribute to the modulation of these signaling pathways. This review summarizes the recent updates regarding the molecular mechanisms underlying the role of the over 30 miRNAs involved in post-MI LVR. In addition, we compare the contradictory roles of several multifunctional miRNAs and highlight their potential use in pressure overload and ischemia-induced fibrosis. Finally, we discuss their attractive role as prognostic biomarkers for HF, highlighting the most relevant human trials involving these miRNAs.
Collapse
|
4
|
Xu L, Hu G, Xing P, Zhou M, Wang D. Paclitaxel alleviates the sepsis-induced acute kidney injury via lnc-MALAT1/miR-370-3p/HMGB1 axis. Life Sci 2020; 262:118505. [PMID: 32998017 DOI: 10.1016/j.lfs.2020.118505] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 09/21/2020] [Accepted: 09/23/2020] [Indexed: 12/11/2022]
Abstract
AIMS To investigate the effects of paclitaxel on lipopolysaccharide (LPS)-induced acute kidney injury (AKI) and its related mechanisms. MAIN METHODS The sepsis-associated AKI was induced by LPS using HK-2 cells. Then the mRNA and protein expression levels of relevant genes in the serum of sepsis patients and HK-2 cells with LPS-induced AKI were detected by qRT-PCR and western blot analyses before and after paclitaxel treatment, respectively. Subsequently, the cell counting kit-8 (CCK-8) and flow cytometry assays were performed to estimate the effects of paclitaxel, lnc-MALAT1, miR-370-3p and HMGB1 on the proliferation and apoptosis of HK-2 cells injured by LPS. KEY FINDINGS Lnc-MALAT1 was increased both in the serum of sepsis patients and cells injured by LPS, which could inhibit the cell proliferation, promote the cell apoptosis and increase the expression of TNF-α, IL-6 and IL-1β caused by paclitaxel. Moreover, lnc-MALAT1 was sponged with miR-370-3p which had the inverse effects with lnc-MALAT1 in LPS induced HK-2 cells. What's more, miR-370-3p targeted HMGB1 which was induced in serum and cells of sepsis. Knockdown of miR-370-3p inhibited the expression of HMGB1 and suppressed the proliferation but promoted the apoptosis of HK-2 cells injured by LPS as well as the expression of TNF-α, IL-6 and IL-1β. Besides, paclitaxel restrained the expression of HMGB1 via regulating lnc-MALAT1/miR-370-3p axis. SIGNIFICANCE Paclitaxel could protect against LPS-induced AKI via the regulation of lnc-MALAT1/miR-370-3p/HMGB1 axis and the expression of TNF-α, IL-6 and IL-1β, revealing that paclitaxel might act as a therapy drug in reducing sepsis-associated AKI.
Collapse
Affiliation(s)
- Lina Xu
- Department of Infectious Diseases, Shanghai University of Medicine & Health Sciences Affiliated Shanghai Sixth People's Hospital East, No. 222, West Three Road Around Lake, Pudong District, Shanghai 201306, PR China
| | - Guyong Hu
- Department of Emergency, Shanghai University of Medicine & Health Sciences Affiliated Shanghai Sixth People's Hospital East, No. 222, West Three Road Around Lake, Pudong District, Shanghai 201306, PR China
| | - Pengcheng Xing
- Department of Emergency, Shanghai University of Medicine & Health Sciences Affiliated Shanghai Sixth People's Hospital East, No. 222, West Three Road Around Lake, Pudong District, Shanghai 201306, PR China.
