1
|
Han B, Zhou L, Shi Y, Zhao F, Ji J, Zhang K, Yin S, Ning X. LncRNA432-miR-21-y-DAPK2 ceRNA crosstalk regulates antibacterial response in hypoxia stress through mediating mitochondrial apoptosis in teleost fish. Int J Biol Macromol 2025; 295:139694. [PMID: 39798738 DOI: 10.1016/j.ijbiomac.2025.139694] [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: 10/16/2024] [Revised: 12/21/2024] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
As cold-blooded vertebrates, fish are sensitive to environmental changes. The outcome of pathogen infections in fish therefore is highly shaped by hypoxia. The epigenetic regulation of competitive endogenous RNA (ceRNA) bridging non-coding RNAs and mRNAs represents a promising mechanism modulating antibacterial response plus environmental stress. Here, we for the first time systematically analyzed the ceRNA crosstalk in fish response to the combined stimulation of hypoxia and bacterial infection (HB) dual-stimulation. We found that mitochondrial apoptosis initiated by loss of mitochondrial membrane potential was the main causative for liver damage induced by HB challenge in fish. Accordingly, through whole transcriptome analysis, an apoptosis-associated ceRNA network was constructed, based on which a key crosstalk consisting of lnc432, miR-21-y and DAPK2 was identified. Mechanistically, DAPK2 acted as a positive regulator, knockdown of which significantly increased the bacterial burden during hypoxia by promoting mitochondrial apoptosis. MiR-21-y inhibited DAPK2 expression at both mRNA and protein levels by interacting with its 3'UTR, thereby enhancing DAPK2-mediated apoptosis determinations, and exacerbating bacterial infection during hypoxia. Lnc432 knockdown significantly increased miR-21-y and decreased DAPK2, and substantially promoted the expression of genes associated with mitochondrial apoptosis and enhanced the bacterial load during hypoxia stress. Finally, we revealed that lnc432 sponged miR-21-y to alleviate its suppression on DAPK2 in the ceRNA regulatory way. Our findings reveal that lnc432-miR-21-y-DAPK2 ceRNA crosstalk occurs in fish response to bacterial infection during hypoxic stress through mediating mitochondrial apoptosis. This study provides novel insights into the mechanism underlying the interactions among pathogens, hosts and environmental factors.
Collapse
Affiliation(s)
- Bing Han
- College of Marine Science and Engineering, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Jiangsu Key Laboratory of Ocean-Land Environmental Change and Ecological Construction, Nanjing Normal University, Nanjing 210023, Jiangsu, China
| | - Linxin Zhou
- College of Marine Science and Engineering, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Jiangsu Key Laboratory of Ocean-Land Environmental Change and Ecological Construction, Nanjing Normal University, Nanjing 210023, Jiangsu, China
| | - Yaxuan Shi
- College of Marine Science and Engineering, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Jiangsu Key Laboratory of Ocean-Land Environmental Change and Ecological Construction, Nanjing Normal University, Nanjing 210023, Jiangsu, China
| | - Feng Zhao
- College of Marine Science and Engineering, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Jiangsu Key Laboratory of Ocean-Land Environmental Change and Ecological Construction, Nanjing Normal University, Nanjing 210023, Jiangsu, China
| | - Jie Ji
- College of Marine Science and Engineering, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Jiangsu Key Laboratory of Ocean-Land Environmental Change and Ecological Construction, Nanjing Normal University, Nanjing 210023, Jiangsu, China; Co-Innovation Center for Marine Bio-Industry Technology of Jiangsu Province, Lianyungang 222005, China
| | - Kai Zhang
- College of Marine Science and Engineering, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Jiangsu Key Laboratory of Ocean-Land Environmental Change and Ecological Construction, Nanjing Normal University, Nanjing 210023, Jiangsu, China; Co-Innovation Center for Marine Bio-Industry Technology of Jiangsu Province, Lianyungang 222005, China
| | - Shaowu Yin
- College of Marine Science and Engineering, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Jiangsu Key Laboratory of Ocean-Land Environmental Change and Ecological Construction, Nanjing Normal University, Nanjing 210023, Jiangsu, China; Co-Innovation Center for Marine Bio-Industry Technology of Jiangsu Province, Lianyungang 222005, China.
| | - Xianhui Ning
- College of Marine Science and Engineering, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Jiangsu Key Laboratory of Ocean-Land Environmental Change and Ecological Construction, Nanjing Normal University, Nanjing 210023, Jiangsu, China; Co-Innovation Center for Marine Bio-Industry Technology of Jiangsu Province, Lianyungang 222005, China.
| |
Collapse
|
2
|
Yang H, Li J, Song C, Li H, Luo Q, Chen M. Emerging Gene Therapy Based on Nanocarriers: A Promising Therapeutic Alternative for Cardiovascular Diseases and a Novel Strategy in Valvular Heart Disease. Int J Mol Sci 2025; 26:1743. [PMID: 40004206 PMCID: PMC11855571 DOI: 10.3390/ijms26041743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/10/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Cardiovascular disease remains a leading cause of global mortality, with many unresolved issues in current clinical treatment strategies despite years of extensive research. Due to the great progress in nanotechnology and gene therapy in recent years, the emerging gene therapy based on nanocarriers has provided a promising therapeutic alternative for cardiovascular diseases. This review outlines the status of nanocarriers as vectors in gene therapy for cardiovascular diseases, including coronary heart disease, pulmonary hypertension, hypertension, and valvular heart disease. It discusses challenges and future prospects, aiming to support emerging clinical treatments. This review is the first to summarize gene therapy using nanocarriers for valvular heart disease, highlighting their potential in targeting challenging tissues.
Collapse
Affiliation(s)
- Haoran Yang
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu 610041, China; (H.Y.); (J.L.); (C.S.)
- Department of Cardiology, West China Hospital, Sichuan University, No.37 Guoxue Street, Chengdu 610041, China
| | - Junli Li
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu 610041, China; (H.Y.); (J.L.); (C.S.)
- Cardiac Structure and Function Research Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chengxiang Song
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu 610041, China; (H.Y.); (J.L.); (C.S.)
- Department of Cardiology, West China Hospital, Sichuan University, No.37 Guoxue Street, Chengdu 610041, China
| | - Hongde Li
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu 610041, China; (H.Y.); (J.L.); (C.S.)
- Department of Cardiology, West China Hospital, Sichuan University, No.37 Guoxue Street, Chengdu 610041, China
| | - Qiang Luo
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu 610041, China; (H.Y.); (J.L.); (C.S.)
- Department of Cardiology, West China Hospital, Sichuan University, No.37 Guoxue Street, Chengdu 610041, China
- Cardiac Structure and Function Research Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Mao Chen
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu 610041, China; (H.Y.); (J.L.); (C.S.)
- Department of Cardiology, West China Hospital, Sichuan University, No.37 Guoxue Street, Chengdu 610041, China
- Cardiac Structure and Function Research Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
3
|
Nagy RN, Makkos A, Baranyai T, Giricz Z, Szabó M, Kravcsenko-Kiss B, Bereczki Z, Ágg B, Puskás LG, Faragó N, Schulz R, Gyöngyösi M, Lukovic D, Varga ZV, Görbe A, Ferdinandy P. Cardioprotective microRNAs (protectomiRs) in a pig model of acute myocardial infarction and cardioprotection by ischaemic conditioning: MiR-450a. Br J Pharmacol 2025; 182:396-416. [PMID: 39294819 DOI: 10.1111/bph.17313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 06/05/2024] [Accepted: 07/04/2024] [Indexed: 09/21/2024] Open
Abstract
BACKGROUND AND PURPOSE Cardioprotective miRNAs (protectomiRs) are promising therapeutic tools. Here, we aimed to identify protectomiRs in a translational porcine model of acute myocardial infarction (AMI) and to validate their cardiocytoprotective effect. EXPERIMENTAL APPROACH ProtectomiR candidates were selected after systematic analysis of miRNA expression changes in cardiac tissue samples from a closed-chest AMI model in pigs subjected to sham operation, AMI and ischaemic preconditioning, postconditioning or remote preconditioning, respectively. Cross-species orthologue protectomiR candidates were validated in simulated ischaemia-reperfusion injury (sI/R) model of isolated rat ocardiomyocytes and in human AC16 cells as well. For miR-450a, we performed target prediction and analysed the potential mechanisms of action by GO enrichment and KEGG pathway analysis. KEY RESULTS Out of the 220 detected miRNAs, four were up-regulated and 10 were down-regulated due to all three conditionings versus AMI. MiR-450a and miR-451 mimics at 25 nM were protective in rat cardiomyocytes, and miR-450a showed protection in human cardiomyocytes as well. MiR-450a has 3987 predicted mRNA targets in pigs, 4279 in rats and 8328 in humans. Of these, 607 genes are expressed in all three species. A total of 421 common enriched GO terms were identified in all three species, whereas KEGG pathway analysis revealed 13 common pathways. CONCLUSION AND IMPLICATIONS This is the first demonstration that miR-450a is associated with cardioprotection by ischaemic conditioning in a clinically relevant porcine model and shows cardiocytoprotective effect in human cardiomyocytes, making it a promising drug candidate. The mechanism of action of miR-450a involves multiple cardioprotective pathways. LINKED ARTICLES This article is part of a themed issue Non-coding RNA Therapeutics. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v182.2/issuetoc.
