1
|
Xu L, Tan X, Bai S, Wu H, Luo H, Ye Y, Fang L, Dai H, Huang L. L-arginine protects cementoblasts against hypoxia-induced apoptosis through Sirt1-enhanced autophagy. J Periodontol 2022; 93:1961-1973. [PMID: 34957557 DOI: 10.1002/jper.21-0473] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 11/16/2021] [Accepted: 11/17/2021] [Indexed: 01/05/2023]
Abstract
BACKGROUND L-arginine (L-arg) can reduce apoptosis in a variety of cells. Cementoblast apoptosis is related to root resorption during orthodontic treatment. In the present study, we aimed to study the regulatory effect and potential mechanism of L-arg on cementoblast apoptosis and root resorption. METHODS The apoptosis-related mRNA and protein expression of murine cementoblast (OCCM-30) was assessed after L-arg treatment. To investigate the role of Sirtuin 1 (Sirt1) and autophagy in L-arg resistance to cementoblast apoptosis and root absorption, resveratrol, and EX527 were used to activate or inhibit Sirt1, and chloroquine (CQ) was used to inhibit autophagy. RESULTS In vitro, L-arg inhibited hypoxia-induced apoptosis in OCCM-30. Further, L-arg increased Sirt1 expression whereas Sirt1 suppression by EX527 reversed the inhibitory effect of L-arg on cell apoptosis. Sirt1 activator resveratrol increased the ratio of microtubule-associated protein light chain 3 (LC3) II/I and decreased the expression of SQSTM1/p62 (p62), suggesting autophagy activation. Autophagy enhancement could reduce apoptosis. Caspase-3 and Bax expression was decreased, and Bcl-2 expression was increased. When autophagy was inhibited by CQ, the positive effects of Sirt1 were attenuated. In vivo, L-arg application reduced root resorption in rats, as demonstrated by decreased root absorption volume. Similarly, L-arg upregulated Sirt1, which activated autophagy in the root resorption model, and less root resorption was observed in the Sirt1 activation group. CONCLUSION L-arg reduced cementoblast apoptosis in hypoxia and reduced root resorption induced by loading force in rats, which may be partly mediated by Sirt1-enhanced autophagy.
Collapse
Affiliation(s)
- Lei Xu
- College of Stomatology, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Xi Tan
- College of Stomatology, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Siyu Bai
- College of Stomatology, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Hongyan Wu
- College of Stomatology, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Hong Luo
- College of Stomatology, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Yusi Ye
- College of Stomatology, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Lingli Fang
- College of Stomatology, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Hongwei Dai
- College of Stomatology, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Lan Huang
- College of Stomatology, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| |
Collapse
|
2
|
LncRNA HOTTIP Knockdown Attenuates Acute Myocardial Infarction via Regulating miR-92a-2/c-Met Axis. Cardiovasc Toxicol 2022; 22:352-364. [PMID: 35044621 PMCID: PMC8907089 DOI: 10.1007/s12012-021-09717-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 12/21/2021] [Indexed: 12/11/2022]
Abstract
Increasing investigations have focused on long non-coding RNAs (lncRNAs) in various human diseases, including acute myocardial infarction (AMI). Although lncRNA HOTTIP has been identified to play an important role in coronary artery diseases, its role and specific mechanism in AMI remain unclear. To investigate the potential role of HOTTIP in MI, HOTTIP expression in hypoxia-treated cardiomyocytes and myocardial tissues of MI mice was evaluated. The potential targets of HOTTIP and miR-92a-2 were predicted using Starbase and Targetscan. To further determine the cardio-protective effects of HOTTIP in vivo, si-HOTTIP and miR-92a-2 mimics were individually or co-injected into mice through intramyocardial injection. Moreover, their roles were further confirmed in rescue experiments. HOTTIP was significantly upregulated in ischemic myocardium of MI mice and hypoxia-induced cardiomyocytes. Moreover, HOTTIP knockdown markedly promoted cardiomyocyte growth and inhibited cardiomyocyte apoptosis in vitro. Luciferase reporter assay showed that HOTTIP could directly sponge miR-92a-2 to negatively regulate miR-92a-2 expression. In addition, c-Met was identified as a direct target of miR-92a-2, and their correlation was confirmed by luciferase reporter assay. MiR-92a-2 overexpression significantly enhanced the protective effect of HOTTIP knockdown against AMI through partially inhibiting c-Met expression. Our results demonstrated that HOTTIP downregulation attenuated AMI progression via the targeting miR-92a-2/c-Met axis and suggested that HOTTIP might be a potential therapeutic target for AMI.