| | - Minjie Zhou
- Department of Emergency, Shanghai University of Medicine & Health Sciences Affiliated Shanghai Sixth People's Hospital East, No. 222, West Three Road Around Lake, Pudong District, Shanghai 201306, PR China
| | - Donglian Wang
- Department of Emergency, Shanghai University of Medicine & Health Sciences Affiliated Shanghai Sixth People's Hospital East, No. 222, West Three Road Around Lake, Pudong District, Shanghai 201306, PR China
| |
Collapse
|
5
|
Yanni J, D'Souza A, Wang Y, Li N, Hansen BJ, Zakharkin SO, Smith M, Hayward C, Whitson BA, Mohler PJ, Janssen PML, Zeef L, Choudhury M, Zi M, Cai X, Logantha SJRJ, Nakao S, Atkinson A, Petkova M, Doris U, Ariyaratnam J, Cartwright EJ, Griffiths-Jones S, Hart G, Fedorov VV, Oceandy D, Dobrzynski H, Boyett MR. Silencing miR-370-3p rescues funny current and sinus node function in heart failure. Sci Rep 2020; 10:11279. [PMID: 32647133 PMCID: PMC7347645 DOI: 10.1038/s41598-020-67790-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 06/02/2020] [Indexed: 01/13/2023] Open
Abstract
Bradyarrhythmias are an important cause of mortality in heart failure and previous studies indicate a mechanistic role for electrical remodelling of the key pacemaking ion channel HCN4 in this process. Here we show that, in a mouse model of heart failure in which there is sinus bradycardia, there is upregulation of a microRNA (miR-370-3p), downregulation of the pacemaker ion channel, HCN4, and downregulation of the corresponding ionic current, If, in the sinus node. In vitro, exogenous miR-370-3p inhibits HCN4 mRNA and causes downregulation of HCN4 protein, downregulation of If, and bradycardia in the isolated sinus node. In vivo, intraperitoneal injection of an antimiR to miR-370-3p into heart failure mice silences miR-370-3p and restores HCN4 mRNA and protein and If in the sinus node and blunts the sinus bradycardia. In addition, it partially restores ventricular function and reduces mortality. This represents a novel approach to heart failure treatment.
Collapse
Affiliation(s)
- Joseph Yanni
- Division of Cardiovascular Sciences, University of Manchester, 46 Grafton Street, Manchester, M13 9NT, UK
| | - Alicia D'Souza
- Division of Cardiovascular Sciences, University of Manchester, 46 Grafton Street, Manchester, M13 9NT, UK
| | - Yanwen Wang
- Division of Cardiovascular Sciences, University of Manchester, 46 Grafton Street, Manchester, M13 9NT, UK
| | - Ning Li
- Physiology and Cell Biology, Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
- Bob and Corrine Frick Center for Heart Failure and Arrhythmia Research and Dorothy M. Davis Heart and Lung Research Institute, Ohio State University, Columbus, OH, 43210, USA
| | - Brian J Hansen
- Physiology and Cell Biology, Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
- Bob and Corrine Frick Center for Heart Failure and Arrhythmia Research and Dorothy M. Davis Heart and Lung Research Institute, Ohio State University, Columbus, OH, 43210, USA
| | - Stanislav O Zakharkin
- Physiology and Cell Biology, Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Matthew Smith
- Division of Cardiovascular Sciences, University of Manchester, 46 Grafton Street, Manchester, M13 9NT, UK
| | - Christina Hayward
- Division of Cardiovascular Sciences, University of Manchester, 46 Grafton Street, Manchester, M13 9NT, UK
| | - Bryan A Whitson
- Bob and Corrine Frick Center for Heart Failure and Arrhythmia Research and Dorothy M. Davis Heart and Lung Research Institute, Ohio State University, Columbus, OH, 43210, USA
- Department of Surgery, Division of Cardiac Surgery, Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Peter J Mohler
- Physiology and Cell Biology, Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
- Bob and Corrine Frick Center for Heart Failure and Arrhythmia Research and Dorothy M. Davis Heart and Lung Research Institute, Ohio State University, Columbus, OH, 43210, USA
- Department of Internal Medicine, Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Paul M L Janssen
- Physiology and Cell Biology, Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
- Bob and Corrine Frick Center for Heart Failure and Arrhythmia Research and Dorothy M. Davis Heart and Lung Research Institute, Ohio State University, Columbus, OH, 43210, USA
- Department of Internal Medicine, Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Leo Zeef
- Bioinformatics Core Facility, University of Manchester, Manchester, UK
| | - Moinuddin Choudhury