Collapse
Grants
- OTKA ANN 107803 Hungarian Scientific Research Fund
- OTKA K-105555 Hungarian Scientific Research Fund
- 2018-1.3.1-VKE-2018-00024 National Research, Development and Innovation Office
- NVKP-16-1-2016-0017 National Heart Program National Research, Development and Innovation Office
- OTKA-FK 134751 National Research, Development and Innovation Office
- TKP/ITM/NFKIH National Research, Development and Innovation Office
- OTKAK21-139105 National Research, Development and Innovation Office
- RRF-2.3.1-21-2022-00003 European Union
- EU COST Action CardioRNA.eu, Cardioprotection.eu
- 88öu1 Austrian-Hungarian Action Scholarship
- 739593 European Union's Horizon 2020
- 2019-1.1.1-PIACI-KFI-2019-00367 National Research, Development and Innovation Fund
- 2020-1.1.5-GYORSÍTÓSÁV-2021-00011 National Research, Development and Innovation Fund
- ÚNKP-20-5 National Research, Development and Innovation Fund
- ÚNKP-23-4-II-SE-34 National Research, Development and Innovation Fund
- János Bolyai Research Scholarship of Hungarian Academy of Sciences
Collapse
Affiliation(s)
- Regina N Nagy
- Cardiometabolic and HUN-REN-SU System Pharmacology Research Group, Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - András Makkos
- Cardiometabolic and HUN-REN-SU System Pharmacology Research Group, Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Tamás Baranyai
- Cardiometabolic and HUN-REN-SU System Pharmacology Research Group, Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Zoltán Giricz
- Cardiometabolic and HUN-REN-SU System Pharmacology Research Group, Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Márta Szabó
- Cardiometabolic and HUN-REN-SU System Pharmacology Research Group, Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Bernadett Kravcsenko-Kiss
- Cardiometabolic and HUN-REN-SU System Pharmacology Research Group, Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Zoltán Bereczki
- Cardiometabolic and HUN-REN-SU System Pharmacology Research Group, Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Bence Ágg
- Cardiometabolic and HUN-REN-SU System Pharmacology Research Group, Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - László G Puskás
- Laboratory of Functional Genomics, Biological Research Centre, Szeged, Hungary
| | - Nóra Faragó
- Laboratory of Functional Genomics, Biological Research Centre, Szeged, Hungary
| | - Rainer Schulz
- Institute of Physiology, Justus-Liebig University of Giessen, Giessen, Germany
| | - Mariann Gyöngyösi
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Dominika Lukovic
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Zoltán V Varga
- Cardiometabolic and HUN-REN-SU System Pharmacology Research Group, Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Semmelweis University, Budapest, Hungary
| | - Anikó Görbe
- Cardiometabolic and HUN-REN-SU System Pharmacology Research Group, Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Péter Ferdinandy
- Cardiometabolic and HUN-REN-SU System Pharmacology Research Group, Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| |
Collapse
|
4
|
Perera N, De Blasio MJ, Febbraio MA. Harnessing the therapeutic potential of exercise in extracellular vesicle-based therapy in metabolic disease associated cardiovascular complications. Free Radic Biol Med 2025; 226:230-236. [PMID: 39549882 DOI: 10.1016/j.freeradbiomed.2024.11.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 10/10/2024] [Accepted: 11/13/2024] [Indexed: 11/18/2024]
Abstract
Cardiovascular disease (CVD) is a leading cause of mortality, affecting ∼18 million individuals each year. Obesity and type 2 diabetes mellitus in particular, both chronic metabolic disorders, are risk factors for CVD. The salutary effects of physical activity in preventing and ameliorating CVD have long been acknowledged, as it improves glucose and lipid homeostasis, alongside attenuating oxidative damage, increasing mitochondrial function, and ultimately improving cardiac function. Exercise serves as a catalyst for the secretion of extracellular vesicles (EVs), facilitating inter-tissue communication, by which tissues can deliver important signals from one tissue to another. In recent years, an increasing number of studies have focused on the cargo encapsulated within exercise-derived EVs, as well as the orchestration of inter-tissue crosstalk aimed at modulating metabolism and tissue function in CVDs. The precise mechanisms underpinning the cardioprotective properties of exercise-derived EVs, however, remains only partially elucidated. This review explores novel EV based therapeutic options in CVD and, in particular, EVs derived from models of exercise to alter metabolism and enhance cardiovascular outcomes.
Collapse
Affiliation(s)
- Nimna Perera
- Monash Institute of Pharmaceutical Sciences, Parkville, Melbourne, Australia
| | - Miles J De Blasio
- Monash Institute of Pharmaceutical Sciences, Parkville, Melbourne, Australia
| | - Mark A Febbraio
- Monash Institute of Pharmaceutical Sciences, Parkville, Melbourne, Australia.
| |
Collapse
|
5
|
Saberiyan M, Zarei M, Safi A, Movahhed P, Khorasanian R, Adelian S, Mousavi P. The role of DAPK2 as a key regulatory element in various human cancers: a systematic review. Mol Biol Rep 2024; 51:886. [PMID: 39105958 DOI: 10.1007/s11033-024-09761-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 06/26/2024] [Indexed: 08/07/2024]
Abstract
Cancer is considered the uncontrolled growth and spread of cells into neighboring tissues, a process governed at the molecular level by many different factors, including abnormalities in the protein family's death-associated kinase (DAPK). DAPK2 is a member of the DAPK protein family, which plays essential roles in several cellular processes. DAPK2 acts as a tumor suppressor, interacting with several proteins, such as TNF, IFN, etc. during apoptosis and autophagy. Expression of DAPK2 causes changes in the structure of the cell, ultimately leading to cell death by apoptosis. In this essay, studies are obtained from Scopus, PubMed, and the Web of Science. According to these investigations, DAPK2 activates autophagy by interacting with AMPK, mTORC1, and p73. Furthermore, DAPK2 induces apoptosis pathway via interacting with the p73 family and JNK. In general, due to the vital role of DAPK2 in cell physiology and its effect on various factors and signaling pathways, it can be a potent target in the treatment of various cancers, including gastric, ovarian, breast, and other prominent cancers.
Collapse
Affiliation(s)
- Mohammadreza Saberiyan
- Student Research Committee, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Mahboobeh Zarei
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Amir Safi
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Parvaneh Movahhed
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, ShahidBeheshti University of Medical Sciences, Tehran, Iran
| | - Reihane Khorasanian
- Department of Medical Genetics and Molecular Biology, Faculty of Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Samaneh Adelian
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Pegah Mousavi
- Molecular Medicine Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.
| |
Collapse
|
6
|
Olson SR, Tang WHW, Liu CF. Non-Coding Ribonucleic Acids as Diagnostic and Therapeutic Targets in Cardiac Fibrosis. Curr Heart Fail Rep 2024; 21:262-275. [PMID: 38485860 PMCID: PMC11090942 DOI: 10.1007/s11897-024-00653-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/26/2024] [Indexed: 05/14/2024]
Abstract
PURPOSE OF REVIEW Cardiac fibrosis is a crucial juncture following cardiac injury and a precursor for many clinical heart disease manifestations. Epigenetic modulators, particularly non-coding RNAs (ncRNAs), are gaining prominence as diagnostic and therapeutic tools. RECENT FINDINGS miRNAs are short linear RNA molecules involved in post-transcriptional regulation; lncRNAs and circRNAs are RNA sequences greater than 200 nucleotides that also play roles in regulating gene expression through a variety of mechanisms including miRNA sponging, direct interaction with mRNA, providing protein scaffolding, and encoding their own products. NcRNAs have the capacity to regulate one another and form sophisticated regulatory networks. The individual roles and disease relevance of miRNAs, lncRNAs, and circRNAs to cardiac fibrosis have been increasingly well described, though the complexity of their interrelationships, regulatory dynamics, and context-specific roles needs further elucidation. This review provides an overview of select ncRNAs relevant in cardiac fibrosis as a surrogate for many cardiac disease states with a focus on crosstalk and regulatory networks, variable actions among different disease states, and the clinical implications thereof. Further, the clinical feasibility of diagnostic and therapeutic applications as well as the strategies underway to advance ncRNA theranostics is explored.
Collapse
Affiliation(s)
- Samuel R Olson
- Medicine Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - W H Wilson Tang
- Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
- Kaufman Center for Heart Failure Treatment and Recovery, Heart Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Chia-Feng Liu
- Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA.
| |
Collapse
|
7
|
Ji XD, Yang D, Cui XY, Lou LX, Nie B, Zhao JL, Zhao MJ, Wu AM. Mechanism of Qili Qiangxin Capsule for Heart Failure Based on miR133a-Endoplasmic Reticulum Stress. Chin J Integr Med 2024; 30:398-407. [PMID: 38386253 DOI: 10.1007/s11655-024-3654-3] [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] [Accepted: 11/27/2023] [Indexed: 02/23/2024]
Abstract
OBJECTIVE To investigate the pharmacological mechanism of Qili Qiangxin Capsule (QLQX) improvement of heart failure (HF) based on miR133a-endoplasmic reticulum stress (ERS) pathway. METHODS A left coronary artery ligation-induced HF after myocardial infarction model was used in this study. Rats were randomly assigned to the sham group, the model group, the QLQX group [0.32 g/(kg·d)], and the captopril group [2.25 mg/(kg·d)], 15 rats per group, followed by 4 weeks of medication. Cardiac function such as left ventricular ejection fraction (EF), fractional shortening (FS), left ventricular systolic pressure (LVSP), left ventricular end diastolic pressure (LVEDP), the maximal rate of increase of left ventricular pressure (+dp/dt max), and the maximal rate of decrease of left ventricular pressure (-dp/dt max) were monitored by echocardiography and hemodynamics. Hematoxylin and eosin (HE) and Masson stainings were used to visualize pathological changes in myocardial tissue. The mRNA expression of miR133a, glucose-regulated protein78 (GRP78), inositol-requiring enzyme 1 (IRE1), activating transcription factor 6 (ATF6), X-box binding protein1 (XBP1), C/EBP homologous protein (CHOP) and Caspase 12 were detected by RT-PCR. The protein expression of GRP78, p-IRE1/IRE1 ratio, cleaved-ATF6, XBP1-s (the spliced form of XBP1), CHOP and Caspase 12 were detected by Western blot. TdT-mediated dUTP nick-end labeling (TUNEL) staining was used to detect the rate of apoptosis. RESULTS QLQX significantly improved cardiac function as evidenced by increased EF, FS, LVSP, +dp/dt max, -dp/dt max, and decreased LVEDP (P<0.05, P<0.01). HE staining showed that QLQX ameliorated cardiac pathologic damage to some extent. Masson staining indicated that QLQX significantly reduced collagen volume fraction in myocardial tissue (P<0.01). Results from RT-PCR and Western blot showed that QLQX significantly increased the expression of miR133a and inhibited the mRNA expressions of GRP78, IRE1, ATF6 and XBP1, as well as decreased the protein expressions of GRP78, cleaved-ATF6 and XBP1-s and decreased p-IRE1/IRE1 ratio (P<0.05, P<0.01). Further studies showed that QLQX significantly reduced the expression of CHOP and Caspase12, resulting in a significant reduction in apoptosis rate (P<0.05, P<0.01). CONCLUSION The pharmacological mechanism of QLQX in improving HF is partly attributed to its regulatory effect on the miR133a-IRE1/XBP1 pathway.