Collapse
|
3
|
Sun J, Wang R, Chao T, Wang C. Long Noncoding RNAs Involved in Cardiomyocyte Apoptosis Triggered by Different Stressors. J Cardiovasc Transl Res 2021; 15:588-603. [PMID: 34855148 DOI: 10.1007/s12265-021-10186-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 11/04/2021] [Indexed: 12/26/2022]
Abstract
Cardiomyocytes are essential to maintain the normal cardiac function. Ischemia, hypoxia, and drug stimulation can induce pathological apoptosis of cardiomyocytes which eventually leads to heart failure, arrhythmia, and other cardiovascular diseases. Understanding the molecular mechanisms that regulate cardiomyocyte apoptosis is of great significance for the prevention and treatment of cardiovascular diseases. In recent years, more and more evidences reveal that long noncoding RNAs (lncRNAs) play important regulatory roles in myocardial cell apoptosis. They can modulate the expression of apoptosis-related genes at post-transcriptional level by altering the translation efficacy of target mRNAs or functioning as a precursor for miRNAs or competing for miRNA-mediated inhibition. Moreover, reversing the abnormal expression of lncRNAs can attenuate and even reverse the pathological apoptosis of cardiomyocytes. Therefore, apoptosis-related lncRNAs may become a potential new field for studying cardiomyocyte apoptosis and provide new ideas for the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Jinghui Sun
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ru Wang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Tiantian Chao
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chenglong Wang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
4
|
Lomis N, Westfall S, Shum-Tim D, Prakash S. Synthesis and characterization of peptide conjugated human serum albumin nanoparticles for targeted cardiac uptake and drug delivery. PLoS One 2021; 16:e0254305. [PMID: 34591850 PMCID: PMC8483410 DOI: 10.1371/journal.pone.0254305] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 09/03/2021] [Indexed: 12/15/2022] Open
Abstract
Congestive heart failure, a prominent cardiovascular disease results primarily from myocardial infarction or ischemia. Milrinone (MRN), a widely used clinical drug for heart failure, improves myocardial contractility and cardiac function through its inotropic and vasodilatory effects. However, lacking target specificity, it exhibits low bioavailability and lower body retention time. Therefore, in this study, angiotensin II (AT1) peptide conjugated human serum albumin nanoparticles (AT1-HSA-MRN-NPs) have been synthesized for targeted delivery of MRN to the myocardium, overexpressing AT1 receptors under heart failure. The NPs were surface functionalized through a covalent conjugation reaction between HSA and AT1. Nanoparticle size was 215.2±4.7 nm and zeta potential -28.8±2.7 mV and cumulative release of MRN was ~72% over 24 hrs. The intracellular uptake of nanoparticles and cell viability was studied in H9c2 cells treated with AT1-MRN-HSA-NPs vs the control non-targeted drug, MRN Lactate under normal, hypoxic and hypertrophic conditions. The uptake of AT1-HSA-MRN-NPs in H9c2 cells was significantly higher as compared to non-targeted nanoparticles, and the viability of H9c2 cells treated with AT1-MRN-HSA-NPs vs MRN Lactate was 73.4±1.4% vs 44.9±1.4%, respectively. Therefore, AT1-HSA-MRN-NPs are safe for in vivo use and exhibit superior targeting and drug delivery characteristics for treatment of heart failure.