- Division of Cardiovascular Sciences, University of Manchester, 46 Grafton Street, Manchester, M13 9NT, UK
| | - Min Zi
- Division of Cardiovascular Sciences, University of Manchester, 46 Grafton Street, Manchester, M13 9NT, UK
| | - Xue Cai
- Division of Cardiovascular Sciences, University of Manchester, 46 Grafton Street, Manchester, M13 9NT, UK
| | - Sunil Jit R J Logantha
- Division of Cardiovascular Sciences, University of Manchester, 46 Grafton Street, Manchester, M13 9NT, UK
- Liverpool Centre for Cardiovascular Science, University of Liverpool, Liverpool, UK
| | - Shu Nakao
- Division of Cardiovascular Sciences, University of Manchester, 46 Grafton Street, Manchester, M13 9NT, UK
| | - Andrew Atkinson
- Division of Cardiovascular Sciences, University of Manchester, 46 Grafton Street, Manchester, M13 9NT, UK
| | - Maria Petkova
- Division of Cardiovascular Sciences, University of Manchester, 46 Grafton Street, Manchester, M13 9NT, UK
| | - Ursula Doris
- Division of Cardiovascular Sciences, University of Manchester, 46 Grafton Street, Manchester, M13 9NT, UK
| | - Jonathan Ariyaratnam
- Division of Cardiovascular Sciences, University of Manchester, 46 Grafton Street, Manchester, M13 9NT, UK
| | - Elizabeth J Cartwright
- Division of Cardiovascular Sciences, University of Manchester, 46 Grafton Street, Manchester, M13 9NT, UK
| | - Sam Griffiths-Jones
- Division of Evolution and Genomics Sciences, University of Manchester, Manchester, UK
| | - George Hart
- Division of Cardiovascular Sciences, University of Manchester, 46 Grafton Street, Manchester, M13 9NT, UK
| | - Vadim V Fedorov
- Physiology and Cell Biology, Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
- Bob and Corrine Frick Center for Heart Failure and Arrhythmia Research and Dorothy M. Davis Heart and Lung Research Institute, Ohio State University, Columbus, OH, 43210, USA
| | - Delvac Oceandy
- Division of Cardiovascular Sciences, University of Manchester, 46 Grafton Street, Manchester, M13 9NT, UK
| | - Halina Dobrzynski
- Division of Cardiovascular Sciences, University of Manchester, 46 Grafton Street, Manchester, M13 9NT, UK
- Department of Anatomy, Jagiellonian University Medical College, Kraków, Poland
| | - Mark R Boyett
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200N, Copenhagen, Denmark.
| |
Collapse
|
6
|
Qiu Z, Wang L, Mao H, Xu F, Sun B, Lian X, Wang J, Kong F, Wang L, Chen Y. miR-370 inhibits the oxidative stress and apoptosis of cardiac myocytes induced by hydrogen peroxide by targeting FOXO1. Exp Ther Med 2019; 18:3025-3031. [PMID: 31555385 PMCID: PMC6755418 DOI: 10.3892/etm.2019.7908] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 07/05/2019] [Indexed: 12/27/2022] Open
Abstract
Myocardial infarction, one of the main factors that threatens human health, leads to cardiac cell death. Myocardial cells suffer ischemia and hypoxia for a long period of time, which can lead to irreversible cell death or apoptosis and cardiac dysfunction. MicroRNAs (miRs) have been reported to play an important role in a wide range of biological processes in cardiac myocytes, which respond to inflammation and oxidative stress. The aim of the present study was to investigate the effect of miR-370 on oxidative stress and apoptosis of cardiac myocytes in ischemic H9C2 cells induced by hydrogen peroxide (H2O2). H9C2 cells were cultured and treated with different concentrations of H2O2 solution. Then, cells were transfected with miR-370 mimic or negative control (NC) mimic, small interfering (si)-RNA-Forkhead box O1 (FOXO1) and NC siRNA. A Cell Counting Kit-8 and flow cytometry assay were conducted to detect cell viability and cell apoptosis. The expression of oxidative stress associated factors were detected by ELISA. The levels of miR-370 and FOXO1 were examined using western blotting and reverse transcription-quantitative PCR. A luciferase reporter gene assay was performed to verify whether FOXO1 was a target gene of miR-370. The results revealed that miR-370 expression was downregulated and FOXO1 expression was increased in H9C2 cells induced by H2O2. Additionally, FOXO1 was proven to be a target of miR-370. The ELISA and flow cytometry assay revealed that miR-370 overexpression and FOXO1 silencing reversed H2O2-induced oxidative stress and apoptosis. The results indicated that miR-370 could inhibit the oxidative stress and apoptosis of H9C2 cells induced by H2O2 by targeting FOXO1. Therefore, miR-370 may be a new therapeutic target for ischemic heart disease.