Collapse
Affiliation(s)
- Xiao-di Ji
- Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Beijing, 100700, China
- Department of Traditional Chinese Medicine, Fuwai Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100037, China
| | - Ding Yang
- Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Beijing, 100700, China
| | - Xi-Yuan Cui
- Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Beijing, 100700, China
| | - Li-Xia Lou
- Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Beijing, 100700, China
| | - Bo Nie
- Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Beijing, 100700, China
| | - Jiu-Li Zhao
- Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Beijing, 100700, China
| | - Ming-Jing Zhao
- Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Beijing, 100700, China
| | - Ai-Ming Wu
- Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Beijing, 100700, China.
| |
Collapse
|
8
|
Weber B, Franz N, Marzi I, Henrich D, Leppik L. Extracellular vesicles as mediators and markers of acute organ injury: current concepts. Eur J Trauma Emerg Surg 2022; 48:1525-1544. [PMID: 33533957 PMCID: PMC7856451 DOI: 10.1007/s00068-021-01607-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 01/19/2021] [Indexed: 12/15/2022]
Abstract
Due to the continued high incidence and mortality rate worldwide, there is a need to develop new strategies for the quick, precise, and valuable recognition of presenting injury pattern in traumatized and poly-traumatized patients. Extracellular vesicles (EVs) have been shown to facilitate intercellular communication processes between cells in close proximity as well as distant cells in healthy and disease organisms. miRNAs and proteins transferred by EVs play biological roles in maintaining normal organ structure and function under physiological conditions. In pathological conditions, EVs change the miRNAs and protein cargo composition, mediating or suppressing the injury consequences. Therefore, incorporating EVs with their unique protein and miRNAs signature into the list of promising new biomarkers is a logical next step. In this review, we discuss the general characteristics and technical aspects of EVs isolation and characterization. We discuss results of recent in vitro, in vivo, and patients study describing the role of EVs in different inflammatory diseases and traumatic organ injuries. miRNAs and protein signature of EVs found in patients with acute organ injury are also debated.
Collapse
Affiliation(s)
- Birte Weber
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Goethe-University, Frankfurt am Main, Germany
| | - Niklas Franz
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Goethe-University, Frankfurt am Main, Germany
| | - Ingo Marzi
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Goethe-University, Frankfurt am Main, Germany
| | - Dirk Henrich
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Goethe-University, Frankfurt am Main, Germany
| | - Liudmila Leppik
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Goethe-University, Frankfurt am Main, Germany.
| |
Collapse
|
9
|
Błażejowska E, Urbanowicz T, Gąsecka A, Olasińska-Wiśniewska A, Jaguszewski MJ, Targoński R, Szarpak Ł, Filipiak KJ, Perek B, Jemielity M. Diagnostic and Prognostic Value of miRNAs after Coronary Artery Bypass Grafting: A Review. BIOLOGY 2021; 10:1350. [PMID: 34943265 PMCID: PMC8698870 DOI: 10.3390/biology10121350] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 12/09/2021] [Accepted: 12/16/2021] [Indexed: 12/23/2022]
Abstract
MiRNAs are noncoding, 21-24 nucleotide-long RNA particles that control over 60% of genes. MiRNAs affect gene expression through binding to the 3'-untranslated region of messenger RNA (mRNA), thus inhibiting mRNA translation or inducing mRNA degradation. MiRNAs have been associated with various cardiovascular diseases, including heart failure, hypertension, left ventricular hypertrophy, or ischemic heart disease. In addition, miRNA expression alters during coronary artery bypass grafting (CABG) surgery, which could be used to predict perioperative outcomes. CABG is an operation in which complex coronary arteries stenosis is treated by bypassing atherosclerotic lesions with venous or arterial grafts. Despite a very low perioperative mortality rate and excellent long-term survival, CABG is associated with postoperative complications, including reperfusion injury, graft failure, atrial fibrillation and perioperative myocardial infarction. So far, no reliable diagnostic and prognostic tools to predict prognosis after CABG have been developed. Changes in the perioperative miRNA expression levels could improve the diagnosis of post-CABG myocardial infarction and atrial fibrillation and could be used to stratify risk after CABG. Herein, we describe the expression changes of different subtypes of miRNAs during CABG and review the diagnostic and prognostic utility of miRNAs in patients undergoing CABG.
Collapse
Affiliation(s)
- Ewelina Błażejowska
- 1st Chair and Department of Cardiology, Medical University of Warsaw, 02-091 Warsaw, Poland;
| | - Tomasz Urbanowicz
- Cardiac Surgery and Transplantology Department, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (T.U.); (A.O.-W.); (B.P.); (M.J.)
| | - Aleksandra Gąsecka
- 1st Chair and Department of Cardiology, Medical University of Warsaw, 02-091 Warsaw, Poland;
| | - Anna Olasińska-Wiśniewska
- Cardiac Surgery and Transplantology Department, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (T.U.); (A.O.-W.); (B.P.); (M.J.)
| | - Miłosz J. Jaguszewski
- 1st Department of Cardiology, Medical University of Gdansk, 80-211 Gdansk, Poland; (M.J.J.); (R.T.)
| | - Radosław Targoński
- 1st Department of Cardiology, Medical University of Gdansk, 80-211 Gdansk, Poland; (M.J.J.); (R.T.)
| | - Łukasz Szarpak
- Department of Clinical Sciences, Maria Sklodowska-Curie Medical Academy, 03-411 Warsaw, Poland; (Ł.S.); (K.J.F.)
| | - Krzysztof J. Filipiak
- Department of Clinical Sciences, Maria Sklodowska-Curie Medical Academy, 03-411 Warsaw, Poland; (Ł.S.); (K.J.F.)
| | - Bartłomiej Perek
- Cardiac Surgery and Transplantology Department, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (T.U.); (A.O.-W.); (B.P.); (M.J.)
| | - Marek Jemielity
- Cardiac Surgery and Transplantology Department, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (T.U.); (A.O.-W.); (B.P.); (M.J.)
| |
Collapse
|
10
|
Wagner KT, Radisic M. A New Role for Extracellular Vesicles in Cardiac Tissue Engineering and Regenerative Medicine. ADVANCED NANOBIOMED RESEARCH 2021; 1:2100047. [PMID: 34927167 PMCID: PMC8680295 DOI: 10.1002/anbr.202100047] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular diseases are the leading cause of death worldwide. Discovering new therapies to treat heart disease requires improved understanding of cardiac physiology at a cellular level. Extracellular vesicles (EVs) are plasma membrane-bound nano- and microparticles secreted by cells and known to play key roles in intercellular communication, often through transfer of biomolecular cargo. Advances in EV research have established techniques for EV isolation from tissue culture media or biofluids, as well as standards for quantitation and biomolecular characterization. EVs released by cardiac cells are known to be involved in regulating cardiac physiology as well as in the progression of myocardial diseases. Due to difficulty accessing the heart in vivo, advanced in vitro cardiac 'tissues-on-a-chip' have become a recent focus for studying EVs in the heart. These physiologically relevant models are producing new insight into the role of EVs in cardiac physiology and disease while providing a useful platform for screening novel EV-based therapeutics for cardiac tissue regeneration post-injury. Numerous hurdles have stalled the clinical translation of EV therapeutics for heart patients, but tissue-on-a-chip models are playing an important role in bridging the translational gap, improving mechanistic understanding of EV signalling in cardiac physiology, disease, and repair.
Collapse
Affiliation(s)
- Karl T Wagner
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 27 King's College Circle, Toronto, Ontario, Canada M5S 1A1
| | - Milica Radisic
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 27 King's College Circle, Toronto, Ontario, Canada M5S 1A1
| |
Collapse
|
11
|
Yarmohammadi F, Hayes AW, Karimi G. The cardioprotective effects of hydrogen sulfide by targeting endoplasmic reticulum stress and the Nrf2 signaling pathway: A review. Biofactors 2021; 47:701-712. [PMID: 34161646 DOI: 10.1002/biof.1763] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/30/2021] [Accepted: 06/01/2021] [Indexed: 12/20/2022]
Abstract
Cardiac diseases are emerging due to lifestyle, urbanization, and the accelerated aging process. Oxidative stress has been associated with cardiac injury progression through interference with antioxidant strategies and endoplasmic reticulum (ER) function. Hydrogen sulfide (H2 S) is generated endogenously from l-cysteine in various tissues including heart tissue. Pharmacological evaluation of H2 S has suggested a potential role for H2 S against diabetic cardiomyopathy, ischemia/reperfusion injury, myocardial infarction, and cardiotoxicity. Nuclear factor E2-related factor 2 (Nrf2) activity is crucial for cell survival in response to oxidative stress. H2 S up-regulates Nrf2 expression and its related signaling pathway in myocytes. H2 S also suppresses the expression and activity of ER stress-related proteins. H2 S has been reported to improve various cardiac conditions through antioxidant and anti-ER stress-related activities.
Collapse
Affiliation(s)
- Fatemeh Yarmohammadi
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - A Wallace Hayes
- Center for Environmental Occupational Risk Analysis and Management, College of Public Health, University of South Florida, Tampa, Florida, USA
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan, USA
| | - Gholamreza Karimi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
12
|
Wang Q, Lin Y, Zhong W, Jiang Y, Lin Y. Regulatory Non-coding RNAs for Death Associated Protein Kinase Family. Front Mol Biosci 2021; 8:649100. [PMID: 34422899 PMCID: PMC8377501 DOI: 10.3389/fmolb.2021.649100] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 07/26/2021] [Indexed: 01/24/2023] Open
Abstract
The death associated protein kinases (DAPKs) are a family of calcium dependent serine/threonine kinases initially identified in the regulation of apoptosis. Previous studies showed that DAPK family members, including DAPK1, DAPK2 and DAPK3 play a crucial regulatory role in malignant tumor development, in terms of cell apoptosis, proliferation, invasion and metastasis. Accumulating evidence has demonstrated that non-coding RNAs, including microRNA (miRNA), long non-coding RNA (lncRNA) and circRNA, are involved in the regulation of gene expression and tumorigenesis. Recent studies indicated that non-coding RNAs participate in the regulation of DAPKs. In this review, we summarized the current knowledge of non-coding RNAs, as well as the potential miRNAs, lncRNAs and circRNAs, that are involved in the regulation of DAPKs.