Collapse
Affiliation(s)
- Nikita Lomis
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Montreal, QC, Canada
- Division of Experimental Medicine, Montréal, QC, Canada
| | - Susan Westfall
- Meakins Christie Laboratories, Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
| | - Dominique Shum-Tim
- Division of Cardiac Surgery and Surgical Research, Royal Victoria Hospital, Montréal, QC, Canada
| | - Satya Prakash
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Montreal, QC, Canada
- Division of Experimental Medicine, Montréal, QC, Canada
- * E-mail:
| |
Collapse
|
5
|
Qian Z, Zhu L, Li Y, Li Y, Wu Y, Fu S, Yang D. Icarrin prevents cardiomyocyte apoptosis in spontaneously hypertensive rats by inhibiting endoplasmic reticulum stress pathways. J Pharm Pharmacol 2021; 73:1023-1032. [PMID: 34018553 DOI: 10.1093/jpp/rgaa016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 10/05/2020] [Indexed: 01/10/2023]
Abstract
OBJECTIVES This study aimed to explore whether icarrin (ICA) can protect cardiomyocytes from hypertension-induced damage by inhibiting endoplasmic reticulum stress (ERS). METHODS Spontaneously hypertensive rats (SHRs) were orally administered water or ICA at 10, 20 and 40 mg/kg once daily for 12 weeks, and Wistar-Kyoto (WKY) rats were used as control. Changes in the growth and blood pressure of rats were assessed. Cardiac function was determined by ultrasound and the left ventricle mass was calculated. Myocardial tissue structure was assessed by haematoxylin and eosin staining, cardiomyocyte apoptosis was observed by TUNEL staining and the expression of ERS-related proteins was determined by western blotting. RESULTS In the SHR group, blood pressure was significantly high, left ventricular function decreased and left ventricular mass index increased. Additionally, left ventricular cardiomyocyte hypertrophy, disordered myofilament arrangement and increased cardiomyocyte apoptosis were observed by histological staining. ERS-induced proteins associated with apoptosis, including GRP78, PERK, ATF-6, ATF-4, CHOP, DR5, Caspase 12, c-JUN and ASK-1 were found to be highly expressed. ICA treatment reduced blood pressure and regulated the expression of proteins induced by ERS. Cardiomyocyte apoptosis decreased and left ventricular function improved. CONCLUSIONS ICA can inhibit ERS-induced apoptosis of cardiomyocytes and protect ventricular function in SHR.
Collapse
Affiliation(s)
- Zhiqiang Qian
- Key Laboratory of Basic Pharmacology of Ministry of Education, Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China.,Taizhou Jiangyan Hospital of TCM, Jiangyan Affiliated Hospital of Nanjing University of traditional Chinese Medicine, Taizhou, Jiangsu, China
| | - Ling Zhu
- Key Laboratory of Basic Pharmacology of Ministry of Education, Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Yeli Li
- Key Laboratory of Basic Pharmacology of Ministry of Education, Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Yiqi Li
- Key Laboratory of Basic Pharmacology of Ministry of Education, Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Yuting Wu
- Key Laboratory of Basic Pharmacology of Ministry of Education, Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Shu Fu
- Key Laboratory of Basic Pharmacology of Ministry of Education, Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Danli Yang
- Key Laboratory of Basic Pharmacology of Ministry of Education, Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
6
|
Chen M, Guo Y, Sun Z, Meng X. Long non-coding RNA SENCR alleviates hypoxia/reoxygenation-induced cardiomyocyte apoptosis and inflammatory response by sponging miR-1. Cardiovasc Diagn Ther 2021; 11:707-715. [PMID: 34295697 DOI: 10.21037/cdt-20-1037] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 02/21/2021] [Indexed: 12/14/2022]
Abstract
Background Myocardial cell apoptosis is one of the main reasons for the occurrence of acute myocardial infarction (AMI). The role of smooth muscle and endothelial cell enriched migration/differentiation-associated lncRNA (SENCR) in the cardiomyocyte apoptosis induced by hypoxia/reoxygenation (H/R) injury and its potential mechanism were investigated in this study to provide a novel biomarker for the development of AMI. Methods The expression levels of SENCR in the serum of AMI patients and non-AMI patients with chest pain (control) were detected by qRT-PCR. The function of SENCR in the cardiomyocyte apoptosis and inflammatory response induced by H/R injury was evaluated by MTT, cell apoptosis, and ELISA assay, respectively. The mechanism underlying the function of SENCR was investigated with the luciferase reporter assay. Results SENCR was significantly downregulated in AMI compared with the control volunteers, which showed negative correlations with the cardiac troponin I (cTnI) and creatine kinase-MB (CK-MB) level of patients. The H/R injury-induced cell apoptosis and inflammatory response in cardiomyocytes, which were attenuated by the overexpression of SENCR. The expression of miR-1 was suppressed by the overexpression of SENCR, while the overexpression of miR-1 could alleviate the cell apoptosis, enhance cell viability, and attenuate inflammatory response in cardiomyocyte. SENCR reversed H/R-induced myocardial cell injury by regulating the expression of miR-1. Conclusions SENCR was correlated with the clinicopathological features of patients and was revealed to alleviate the cardiomyocyte apoptosis and inflammatory response induced by H/R injury via sponging miR-1.