Collapse
Affiliation(s)
- Zhanjun Qiu
- Department of Emergency Medicine and Chest Pain Center, Affiliated Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China.,Department of Emergency and Critical Care Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, P.R. China.,Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Jinan, Shandong 250012, P.R. China.,Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China.,The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine; Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Lei Wang
- Department of Emergency and Critical Care Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, P.R. China
| | - Huaiyu Mao
- Department of Emergency Medicine, The Second People's Hospital of Dongying, Dongying, Shandong 257335, P.R. China
| | - Feng Xu
- Department of Emergency Medicine and Chest Pain Center, Affiliated Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China.,Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Jinan, Shandong 250012, P.R. China.,Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China.,The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine; Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Bin Sun
- Department of Emergency Medicine, Yidu Center Hospital of Weifang, Weifang, Shandong 262500, P.R. China
| | - Xinbao Lian
- Department of Emergency and Critical Care Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, P.R. China
| | - Jiali Wang
- Department of Emergency Medicine and Chest Pain Center, Affiliated Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China.,Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Jinan, Shandong 250012, P.R. China.,Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China.,The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine; Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Feng Kong
- Department of Central Laboratory, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Lina Wang
- Department of Clinical Laboratory Medicine, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Yuguo Chen
- Department of Emergency Medicine and Chest Pain Center, Affiliated Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China.,Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Jinan, Shandong 250012, P.R. China.,Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China.,The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine; Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
7
|
Zhang Y, Zhu Y, Gao G, Zhou Z. Knockdown XIST alleviates LPS-induced WI-38 cell apoptosis and inflammation injury via targeting miR-370-3p/TLR4 in acute pneumonia. Cell Biochem Funct 2019; 37:348-358. [PMID: 31066476 PMCID: PMC6618287 DOI: 10.1002/cbf.3392] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Accepted: 04/02/2019] [Indexed: 12/14/2022]
Abstract
Pneumonia is an inflammatory disease that occurs in the lungs associated with pathogens or other factors. It has been well established that long noncoding RNA X inactivate-specific transcript (XIST) is involved in several cancers. The present study focused on the effect and detailed mechanism of XIST in lipopolysaccharide (LPS)-induced injury in pneumonia. Here, XIST was silenced by transfection with XIST-targeted siRNA, and then, mRNA expression, cell viability, apoptosis, and protein expression were, respectively, assessed by qRT-PCR, CCK-8, flow cytometry, and Western blotting. Luciferase reporter, RIP, and RNA pull-down assays were used to detect the combination of miR-370-3p and XIST. Besides, the tested proinflammatory factors were analysed by qRT-PCR and Western blot, and their productions were quantified by ELISA. The results showed that XIST expression was robustly increased in serum of patients with acute-stage pneumonia and LPS-induced WI-38 human lung fibroblasts cells. Functional analyses demonstrated that knockdown of XIST remarkably alleviated LPS-induced cell injury through increasing cell viability and inhibiting apoptosis and inflammatory cytokine levels. Mechanistically, XIST functioned as a competitive endogenous RNA (ceRNA) by effectively binding to miR-370-3p and then restoring TLR4 expression. More importantly, miR-370-3p inhibitor abolished the function of XIST knockdown on cell injury and JAK/STAT and NF-κB pathways. Taken together, XIST may be involved in progression of cell inflammatory response, and XIST/miR-370-3p/TLR4 axis thus may shed light on the development of novel therapeutics to the treatment of acute stage of pneumonia. SIGNIFICANCE OF THE STUDY: Our study demonstrated that XIST was highly expressed in patients with acute stage of pneumonia. Knockdown of XIST remarkably alleviated LPS-induced cell injury through increasing cell viability and inhibiting apoptosis and inflammatory cytokine levels through regulating JAK/STAT and NF-κB pathways.