Collapse
Affiliation(s)
- Qingshui Wang
- Central Laboratory at the Second Affiliated Hospital of Fujian Traditional Chinese Medical University, Collaborative Innovation Center for Rehabilitation Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, College of Life Sciences, Fujian Normal University, Fuzhou, China
| | - Youyu Lin
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, College of Life Sciences, Fujian Normal University, Fuzhou, China
| | - Wenting Zhong
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, College of Life Sciences, Fujian Normal University, Fuzhou, China
| | - Yu Jiang
- Prenatal Diagnosis Centre, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Yao Lin
- Central Laboratory at the Second Affiliated Hospital of Fujian Traditional Chinese Medical University, Collaborative Innovation Center for Rehabilitation Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, College of Life Sciences, Fujian Normal University, Fuzhou, China
| |
Collapse
|
13
|
Bian Y, Pang P, Li X, Yu S, Wang X, Liu K, Ju J, Wu H, Gao Y, Liu Q, Jia Y, Qu Z, Bi X, Mei Z, Yin X, Wang N, Du W, Yang B. CircHelz activates NLRP3 inflammasome to promote myocardial injury by sponging miR-133a-3p in mouse ischemic heart. J Mol Cell Cardiol 2021; 158:128-139. [PMID: 34043986 DOI: 10.1016/j.yjmcc.2021.05.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 05/17/2021] [Accepted: 05/18/2021] [Indexed: 01/14/2023]
Abstract
Myocardial infarction (MI)-induced the activation of NLRP3 inflammasome has been well known to aggravate myocardial injury and cardiac dysfunction by causing inflammation and pyroptosis in the heart. Circular RNAs (circRNAs) have been demonstrated to play critical roles in cardiovascular diseases. However, the functions and mechanisms of circRNAs in modulating cardiac inflammatory response and cardiomyocyte pyroptosis remain largely unknown. We revealed that circHelz, a novel circRNA transcribed from the helicase with zinc finger (Helz) gene, was significantly upregulated in both the ischemic myocardium of MI mouse and neonatal mouse ventricular cardiomyocytes (NMVCs) exposed to hypoxia. Overexpression of circHelz caused cardiomyocyte injury in NMVCs by activating the NLRP3 inflammasome and inducing pyroptosis, while circHelz silencing reduced these effects induced by hypoxia. Furthermore, knockdown of circHelz remarkably attenuated NLRP3 expression, decreased myocardial infarct size, pyroptosis, inflammation, and increased cardiac function in vivo after MI. Overexpression of miR-133a-3p in cardiomyocytes greatly prevented pyroptosis in the presence of hypoxia or circHelz by targeting NLRP3 in NMVCs. Mechanistically, circHelz functioned as an endogenous sponge for miR-133a-3p via suppressing its activity. Overall, our results demonstrate that circHelz causes myocardial injury by triggering the NLRP3 inflammasome-mediated pro-inflammatory response and subsequent pyroptosis in cardiomyocytes by inhibiting miR-133a-3p function. Therefore, interfering with circHelz/miR-133a-3p/NLRP3 axis might be a promising therapeutic approach for ischemic cardiac diseases.
Collapse
Affiliation(s)
- Yu Bian
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, Heilongjiang 150081, PR China; Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, PR China
| | - Ping Pang
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Xin Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Shuting Yu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Xiuzhu Wang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Kuiwu Liu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Jiaming Ju
- Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, PR China
| | - Han Wu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Yuelin Gao
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Qian Liu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Yingqiong Jia
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Zhezhe Qu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Xiaoqian Bi
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Zhongting Mei
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Xinda Yin
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Ning Wang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, Heilongjiang 150081, PR China; Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, PR China.
| | - Weijie Du
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, Heilongjiang 150081, PR China; Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, PR China.
| | - Baofeng Yang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, Heilongjiang 150081, PR China; Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, PR China; Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, China.
| |
Collapse
|
14
|
A Brief Review on the Biology and Effects of Cellular and Circulating microRNAs on Cardiac Remodeling after Infarction. Int J Mol Sci 2021; 22:ijms22094995. [PMID: 34066757 PMCID: PMC8125864 DOI: 10.3390/ijms22094995] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/03/2021] [Accepted: 05/05/2021] [Indexed: 01/21/2023] Open
Abstract
Despite advances in diagnostic, prognostic, and treatment modalities, myocardial infarction (MI) remains a leading cause of morbidity and mortality. Impaired cellular signaling after an MI causes maladaptive changes resulting in cardiac remodeling. MicroRNAs (miRNAs/miR) along with other molecular components have been investigated for their involvement in cellular signaling in the pathogenesis of various cardiac conditions like MI. miRNAs are small non-coding RNAs that negatively regulate gene expression. They bind to complementary mRNAs and regulate the rate of protein synthesis by altering the stability of their targeted mRNAs. A single miRNA can modulate several cellular signaling pathways by targeting hundreds of mRNAs. This review focuses on the biogenesis and beneficial effects of cellular and circulating (exosomal) miRNAs on cardiac remodeling after an MI. Particularly, miR-1, -133, 135, and -29 that play an essential role in cardiac remodeling after an MI are described in detail. The limitations that will need to be addressed in the future for the further development of miRNA-based therapeutics for cardiovascular conditions will also be discussed.
Collapse
|
15
|
Yang M, Wang L. MALAT1 knockdown protects from bronchial/tracheal smooth muscle cell injury via regulation of microRNA-133a/ryanodine receptor 2 axis. J Biosci 2021. [DOI: 10.1007/s12038-021-00149-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
16
|
Wang YZ, Ngowi EE, Wang D, Qi HW, Jing MR, Zhang YX, Cai CB, He QL, Khattak S, Khan NH, Jiang QY, Ji XY, Wu DD. The Potential of Hydrogen Sulfide Donors in Treating Cardiovascular Diseases. Int J Mol Sci 2021; 22:2194. [PMID: 33672103 PMCID: PMC7927090 DOI: 10.3390/ijms22042194] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/05/2021] [Accepted: 02/10/2021] [Indexed: 02/08/2023] Open
Abstract
Hydrogen sulfide (H2S) has long been considered as a toxic gas, but as research progressed, the idea has been updated and it has now been shown to have potent protective effects at reasonable concentrations. H2S is an endogenous gas signaling molecule in mammals and is produced by specific enzymes in different cell types. An increasing number of studies indicate that H2S plays an important role in cardiovascular homeostasis, and in most cases, H2S has been reported to be downregulated in cardiovascular diseases (CVDs). Similarly, in preclinical studies, H2S has been shown to prevent CVDs and improve heart function after heart failure. Recently, many H2S donors have been synthesized and tested in cellular and animal models. Moreover, numerous molecular mechanisms have been proposed to demonstrate the effects of these donors. In this review, we will provide an update on the role of H2S in cardiovascular activities and its involvement in pathological states, with a special focus on the roles of exogenous H2S in cardiac protection.
Collapse
Affiliation(s)
- Yi-Zhen Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.-Z.W.); (E.E.N.); (D.W.); (H.-W.Q.); (M.-R.J.); (Y.-X.Z.); (C.-B.C.); (Q.-L.H.); (S.K.); (N.H.K.)
| | - Ebenezeri Erasto Ngowi
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.-Z.W.); (E.E.N.); (D.W.); (H.-W.Q.); (M.-R.J.); (Y.-X.Z.); (C.-B.C.); (Q.-L.H.); (S.K.); (N.H.K.)
- Department of Biological Sciences, Faculty of Science, Dar es Salaam University College of Education, Dar es Salaam 2329, Tanzania
| | - Di Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.-Z.W.); (E.E.N.); (D.W.); (H.-W.Q.); (M.-R.J.); (Y.-X.Z.); (C.-B.C.); (Q.-L.H.); (S.K.); (N.H.K.)
| | - Hui-Wen Qi
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.-Z.W.); (E.E.N.); (D.W.); (H.-W.Q.); (M.-R.J.); (Y.-X.Z.); (C.-B.C.); (Q.-L.H.); (S.K.); (N.H.K.)
| | - Mi-Rong Jing
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.-Z.W.); (E.E.N.); (D.W.); (H.-W.Q.); (M.-R.J.); (Y.-X.Z.); (C.-B.C.); (Q.-L.H.); (S.K.); (N.H.K.)
| | - Yan-Xia Zhang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.-Z.W.); (E.E.N.); (D.W.); (H.-W.Q.); (M.-R.J.); (Y.-X.Z.); (C.-B.C.); (Q.-L.H.); (S.K.); (N.H.K.)
| | - Chun-Bo Cai
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.-Z.W.); (E.E.N.); (D.W.); (H.-W.Q.); (M.-R.J.); (Y.-X.Z.); (C.-B.C.); (Q.-L.H.); (S.K.); (N.H.K.)
| | - Qing-Lin He
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.-Z.W.); (E.E.N.); (D.W.); (H.-W.Q.); (M.-R.J.); (Y.-X.Z.); (C.-B.C.); (Q.-L.H.); (S.K.); (N.H.K.)
- School of Nursing and Health, Henan University, Kaifeng 475004, China
| | - Saadullah Khattak
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.-Z.W.); (E.E.N.); (D.W.); (H.-W.Q.); (M.-R.J.); (Y.-X.Z.); (C.-B.C.); (Q.-L.H.); (S.K.); (N.H.K.)
- Kaifeng Municipal Key Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular Medicine, Henan University, Kaifeng 475004, China
- School of Life Sciences, Henan University, Kaifeng 475004, China
| | - Nazeer Hussain Khan
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.-Z.W.); (E.E.N.); (D.W.); (H.-W.Q.); (M.-R.J.); (Y.-X.Z.); (C.-B.C.); (Q.-L.H.); (S.K.); (N.H.K.)
- Kaifeng Municipal Key Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular Medicine, Henan University, Kaifeng 475004, China
- School of Life Sciences, Henan University, Kaifeng 475004, China
| | - Qi-Ying Jiang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.-Z.W.); (E.E.N.); (D.W.); (H.-W.Q.); (M.-R.J.); (Y.-X.Z.); (C.-B.C.); (Q.-L.H.); (S.K.); (N.H.K.)
| | - Xin-Ying Ji
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.-Z.W.); (E.E.N.); (D.W.); (H.-W.Q.); (M.-R.J.); (Y.-X.Z.); (C.-B.C.); (Q.-L.H.); (S.K.); (N.H.K.)