Collapse
Affiliation(s)
- Minghe Chen
- Second Department of Cardiology, Changle People's Hospital, Weifang, China
| | - Yini Guo
- First Department of Cardiology, Changle People's Hospital, Weifang, China
| | - Zongli Sun
- Second Department of Cardiology, Changle People's Hospital, Weifang, China
| | - Xiangjiang Meng
- Second Department of Cardiology, Changle People's Hospital, Weifang, China
| |
Collapse
|
7
|
Long R, Gao L, Li Y, Li G, Qin P, Wei Z, Li D, Qian C, Li J, Yang G. M2 macrophage-derived exosomes carry miR-1271-5p to alleviate cardiac injury in acute myocardial infarction through down-regulating SOX6. Mol Immunol 2021; 136:26-35. [PMID: 34058620 DOI: 10.1016/j.molimm.2021.05.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/20/2021] [Accepted: 05/13/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND Emerging evidence has indicated that exosomes serve as key regulators in acute myocardial infarction (AMI). This study was determined to investigate the effect of M2 macrophage-derived exosomes (M2-Exos) in AMI and the further mechanism. METHODS M2 macrophages were induced and M2-exos were isolated and verified. The AMI mouse model was prepared by ligation of the left anterior descending coronary artery (LAD) and then intravenously injected with the isolated M2-exos. The mouse cardiac function was assessed by echocardiography. Hematoxylin and eosin (HE) staining and TUNEL assay were conducted to examine myocardial lesion and apoptosis in cardiac tissues. The expressions of associated molecules were detected by quantitative real time-PCR (qRT-PCR) and western blot. MTT assay, Flow cytometry and Dual-luciferase reporter assay were carried out to detect cell viability, apoptosis and the interaction of miRNA and the target. RESULT M2-Exos could promote cardiac repair in AMI mice. M2-Exos suppressed apoptosis and enhanced viability of hypoxia-induced cardiomyocytes through delivery of miR-1271-5p. SOX6 is a direct target of miR-1271-5p. miR-1271-5p decreased cardiomyocyte apoptosis induced by hypoxia and alleviated cardiac injury in AMI via down-regulating SOX6 expression. CONCLUSION We identified that M2-Exos could carry miR-1271-5p to reduce apoptosis of cardiomyocytes and promote cardiac repair via down-regulating SOX6.
Collapse
Affiliation(s)
- Rui Long
- Department of Geriatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lu Gao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yunpeng Li
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guodong Li
- Department of Geriatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Peng Qin
- Department of Geriatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zihan Wei
- Department of Geriatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Dongbo Li
- Department of Geriatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Cheng Qian
- Department of Geriatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jing Li
- Department of Geriatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guojie Yang
- Department of Geriatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
8
|
How Can Malnutrition Affect Autophagy in Chronic Heart Failure? Focus and Perspectives. Int J Mol Sci 2021; 22:ijms22073332. [PMID: 33805128 PMCID: PMC8036550 DOI: 10.3390/ijms22073332] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/15/2021] [Accepted: 03/21/2021] [Indexed: 12/20/2022] Open
Abstract
Chronic heart failure (CHF) is a disease with important clinical and socio-economic ramifications. Malnutrition and severe alteration of the protein components of the body (protein disarrangements), common conditions in CHF patients, are independent correlates of heart dysfunction, disease progression, and mortality. Autophagy, a prominent occurrence in the heart of patients with advanced CHF, is a self-digestive process that prolongs myocardial cell lifespan by the removal of cytosolic components, such as aging organelles and proteins, and recycles the constituent elements for new protein synthesis. However, in specific conditions, excessive activation of autophagy can lead to the destruction of molecules and organelles essential to cell survival, ultimately leading to organ failure and patient death. In this review, we aim to describe the experimental and clinical evidence supporting a pathophysiological role of nutrition and autophagy in the progression of CHF. The understanding of the mechanisms underlying the interplay between nutrition and autophagy may have important clinical implications by providing molecular targets for innovative therapeutic strategies in CHF patients.