Collapse
Affiliation(s)
- Yena Zhang
- Department of Pulmonary Medicine, HwaMei HospitalUniversity Of Chinese Academy Of SciencesNingboChina
| | - Yuyin Zhu
- Department of Pulmonary Medicine, HwaMei HospitalUniversity Of Chinese Academy Of SciencesNingboChina
| | - Guosheng Gao
- Department of Laboratory, HwaMei HospitalUniversity Of Chinese Academy Of SciencesNingboChina
| | - Zhiming Zhou
- Department of Pulmonary Medicine, HwaMei HospitalUniversity Of Chinese Academy Of SciencesNingboChina
| |
Collapse
|
8
|
Tabish AM, Arif M, Song T, Elbeck Z, Becker RC, Knöll R, Sadayappan S. Association of intronic DNA methylation and hydroxymethylation alterations in the epigenetic etiology of dilated cardiomyopathy. Am J Physiol Heart Circ Physiol 2019; 317:H168-H180. [PMID: 31026178 PMCID: PMC6692731 DOI: 10.1152/ajpheart.00758.2018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 04/15/2019] [Accepted: 04/22/2019] [Indexed: 01/03/2023]
Abstract
In this study, we investigated the role of DNA methylation [5-methylcytosine (5mC)] and 5-hydroxymethylcytosine (5hmC), epigenetic modifications that regulate gene activity, in dilated cardiomyopathy (DCM). A MYBPC3 mutant mouse model of DCM was compared with wild type and used to profile genomic 5mC and 5hmC changes by Chip-seq, and gene expression levels were analyzed by RNA-seq. Both 5mC-altered genes (957) and 5hmC-altered genes (2,022) were identified in DCM hearts. Diverse gene ontology and KEGG pathways were enriched for DCM phenotypes, such as inflammation, tissue fibrosis, cell death, cardiac remodeling, cardiomyocyte growth, and differentiation, as well as sarcomere structure. Hierarchical clustering of mapped genes affected by 5mC and 5hmC clearly differentiated DCM from wild-type phenotype. Based on these data, we propose that genomewide 5mC and 5hmC contents may play a major role in DCM pathogenesis. NEW & NOTEWORTHY Our data demonstrate that development of dilated cardiomyopathy in mice is associated with significant epigenetic changes, specifically in intronic regions, which, when combined with gene expression profiling data, highlight key signaling pathways involved in pathological cardiac remodeling and heart contractile dysfunction.
Collapse
Affiliation(s)
- Ali M Tabish
- Integrated Cardio-Metabolic Centre, Karolinska Institutet , Stockholm , Sweden
| | - Mohammed Arif
- Heart, Lung, Vascular Institute, Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati , Cincinnati, Ohio
| | - Taejeong Song
- Heart, Lung, Vascular Institute, Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati , Cincinnati, Ohio
| | - Zaher Elbeck
- Integrated Cardio-Metabolic Centre, Karolinska Institutet , Stockholm , Sweden
| | - Richard C Becker
- Heart, Lung, Vascular Institute, Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati , Cincinnati, Ohio
| | - Ralph Knöll
- Integrated Cardio-Metabolic Centre, Karolinska Institutet , Stockholm , Sweden
- Cardiovascular and Metabolic Disease Innovative Medicines and Early Development Unit, AstraZeneca R&D, Gothenburg , Sweden
| | - Sakthivel Sadayappan
- Heart, Lung, Vascular Institute, Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati , Cincinnati, Ohio
| |
Collapse
|
9
|
Zhao YB, Zhao J, Zhang LJ, Shan RG, Sun ZZ, Wang K, Chen JQ, Mu JX. MicroRNA-370 protects against myocardial ischemia/reperfusion injury in mice following sevoflurane anesthetic preconditioning through PLIN5-dependent PPAR signaling pathway. Biomed Pharmacother 2019; 113:108697. [PMID: 30856533 DOI: 10.1016/j.biopha.2019.108697] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 02/13/2019] [Accepted: 02/14/2019] [Indexed: 12/16/2022] Open
Abstract