- Kaifeng Key Laboratory of Infection and Biological Safety, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Dong-Dong Wu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.-Z.W.); (E.E.N.); (D.W.); (H.-W.Q.); (M.-R.J.); (Y.-X.Z.); (C.-B.C.); (Q.-L.H.); (S.K.); (N.H.K.)
- School of Stomatology, Henan University, Kaifeng 475004, China
| |
Collapse
|
17
|
Liu Y, Wang M, Liang Y, Wang C, Naruse K, Takahashi K. Treatment of Oxidative Stress with Exosomes in Myocardial Ischemia. Int J Mol Sci 2021; 22:ijms22041729. [PMID: 33572188 PMCID: PMC7915208 DOI: 10.3390/ijms22041729] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/02/2021] [Accepted: 02/04/2021] [Indexed: 02/06/2023] Open
Abstract
A thrombus in a coronary artery causes ischemia, which eventually leads to myocardial infarction (MI) if not removed. However, removal generates reactive oxygen species (ROS), which causes ischemia–reperfusion (I/R) injury that damages the tissue and exacerbates the resulting MI. The mechanism of I/R injury is currently extensively understood. However, supplementation of exogenous antioxidants is ineffective against oxidative stress (OS). Enhancing the ability of endogenous antioxidants may be a more effective way to treat OS, and exosomes may play a role as targeted carriers. Exosomes are nanosized vesicles wrapped in biofilms which contain various complex RNAs and proteins. They are important intermediate carriers of intercellular communication and material exchange. In recent years, diagnosis and treatment with exosomes in cardiovascular diseases have gained considerable attention. Herein, we review the new findings of exosomes in the regulation of OS in coronary heart disease, discuss the possibility of exosomes as carriers for the targeted regulation of endogenous ROS generation, and compare the advantages of exosome therapy with those of stem-cell therapy. Finally, we explore several miRNAs found in exosomes against OS.
Collapse
|
18
|
Guo H, Wang Y, Jia W, Liu L. MiR-133a-3p relieves the oxidative stress induced trophoblast cell apoptosis through the BACH1/Nrf2/HO-1 signaling pathway. Physiol Res 2021; 70:67-78. [PMID: 33453713 DOI: 10.33549/physiolres.934506] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Preeclampsia (PE) is a major cause of the pregnancy morbidity and mortality over the world. Disorganized placentation caused by trophoblast cell abnormity is one of main risk factors to induce PE. MiR-133a-3p has been shown to contain regulatory effects on oxidative stress in the cardiomyocytes. But the effects of miR-133a-3p on oxidative stress-induced apoptosis in the trophoblast cells remain unknown. In this study, trophoblast HTR-8/SVneo cells were transfected with miR-133a-3p mimics and inhibitor. H2O2 (250 microM) treatment of cells was adopted to induce oxidative stress. A series of typical molecular and cellular experiments was subsequently performed in order to investigate this issue. It was found that miR-133a-3p overexpression attenuated the oxidative stress induced by H2O2 through reduced ROS and MDA levels and enhanced antioxidase activities in the trophoblast cells. Overexpressed miR-133a-3p was shown to relieve the oxidative stress-induced apoptosis of HTR-8/SVneo cells. At molecular levels, a direct binding effect of miR-133a-3p on BACH1 was verified. Moreover, miR-133a-3p overexpression also enhanced BACH1 downstream Nrf2/HO-1 signaling to activate antioxidant genes. It is collectively demonstrated that miR-133a-3p can relieve the oxidative stress-induced apoptosis in the trophoblast cells through the BACH1/Nrf2/HO-1 signaling pathway via targeting BACH1 directly. This regulatory mechanism of miR-133a-3p in the trophoblast cells under oxidative stress may give a new perspective for oxidative stress-induced trophoblast cell abnormality and be useful to study more pathological mechanisms of PE.
Collapse
Affiliation(s)
- H Guo
- Department of Obstetrics, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China.
| | | | | | | |
Collapse
|
19
|
miR-133a-3p attenuates cardiomyocyte hypertrophy through inhibiting pyroptosis activation by targeting IKKε. Acta Histochem 2021; 123:151653. [PMID: 33246224 DOI: 10.1016/j.acthis.2020.151653] [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: 04/08/2020] [Revised: 10/27/2020] [Accepted: 10/28/2020] [Indexed: 01/09/2023]
Abstract
OBJECTIVE Cardiac hypertrophy is an adaptive response to physiological and pathological stimuli, the latter of which frequently progresses to valvulopathy, heart failure and sudden death. Recent reports revealed that pyroptosis is involved in regulating multiple cardiovascular diseases progression, including cardiac hypertrophy. However, the underlying mechanisms remain poorly understood. This study aims to extensively investigate the regulation of miR-133a-3p on pyroptosis in angiotensin II (Ang II)-induced cardiac hypertrophyin vitro. METHODS The in vitro model of cardiac hypertrophy was induced by Ang II, which was validated by qPCR combined with measurement of cell surface area by immunofluorescence assay. CCK-8 assay and Hochest33342/PI staining was performed to assess pyroptosis. Dual luciferase reporter system was used to verify the direct interaction between miR-133a-3p and IKKε. The effects of miR-133a-3p/IKKε on pyroptosis activation and cardiac hypertrophy markers (Caspase-1, NLRP3, IL-1β, IL-18, GSDMD, ASC, ANP, BNP and β-MHC) were evaluated by western blot, ELISA and qPCR. RESULTS Ang II treatment could induce cardiomyocyte hypertrophy and pyroptosis. The expression of miR-133a-3p was repressed in Ang II-treated HCM cells, and its overexpression could attenuate both pyroptosis and cardiac hypertrophyin vitro. Additionally, IKKε expression was significantly up-regulated in Ang II-induced HCM cells. Dual luciferase reporter system and qPCR validated that miR-133a-3p directly targeted the 3'-UTR of IKKε and suppressed its expression. Moreover, IKKε overexpression impaired the protective function of miR-133a-3p in cardiomyocyte hypertrophy. CONCLUSION Collectively, miR-133a-3p attenuates Ang II induced cardiomyocyte hypertrophy via inhibition of pyroptosis by targeting IKKε. Therefore, miR-133a-3p up-regulation may be a promising strategy for cardiac hypertrophy treatment.
Collapse
|
20
|
Henning RJ. Cardiovascular Exosomes and MicroRNAs in Cardiovascular Physiology and Pathophysiology. J Cardiovasc Transl Res 2020; 14:195-212. [PMID: 32588374 DOI: 10.1007/s12265-020-10040-5] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 05/26/2020] [Indexed: 12/16/2022]
Abstract
Cardiac exosomes mediate cell-to-cell communication, stimulate or inhibit the activities of target cells, and affect myocardial hypertrophy, injury and infarction, ventricular remodeling, angiogenesis, and atherosclerosis. The exosomes that are released in the heart from cardiomyocytes, vascular cells, fibroblasts, and resident stem cells are hypoimmunogenic, are physiologically more stable than cardiac cells, can circulate in the body, and are able to cross the blood-brain barrier. Exosomes utilize three mechanisms for cellular communication: (1) internalization by cells, (2) direct fusion to the cell membrane, and (3) receptor-ligand interactions. Cardiac exosomes transmit proteins, mRNA, and microRNAs to other cells during both physiological and pathological process. Cardiac-specific exosome miRNAs can regulate the expression of sarcomeric genes, ion channel genes, autophagy, anti-apoptotic and anti-fibrotic activity, and angiogenesis. This review discusses the role of exosomes and microRNAs in normal myocardium, myocardial injury and infarction, atherosclerosis, and the importance of circulating microRNAs as biomarkers of cardiac disease. Graphical Abstract.
Collapse
Affiliation(s)
- Robert J Henning
- University of South Florida, 13201 Bruce B. Downs Blvd., Tampa, FL, 33612-3805, USA.
| |
Collapse
|
21
|
RGD-PEG-PLA Delivers MiR-133 to Infarct Lesions of Acute Myocardial Infarction Model Rats for Cardiac Protection. Pharmaceutics 2020; 12:pharmaceutics12060575. [PMID: 32575874 PMCID: PMC7356814 DOI: 10.3390/pharmaceutics12060575] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/15/2020] [Accepted: 06/16/2020] [Indexed: 12/16/2022] Open
Abstract
Studies have shown that microRNA-133 (miR-133) plays a positive role in the growth of cardiac myocytes, the maintenance of cardiac homeostasis, and the recovery of cardiac function, which is of great significance for the recovery of acute myocardial infarction. However, the delivery of miRNA to the site of action remains a challenge at present. The purpose of this study was to design an ideal carrier to facilitate the delivery of miR-133 to the infarct lesion for cardiac protection. A disease model was constructed by ligating the left anterior descending coronary artery of rats, and polyethylene glycol (PEG)-polylactic acid (PLA) nanoparticles modified with arginine-glycine-aspartic acid tripeptide (RGD) carrying miR-133 were injected via the tail vein. The effects of miR-133 were evaluated from multiple perspectives, including cardiac function, blood indexes, histopathology, and myocardial cell apoptosis. The results showed that RGD-PEG-PLA maintained a high level of distribution in the hearts of model rats, indicating the role of the carrier in targeting the heart infarction lesions. RGD-PEG-PLA/miR-133 alleviated cardiac histopathological changes, reduced the apoptosis of cardiomyocytes, and reduced the levels of factors associated with myocardial injury. Studies on the mechanism of miR-133 by immunohistochemistry and polymerase chain reaction demonstrated that the expression level of Sirtuin3 (SIRT3) was increased and that the expression of adenosine monophosphate activated protein kinase (AMPK) decreased in myocardial tissue. In summary, the delivery of miR-133 by RGD-PEG-PLA carrier can achieve cardiac lesion accumulation, thereby improving the cardiac function damage and reducing the myocardial infarction area. The inhibition of cardiomyocyte apoptosis, inflammation, and oxidative stress plays a protective role in the heart. The mechanism may be related to the regulation of the SIRT3/AMPK pathway.
Collapse
|
22
|
Kmecova Z, Veteskova J, Lelkova-Zirova K, Bies Pivackova L, Doka G, Malikova E, Paulis L, Krenek P, Klimas J. Disease severity-related alterations of cardiac microRNAs in experimental pulmonary hypertension. J Cell Mol Med 2020; 24:6943-6951. [PMID: 32395887 PMCID: PMC7299706 DOI: 10.1111/jcmm.15352] [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: 02/10/2020] [Revised: 03/24/2020] [Accepted: 04/01/2020] [Indexed: 12/16/2022] Open
Abstract
Right ventricular (RV) failure is the primary cause of death in pulmonary arterial hypertension (PAH). We hypothesized that heart‐relevant microRNAs, that is myomiRs (miR‐1, miR‐133a, miR‐208, miR‐499) and miR‐214, can have a role in the right ventricle in the development of PAH. To mimic PAH, male Wistar rats were injected with monocrotaline (MCT, 60 mg/kg, s.c.); control group received vehicle. MCT rats were divided into two groups, based on the clinical presentation: MCT group terminated 4 weeks after MCT administration and prematurely terminated group (ptMCT) displaying signs of terminal disease. Myocardial damage genes and candidate microRNAs expressions were determined by RT‐qPCR. Reduced blood oxygen saturation, breathing disturbances, RV enlargement as well as elevated levels of markers of myocardial damage confirmed PH in MCT animals and were more pronounced in ptMCT. MyomiRs (miR‐1/miR‐133a/miR‐208a/miR‐499) were decreased and the expression of miR‐214 was increased only in ptMCT group (P < 0.05). The myomiRs negatively correlated with Fulton index as a measure of RV hypertrophy in MCT group (P < 0.05), whereas miR‐214 showed a positive correlation (P < 0.05). We conclude that the expression of determined microRNAs mirrored the disease severity and targeting their pathways might represent potential future therapeutic approach in PAH.
Collapse
Affiliation(s)
- Zuzana Kmecova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, Bratislava, Slovakia
| | - Jana Veteskova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, Bratislava, Slovakia
| | - Katarina Lelkova-Zirova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, Bratislava, Slovakia
| | - Lenka Bies Pivackova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, Bratislava, Slovakia
| | - Gabriel Doka
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, Bratislava, Slovakia
| | - Eva Malikova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, Bratislava, Slovakia
| | - Ludovit Paulis
- Institute of Pathophysiology, Faculty of Medicine, Comenius University, Bratislava, Slovakia.,Institute of Normal and Pathological Physiology, Centre of Experimental Medicine, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Peter Krenek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, Bratislava, Slovakia
| | - Jan Klimas
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, Bratislava, Slovakia
| |
Collapse
|
23
|
Guo Y, Chen J, Qiu H. Novel Mechanisms of Exercise-Induced Cardioprotective Factors in Myocardial Infarction. Front Physiol 2020; 11:199. [PMID: 32210839 PMCID: PMC7076164 DOI: 10.3389/fphys.2020.00199] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 02/21/2020] [Indexed: 12/12/2022] Open
Abstract
Exercise training has been reported to ameliorate heart dysfunction in both humans and animals after myocardial infarction (MI). Exercise-induced cardioprotective factors have been implicated in mediating cardiac repair under pathological conditions. These protective factors secreted by or enriched in the heart could exert cardioprotective functions in an autocrine or paracrine manner. Extracellular vesicles, especially exosomes, contain key molecules and play an essential role in cell-to-cell communication via delivery of various factors, which may be a novel target to study the mechanism of exercise-induced benefits, besides traditional signaling pathways. This review is designed to demonstrate the function and underlying protective mechanism of exercise-induced cardioprotective factors in MI, with an aim to offer more potential therapeutic targets for MI.
Collapse
Affiliation(s)
- Yuan Guo
- Department of Cardiovascular Medicine, The Affiliated Zhuzhou Hospital Xiangya Medical College, Central South University, Zhuzhou, China
| | - Jingyuan Chen
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Haihua Qiu
- Department of Cardiovascular Medicine, The Affiliated Zhuzhou Hospital Xiangya Medical College, Central South University, Zhuzhou, China
| |
Collapse
|
24
|
Yang H, He X, Wang C, Zhang L, Yu J, Wang K. Knockdown of TUG 1 suppresses hypoxia-induced apoptosis of cardiomyocytes by up-regulating miR-133a. Arch Biochem Biophys 2020; 681:108262. [DOI: 10.1016/j.abb.2020.108262] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 01/01/2020] [Accepted: 01/07/2020] [Indexed: 01/23/2023]
|
25
|
Hawkins LJ, Storey KB. Advances and applications of environmental stress adaptation research. Comp Biochem Physiol A Mol Integr Physiol 2019; 240:110623. [PMID: 31778815 DOI: 10.1016/j.cbpa.2019.110623] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 11/19/2019] [Accepted: 11/21/2019] [Indexed: 02/06/2023]
Abstract
Evolution has produced animals that survive extreme fluctuations in environmental conditions including freezing temperatures, anoxia, desiccating conditions, and prolonged periods without food. For example, the wood frog survives whole-body freezing every winter, arresting all gross physiological functions, but recovers functions upon thawing in the spring. Likewise, many small mammals hibernate for months at a time with minimal metabolic activity, organ perfusion, and movement, yet do not suffer significant muscle atrophy upon arousal. These conditions and the biochemical adaptations employed to deal with them can be viewed as Nature's answer to problems that humans wish to answer, particularly in a biomedical context. This review focuses on recent advances in the field of animal environmental stress adaptation, starting with an emphasis on new areas of research such as epigenetics and microRNA. We then examine new and emerging technologies such as genome editing, novel sequencing applications, and single cell analysis and how these can push us closer to a deeper understanding of biochemical adaptation. Next, evaluate the potential contributions of new high-throughput technologies (e.g. next-generation sequencing, mass spectrometry proteomics) to better understanding the adaptations that support these extreme phenotypes. Concluding, we examine some of the human applications that can be gained from understanding the principles of biochemical adaptation including organ preservation and treatments for conditions such as ischemic stroke and muscle disuse atrophy.
Collapse
Affiliation(s)
- Liam J Hawkins
- Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada
| | - Kenneth B Storey
- Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada.
| |
Collapse
|
26
|
Li Z, Zhang Y, Ding N, Zhao Y, Ye Z, Shen L, Yi H, Zhu Y. Inhibition of lncRNA XIST Improves Myocardial I/R Injury by Targeting miR-133a through Inhibition of Autophagy and Regulation of SOCS2. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 18:764-773. [PMID: 31734557 PMCID: PMC6861669 DOI: 10.1016/j.omtn.2019.10.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 09/26/2019] [Accepted: 10/04/2019] [Indexed: 12/14/2022]
Abstract
The objective of this study was to investigate the role of lncRNA XIST and its relationship with miR-133a in myocardial I/R injury. H9C2 cells treated by hypoxia/reoxygenation (H/R) were used to establish an in vitro I/R model. The small interfering RNA (siRNA) for XIST and miR-133 mimics, inhibitor, and suppressor of cytokine signaling (SOCS2) recombinant plasmids were used to transfect the cells. Cell apoptosis was determined by flow cytometry analysis, and cell viability was used for 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide, Thiazolyl Blue Tetrazolium Bromide (MTT) assay. The dual-luciferase reporter assay was performed to confirm binding between XIST and miR-133a, as well as miR-133a and SOCS2. To inhibit or overexpress XIST, miR-133a, or SOCS2 in I/R mice, we used recombinant lentivirus vectors and adenovirus vectors for tail vein injection. The expression of XIST, miR-133a, and SOCS2 was determined by quantitative real-time PCR, and LC3 I/II and Beclin1 was determined by western blotting. The expression of XIST and SOCS2 was significantly upregulated, whereas the miR-133a level was remarkably downregulated in both H/R H9C2 cells and I/R mice myocardial tissues. In both H/R H9C2 cells and I/R mice, the inhibition of XIST led to decreased apoptosis and autophagy, and inhibition of miR-133a reversed these effects. Similarly, overexpression of miR-133a resulted in reduced apoptosis and autophagy, which were reversed by overexpression of SOCS2. The inhibition of XIST and overexpression of miR-133a also promote cell viability of H/R cells. The dual-luciferase reporter assay significantly showed that XIST directly targeted on miR-133a, and miR-133a directly targeted on SOCS2. The inhibition of XIST could improve myocardial I/R injury by regulation of the miR-133a/SOCS2 axis and inhibition of autophagy.
Collapse
Affiliation(s)
- Zhiqiang Li
- Department of Cardiovascular Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Yaping Zhang
- Department of Heart Center, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Nan Ding
- Department of Cardiovascular Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Yudong Zhao
- Department of Cardiovascular Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Zankai Ye
- Department of Cardiovascular Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Lei Shen
- Department of Cardiovascular Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Hanlu Yi
- Department of Cardiovascular Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Yaobin Zhu
- Department of Cardiovascular Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China.
| |
Collapse
|
27
|
The Impact of MicroRNA-133a on Prognosis and Clinicopathological Parameters for Digestive System Cancers: a Comprehensive Study Based on Meta-Analysis and TCGA Database. Pathol Oncol Res 2019; 26:771-781. [PMID: 30810894 DOI: 10.1007/s12253-019-00619-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 02/18/2019] [Indexed: 01/11/2023]
Abstract
We conducted a meta-analysis on the impact of microRNA-133a (miR-133a) on digestive system cancers, and verified the results through The Cancer Genome Atlas (TCGA). Relevant studies were searched in English and Chinese database and meta-analysis was performed using Stata 12.0. The corresponding information of miR-133a and digestive system cancers were obtained from TCGA database and analysis was performed using SPSS. Increased miR-133a expression was linked with favorable overall survival (OS) in digestive system cancers (HR = 0.539, 95% CI: 0.416-0.698, P < 0.001), digestive tract cancers (HR =0.558, 95% CI: 0.406-0.767, P < 0.001), esophageal squamous cell carcinoma (ESCC) (HR = 0.427, 95% CI: 0.265-0.690, P = 0.001) and gastric cancer (HR = 0.541, 95% CI: 0.385-0.761, P < 0.001). The expression of miR-133a was significantly lower in cancer tissue compared with adjacent tissue for ESCC (P < 0.001), gastric cancer (P < 0.001), colorectal cancer (P < 0.001) and hepatocellular carcinoma (P = 0.002). Meanwhile, the area under the ROC curve (AUC) value for miR-133a was 0.836, 0.888, and 0.99 in ESCC, gastric cancer and colorectal cancer. MiR-133a is a tumor suppressor with prognostic and diagnostic values for digestive system cancers. High miR-133a expression was associated with better prognosis and less adverse clinicopathological parameters. More research should be performed to test these findings.