Myocardial ischemia/reperfusion injury (IRI) has long been identified to be a contributor to adverse cardiovascular outcomes following myocardial ischemia, cardiac surgery or circulatory arrest. This study aims to investigate the effects of microRNA (miR-370) targeting perilipin-5 (PLIN5) in mice following sevoflurane anesthetic preconditioning (SAP). A mouse model of left ventricular myocardial IRI was established, followed by the evaluation of myocardial infarction size and cardiac function to determine the effects of SAP. The underlying regulatory mechanisms of miR-370 were analyzed in concert with the treatment of miR-370 mimic, miR-370 inhibitor, or siRNA against PLIN5 in cardiomyocytes isolated from mice with IRI. Also, cardiomyocyte proliferation, cell cycle distribution and apoptosis were evaluated following treatment. Lastly, SAP-treated I/R mice were injected with miR-370 inhibitor to verify the mechanism of SAP. The use of SAP conferred cardioprotective effects on myocardial IRI. MiR-370 was downregulated in mice that exhibited IRI, but SAP elevated the miR-370 expression. Functionally, miR-370 negatively targeted PLIN5 and activated the peroxisome proliferator activated-receptor (PPAR) signaling pathway, leading to decreased PPARγ expression but increased PPARα expression. The results also showed that elevation of miR-370 or the silencing of PLIN5 promoted cardiomyocyte proliferation. miR-370 also inhibited cardiomyocyte apoptosis as reflected by decreased caspase-3 expression and increased Bcl-2 expression. Additionally, SAP also alleviated I/R injury by inhibiting PPARγ. This study demonstrates that SAP induces miR-370 and exerts cardioprotective effects on myocardial IRI, where upregulation of miR-370 alleviates myocardial IRI via inhibiting the PLIN5-dependent PPAR signaling pathway.
Collapse
Affiliation(s)
- Yan-Bin Zhao
- Department of Anesthesiology, the First Hospital of the PLA, Lanzhou 730031, PR China
| | - Juan Zhao
- Department of Anesthesiology, the First Hospital of Lanzhou University, Donggang Courtyard Area, Lanzhou 730030, PR China
| | - Li-Jun Zhang
- Department of Anesthesiology, Lanzhou Time Laser Cosmetic Surgery Hospital, Lanzhou 730030, PR China
| | - Run-Gang Shan
- Department of Anesthesiology, the First Hospital of the PLA, Lanzhou 730031, PR China
| | - Zhen-Zhong Sun
- Department of Anesthesiology, Guangdong Armed Police Corps Hospital, Guangzhou 510507, PR China
| | - Kai Wang
- Department of Anesthesiology, the First Hospital of the PLA, Lanzhou 730031, PR China
| | - Jin-Quan Chen
- Department of Anesthesiology, the First People's Hospital of Xianyang City, Xianyang 712000, PR China.
| | - Ji-Xue Mu
- Lanzhou Minimally Invasive Orthopedic Hospital, Lanzhou 730050, PR China.
| |
Collapse
|
10
|
Emerging Roles for Immune Cells and MicroRNAs in Modulating the Response to Cardiac Injury. J Cardiovasc Dev Dis 2019; 6:jcdd6010005. [PMID: 30650599 PMCID: PMC6462949 DOI: 10.3390/jcdd6010005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 01/04/2019] [Accepted: 01/09/2019] [Indexed: 01/13/2023] Open
Abstract
Stimulating cardiomyocyte regeneration after an acute injury remains the central goal in cardiovascular regenerative biology. While adult mammals respond to cardiac damage with deposition of rigid scar tissue, adult zebrafish and salamander unleash a regenerative program that culminates in new cardiomyocyte formation, resolution of scar tissue, and recovery of heart function. Recent studies have shown that immune cells are key to regulating pro-inflammatory and pro-regenerative signals that shift the injury microenvironment toward regeneration. Defining the genetic regulators that control the dynamic interplay between immune cells and injured cardiac tissue is crucial to decoding the endogenous mechanism of heart regeneration. In this review, we discuss our current understanding of the extent that macrophage and regulatory T cells influence cardiomyocyte proliferation and how microRNAs (miRNAs) regulate their activity in the injured heart.
Collapse
|