Collapse
|
28
|
Wang W, Zhong P, Yi JQ, Xu AX, Lin WY, Guo ZC, Wang CG, Sun CB, Chan S. Potential role for microRNA in facilitating physiological adaptation to hypoxia in the Pacific whiteleg shrimp Litopenaeus vannamei. FISH & SHELLFISH IMMUNOLOGY 2019; 84:361-369. [PMID: 30291981 DOI: 10.1016/j.fsi.2018.09.079] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 09/27/2018] [Accepted: 09/29/2018] [Indexed: 06/08/2023]
Abstract
Hypoxia is one of the most common physiological stressors in shrimp farming. Post-transcriptional regulation by microRNAs has been recognized as a ubiquitous strategy to enable transient phenotypic plasticity and adaptation to stressful environment, but involvement of microRNAs in hypoxia stress response of penaeid shrimp remains elusive. In this study, small RNA sequencing and comparative transcriptomic analysis was conducted to construct a comprehensive microRNA dataset for the whiteleg shrimp Litopenaeus vannamei exposed to hypoxia challenge. A total of 3324 known miRNAs and 8 putative novel miRNAs were identified, providing a valuable resource for future investigation on the functional mechanism of miRNAs in shrimp. Upon hypoxia, 1213 miRNAs showed significant differential expression, and many well-known miRNAs involved in hypoxia tolerance such as miR-210, let-7, miR-143 and miR-101 were identified. Remarkably, the vast majority of these miRNAs were up-regulated, suggesting that up-regulation of miRNAs may represent an effective strategy to inhibit protein translation under stressful hypoxic condition. The differentially expressed miRNAs were potentially targeting a wide variety of genes, including those with essential roles in hypoxia tolerance such as HIF1a and p53. GO and KEGG enrichment analysis further revealed that a broad range of biological processes and metabolic pathways were over-represented. Several GO terms associated with gene transcription and translation and KEGG pathways related to cytoskeleton remodeling, immune defense and signaling transduction were enriched, highlighting the crucial roles of these cellular events in the adaptation to hypoxia. Taken together, our study revealed that the differentially expressed miRNAs may regulate host response to hypoxia by modulating the expression of stress response genes such as HIF1a and p53 and affecting key cellular events involved in hypoxia adaptation. The findings would expand our knowledge of the biochemical and molecular underpinnings of hypoxia response strategies used by penaeid shrimp, and contribute to a better understanding of the molecular mechanisms of hypoxia tolerance in decapod crustaceans.
Collapse
Affiliation(s)
- Wei Wang
- College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, PR China
| | - Ping Zhong
- College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, PR China
| | - Jun-Qiao Yi
- College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, PR China
| | - Ai-Xuan Xu
- College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, PR China
| | - Wen-Yi Lin
- College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, PR China
| | - Zhen-Cong Guo
- College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, PR China
| | - Cheng-Gui Wang
- College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, PR China
| | - Cheng-Bo Sun
- College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, PR China.
| | - Siuming Chan
- College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, PR China.
| |
Collapse
|
29
|
Wang Y, Liu LL, Tian Y, Chen Y, Zha WH, Li Y, Wu FJ. Upregulation of DAPK2 ameliorates oxidative damage and apoptosis of placental cells in hypertensive disorder complicating pregnancy by suppressing human placental microvascular endothelial cell autophagy through the mTOR signaling pathway. Int J Biol Macromol 2018; 121:488-497. [PMID: 30243997 DOI: 10.1016/j.ijbiomac.2018.09.111] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 08/27/2018] [Accepted: 09/19/2018] [Indexed: 12/17/2022]
Abstract
Death-associated protein kinase 2 (DAPK2) has indicated functional roles in cellular processes, including survival, apoptosis, and autophagy. This study is aimed to identify the effect of DAPK2 on oxidative damage and apoptosis of placental cells in hypertensive disorder complicating pregnancy (HDCP) through mTOR pathway. Microarray-based gene expression analysis was performed to predict the differentially expressed genes related to HDCP. To investigate the specific mechanism of DAPK2 in HDCP cells, placental microvascular endothelial cells were treated with mimic or siRNA of DAPK2 and mTOR to detect the expression of related genes, cell autophagy and apoptosis and oxidative damage. Finally, rats were modeled with HDCP to verify the cell experiment results. DAPK2 was downregulated in HDCP, and could activate mTOR. Besides, DAPK2 overexpression led to decreases in autophagy in HPVECs as well as apoptosis and oxidative damage in placental cells indicated by a substantial decrease in Beclin-1, LC3 II/LC3 I and Bax along with an increase in Bcl-2, 4EBP1 and p70S6K. It also ameliorates blood pressure elevation in HDCP rats. The study defined remission effect of DAPK2 on placental cell oxidative damage and apoptosis in HDCP via mTOR activation. Together, DAPK2 regulating mTOR pathway presents a promising therapy for HDCP treatment.
Collapse
Affiliation(s)
- Yan Wang
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun 130041, PR China
| | - Lian-Lian Liu
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun 130041, PR China
| | - Yuan Tian
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun 130041, PR China
| | - Yang Chen
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun 130041, PR China
| | - Wen-Hui Zha
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun 130041, PR China
| | - Yang Li
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun 130041, PR China
| | - Fu-Ju Wu
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun 130041, PR China.
| |
Collapse
|
30
|
Integrative analysis of differentially expressed genes and miRNAs predicts complex T3-mediated protective circuits in a rat model of cardiac ischemia reperfusion. Sci Rep 2018; 8:13870. [PMID: 30218079 PMCID: PMC6138681 DOI: 10.1038/s41598-018-32237-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 07/12/2018] [Indexed: 12/22/2022] Open
Abstract
Thyroid hormone (T3) dyshomeostasis in the cardiac ischemia-reperfusion (IR) setting negatively impacts on mitochondria function and extracellular matrix remodeling. The modulation of cardiac miRNAs may represent the underlying molecular mechanisms, but a systems biology perspective investigating this critical issue in depth is still lacking. A rat model of myocardial IR, with or without an early short-term T3-replacement, was used to predict putative T3-dependent miRNA-gene interactions targeted to mitochondria quality control and wound healing repair. As evidenced by mRNA and miRNA expression profiling, the T3 supplementation reverted the expression of 87 genes and 11 miRNAs that were dysregulated in the untreated group. In silico crossing and functional analysis of the T3-associated differentially expressed transcripts, identified a signature of interconnected miRNA-gene regulatory circuits that confer resistance to noxious cascades of acute stress. In this network the T3-down-regulated Tp53, Jun and Sp1 transcription factors emerge as critical nodes linking intrinsic cell death and oxidative stress pathways to adverse remodeling cascades. The data presented here provide a novel insight into the molecular basis of T3 cardioprotection in the early post-IR phase and highlight the contribution of a previously unappreciated complex T3-regulatory network that may be helpful in translating T3 replacement into clinical practice.
Collapse
|
31
|
Potential role for microRNA in regulating hypoxia-induced metabolic suppression in jumbo squids. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2018; 1861:586-593. [DOI: 10.1016/j.bbagrm.2018.04.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 04/30/2018] [Accepted: 04/30/2018] [Indexed: 12/19/2022]
|
32
|
Ong SB, Katwadi K, Kwek XY, Ismail NI, Chinda K, Ong SG, Hausenloy DJ. Non-coding RNAs as therapeutic targets for preventing myocardial ischemia-reperfusion injury. Expert Opin Ther Targets 2018; 22:247-261. [DOI: 10.1080/14728222.2018.1439015] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Sang-Bing Ong
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Khairunnisa Katwadi
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Xiu-Yi Kwek
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Nur Izzah Ismail
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore Medical School, Singapore, Singapore
- Department of Biomedical Engineering, Faculty of Engineering, University of Malaya, Kuala Lumpur, Malaysia
| | - Kroekkiat Chinda
- Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
- Biomedical Research Unit in Cardiovascular Sciences (BRUCS), Naresuan University, Phitsanulok, Thailand
| | - Sang-Ging Ong
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Derek J Hausenloy
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore Medical School, Singapore, Singapore
- National Heart Research Institute of Singapore, National Heart CentreSingapore, Singapore
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, London, UK
- Yong Loo Lin School of Medicine, National University Singapore, Singapore, Singapore
| |
Collapse
|
33
|
Hao W, Zhao ZH, Meng QT, Tie ME, Lei SQ, Xia ZY. Propofol protects against hepatic ischemia/reperfusion injury via miR-133a-5p regulating the expression of MAPK6. Cell Biol Int 2017; 41:495-504. [PMID: 28198596 DOI: 10.1002/cbin.10745] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 02/13/2017] [Indexed: 12/16/2022]
Affiliation(s)
- Wei Hao
- Department of Anesthesiology; Renmin Hospital of Wuhan University; Wuhan 430060 China
| | - Zhi-Hui Zhao
- Department of Anesthesiology; Inner Mongolia Autonomous Region People's Hospital; Huhhot 010017 China
| | - Qing-Tao Meng
- Department of Anesthesiology; Renmin Hospital of Wuhan University; Wuhan 430060 China
| | - Mu-Er Tie
- Department of Anesthesiology; Inner Mongolia Autonomous Region People's Hospital; Huhhot 010017 China
| | - Shao-Qing Lei
- Department of Anesthesiology; Renmin Hospital of Wuhan University; Wuhan 430060 China
| | - Zhong-Yuan Xia
- Department of Anesthesiology; Renmin Hospital of Wuhan University; Wuhan 430060 China
| |
Collapse
|
34
|
Matsuzaka Y, Tanihata J, Komaki H, Ishiyama A, Oya Y, Rüegg U, Takeda SI, Hashido K. Characterization and Functional Analysis of Extracellular Vesicles and Muscle-Abundant miRNAs (miR-1, miR-133a, and miR-206) in C2C12 Myocytes and mdx Mice. PLoS One 2016; 11:e0167811. [PMID: 27977725 PMCID: PMC5158003 DOI: 10.1371/journal.pone.0167811] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 11/21/2016] [Indexed: 12/20/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a progressive neuromuscular disorder. Here, we show that the CD63 antigen, which is located on the surface of extracellular vesicles (EVs), is associated with increased levels of muscle-abundant miRNAs, namely myomiRs miR-1, miR-133a, and miR-206, in the sera of DMD patients and mdx mice. Furthermore, the release of EVs from the murine myoblast C2C12 cell line was found to be modulated by intracellular ceramide levels in a Ca2+-dependent manner. Next, to investigate the effects of EVs on cell survival, C2C12 myoblasts and myotubes were cultured with EVs from the sera of mdx mice or C2C12 cells overexpressing myomiRs in presence of cellular stresses. Both the exposure of C2C12 myoblasts and myotubes to EVs from the serum of mdx mice, and the overexpression of miR-133a in C2C12 cells in presence of cellular stress resulted in a significant decrease in cell death. Finally, to assess whether miRNAs regulate skeletal muscle regeneration in vivo, we intraperitoneally injected GW4869 (an inhibitor of exosome secretion) into mdx mice for 5 and 10 days. Levels of miRNAs and creatine kinase in the serum of GW4869-treated mdx mice were significantly downregulated compared with those of controls. The tibialis anterior muscles of the GW4869-treated mdx mice showed a robust decrease in Evans blue dye uptake. Collectively, these results indicate that EVs and myomiRs might protect the skeletal muscle of mdx mice from degeneration.
Collapse
Affiliation(s)
- Yasunari Matsuzaka
- Administrative Section of Radiation Protection, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Jun Tanihata
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Hirofumi Komaki
- Department of Child Neurology, Hospital, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Akihiko Ishiyama
- Department of Child Neurology, Hospital, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Yasushi Oya
- Department of Neurology, Hospital, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Urs Rüegg
- Department of Pharmacology, Geneva-Lausanne School of Pharmaceutical Sciences, University of Geneva and University of Lausanne, Geneva, Switzerland
| | - Shin-ichi Takeda
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Kazuo Hashido
- Administrative Section of Radiation Protection, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
- * E-mail:
| |
Collapse
|
35
|
Zhang Y, Huang R, Zhou W, Zhao Q, Lü Z. miR-192-5p mediates hypoxia/reoxygenation-induced apoptosis in H9c2 cardiomyocytes via targeting of FABP3. J Biochem Mol Toxicol 2016; 31. [PMID: 27780314 DOI: 10.1002/jbt.21873] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 10/02/2016] [Indexed: 12/21/2022]
Abstract
Myocardial ischemia/reperfusion (I/R) injury is a leading cause of morbidity and mortality. In this study, we investigated the role of miR-192-5p in hypoxia/reoxygenation (H/R)-induced cardiomyocyte apoptosis. H9c2 cardiomyocytes were subjected to H/R and tested for miR-192-5p expression. Overexpression and knockdown experiments were performed to determine the effects of manipulating miR-192-5p on apoptotic responses. H/R-treated H9c2 cells exhibited a 2.2-fold increase in miR-192-5p levels. Overexpression of miR-192-5p significantly augmented apoptosis in H9c2 cells after H/R, which was accompanied by a significant increase in the ratio of Bax/Bcl-2. In contrast, delivery of anti-miR-192-5p inhibitors significantly reduced apoptosis induced by H/R. FABP3 was identified to be a functional target of miR-192-5p. Restoration of FABP3 prevented apoptosis in miR-192-5p-transfected H9c2 cells, whereas downregulation of FABP3 enhanced apoptosis in H/R-exposed H9c2 cells. In conclusion, miR-192-5p mediates H/R-induced apoptosis in cardiomyocytes by targeting FABP3 and represents a potential target for prevention of myocardial I/R injury.
Collapse
Affiliation(s)
- Yuefeng Zhang
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Risheng Huang
- Department of Cardiothoracic Surgery, Wenzhou Central Hospital, Wenzhou, People's Republic of China
| | - Weihe Zhou
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Qifeng Zhao
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Zhenye Lü
- Department of Cardiothoracic Surgery, Wenzhou People's Hospital, Wenzhou, People's Republic of China
| |
Collapse
|
36
|
Chistiakov DA, Orekhov AN, Bobryshev YV. Cardiac-specific miRNA in cardiogenesis, heart function, and cardiac pathology (with focus on myocardial infarction). J Mol Cell Cardiol 2016; 94:107-121. [PMID: 27056419 DOI: 10.1016/j.yjmcc.2016.03.015] [Citation(s) in RCA: 218] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 02/09/2016] [Accepted: 03/24/2016] [Indexed: 12/21/2022]
Abstract
Cardiac miRNAs (miR-1, miR133a, miR-208a/b, and miR-499) are abundantly expressed in the myocardium. They play a central role in cardiogenesis, heart function and pathology. While miR-1 and miR-133a predominantly control early stages of cardiogenesis supporting commitment of cardiac-specific muscle lineage from embryonic stem cells and mesodermal precursors, miR-208 and miR-499 are involved in the late cardiogenic stages mediating differentiation of cardioblasts to cardiomyocytes and fast/slow muscle fiber specification. In the heart, miR-1/133a control cardiac conductance and automaticity by regulating all phases of the cardiac action potential. miR-208/499 located in introns of the heavy chain myosin genes regulate expression of sarcomeric contractile proteins. In cardiac pathology including myocardial infarction (MI), expression of cardiac miRNAs is markedly altered that leads to deleterious effects associated with heart wounding, arrhythmia, increased apoptosis, fibrosis, hypertrophy, and tissue remodeling. In acute MI, circulating levels of cardiac miRNAs are significantly elevated making them to be a promising diagnostic marker for early diagnosis of acute MI. Great cardiospecific capacity of these miRNAs is very helpful for enhancing regenerative properties and survival of stem cell and cardiac progenitor transplants and for reprogramming of mature non-cardiac cells to cardiomyocytes.
Collapse
Affiliation(s)
- Dimitry A Chistiakov
- Department of Molecular Genetic Diagnostics and Cell Biology, Division of Laboratory Medicine, Institute of Pediatrics, Research Center for Children's Health, 119991 Moscow, Russia
| | - Alexander N Orekhov
- Institute of General Pathology and Pathophysiology, Russian Academy of Sciences, Moscow 125315, Russia; Department of Biophysics, Biological Faculty, Moscow State University, Moscow 119991, Russia; Institute for Atherosclerosis Research, Skolkovo Innovative Center, Moscow 121609, Russia
| | - Yuri V Bobryshev
- Institute of General Pathology and Pathophysiology, Russian Academy of Sciences, Moscow 125315, Russia; Faculty of Medicine, School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia; School of Medicine, University of Western Sydney, Campbelltown, NSW 2560, Australia.
| |
Collapse
|
37
|
Chistiakov DA, Orekhov AN, Bobryshev YV. Cardiac Extracellular Vesicles in Normal and Infarcted Heart. Int J Mol Sci 2016; 17:ijms17010063. [PMID: 26742038 PMCID: PMC4730308 DOI: 10.3390/ijms17010063] [Citation(s) in RCA: 128] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 12/10/2015] [Accepted: 12/18/2015] [Indexed: 02/06/2023] Open
Abstract
Heart is a complex assembly of many cell types constituting myocardium, endocardium and epicardium that intensively communicate to each other in order to maintain the proper cardiac function. There are many types of intercellular intracardiac signals, with a prominent role of extracellular vesicles (EVs), such as exosomes and microvesicles, for long-distant delivering of complex messages. Cardiomyocytes release EVs, whose content could significantly vary depending on the stimulus. In stress, such as hypoxia, inflammation or injury, cardiomyocytes increase secretion of EVs. In hypoxic conditions, cardiac EVs are enriched with angiogenic and prosurvival factors. In acute myocardial infarction (AMI), damaged cardiac muscle cells produce EVs with increased content of angiogenic, anti-apoptotic, mitogenic and growth factors in order to induce repair and healing of the infarcted myocardium. Exosomal microRNAs play a central role in cardiac regeneration. In AMI, circulating cardiac EVs abundantly contain cardiac-specific miRNAs that serve as indicators of cardiac damage and have a big diagnostic potential as AMI biomarkers. Cardioprotective and regenerative properties of exosomes derived from cardiac and non-cardiac stem/progenitor cells are very helpful to be used in cell-free cardiotherapy and regeneration of post-infarct myocardium.
Collapse
Affiliation(s)
- Dimitry A Chistiakov
- Department of Molecular Genetic Diagnostics and Cell Biology, Division of Laboratory Medicine, Institute of Pediatrics, Research Center for Children's Health, 119991 Moscow, Russia.
| | - Alexander N Orekhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315 Moscow, Russia.
- Institute for Atherosclerosis Research, Skolkovo Innovative Center, 143025 Moscow, Russia.
- Department of Biophysics, Biological Faculty, Moscow State University, 119991 Moscow, Russia.
| | - Yuri V Bobryshev
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315 Moscow, Russia.
- Faculty of Medicine, School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia.
- School of Medicine, University of Western Sydney, Campbelltown, NSW 2560, Australia.
| |
Collapse
|
38
|
Wu Z, Qi Y, Guo Z, Li P, Zhou D. miR-613 suppresses ischemia-reperfusion-induced cardiomyocyte apoptosis by targeting the programmed cell death 10 gene. Biosci Trends 2016; 10:251-7. [DOI: 10.5582/bst.2016.01122] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Zhenhua Wu
- ICU, Departmentof Cardiac Surgery, Tianjin Chest Hospital
| | - Yujuan Qi
- ICU, Departmentof Cardiac Surgery, Tianjin Chest Hospital
| | - Zhigang Guo
- Department of Cardiac Surgery, Tianjin Chest Hospital
| | - Peijun Li
- ICU, Departmentof Cardiac Surgery, Tianjin Chest Hospital
| | - Ding Zhou
- TEDA International Cardiovascular Hospital
| |
Collapse
|