Collapse
|
9
|
Wang J, Dong G, Chi W, Nie Y. MiR-96 promotes myocardial infarction-induced apoptosis by targeting XIAP. Biomed Pharmacother 2021; 138:111208. [PMID: 33752931 DOI: 10.1016/j.biopha.2020.111208] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 12/01/2020] [Accepted: 12/26/2020] [Indexed: 12/12/2022] Open
Abstract
Acute myocardial infarction (AMI) has becoming a common leading cause of sudden death worldwide. MiR-96 has been identified that can target anti-apoptotic related genes in various human diseases. However, its role in AMI remains unclear. In this study, we found that miR-96 was significantly upregulated in the ischemic heart of MI mice (mice with myocardial infarction) and also in the H2O2-treated neonatal rat ventricular cardiomyocytes (CMs). In response H2O2, miR-96 inhibitor could significantly promote cell viability and reduce cell apoptosis of CMs, and inhibit the expression of Cleaved caspase-3 and Bax, while promote Bcl-2 expression. In addition, downregulation of miR-96 remarkedly reduced the infarct size and the percentages of apoptotic cells in the heart tissues of MI mice, and then protected against the damaged cardiac function. Moreover, we identified that XIAP (X-linked inhibitor of apoptosis) acted as a direct target gene of miR-96, meanwhile si-XIAP could obviously reverse miR-96 inhibitor induced protective effect in H2O2-treated CMs Taken together, our study demonstrated that miR-96 promoted AMI progression by directly targeting XIAP, and inhibiting the anti-apoptotic function of XIAP (Graphical abstract), which provided a novel therapeutic target for AMI treatment.
Collapse
Affiliation(s)
- Jianxiu Wang
- Hiser Medical Center of Qingdao, Qingdao City, Shandong Province, 266033, PR China.
| | - Guiling Dong
- Hiser Medical Center of Qingdao, Qingdao City, Shandong Province, 266033, PR China
| | - Weifeng Chi
- Hiser Medical Center of Qingdao, Qingdao City, Shandong Province, 266033, PR China
| | - Yingying Nie
- Hiser Medical Center of Qingdao, Qingdao City, Shandong Province, 266033, PR China.
| |
Collapse
|
10
|
Gao J, Feng W, Lv W, Liu W, Fu C. HIF-1/AKT Signaling-Activated PFKFB2 Alleviates Cardiac Dysfunction and Cardiomyocyte Apoptosis in Response to Hypoxia. Int Heart J 2021; 62:350-358. [PMID: 33678793 DOI: 10.1536/ihj.20-315] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Myocardial infarction (MI) is the most prevalent disease with severe mortality, and hypoxia-induced cardiac injury and cardiomyocyte apoptosis are the significant and harmful consequences of this disease. The cross talk between hypoxia signaling and glycolysis energy flux plays a critical role in modulating MI-related heart disorder. However, the underlying mechanism remains unclear. Here, we aimed to explore the effect of a key glycolytic enzyme of 6-phosphofructo-2-kinase/fructose-2, 6-bisphosphatase 2 (PFKFB2) on cardiac dysfunction and apoptosis in response to hypoxia. Our data demonstrated that the mRNA and protein expression of PFKFB2 were significantly elevated in the MI mice. The MI treatment promoted the activation of PFKFB2 in vivo, as presented by the remarkably increased phosphorylation levels of PFKFB2. PFKFB2 depletion enhanced MI-induced cardiac dysfunction and cardiomyocyte apoptosis in the MI mouse model. Moreover, hypoxia treatment dramatically upregulated the expression and activation of PFKFB2 in a time-dependent manner in cardiomyocytes. Hypoxia-stimulated PFKFB2 relieved hypoxia-induced cardiomyocyte apoptosis in vitro. PFKFB2 activated the fructose-2, 6-bisphosphate (Fru-2, 6-p2) /PFK/anaerobic adenosine triphosphate (ATP) glycolysis energy flux in response to hypoxia in cardiomyocytes. Mechanically, hypoxia-activated PFKFB2 by stimulating the hypoxia-inducible factor 1 (HIF-1) /ATK signaling. Thus, we conclude that HIF-1/AKT axis-activated PFKFB2 alleviates cardiac dysfunction and cardiomyocyte apoptosis in response to hypoxia. Our finding presents a new insight into the mechanism by which HIF-1/AKT/PFKFB2 signaling modulates MI-related heart disorder under the hypoxia condition, providing potential therapeutic targets and strategy for hypoxia-related myocardial injury.
Collapse
Affiliation(s)
- Juanyu Gao
- Department of Cardiology, Central Hospital Affiliated to Shandong First Medical University
| | - Wenjing Feng
- Department of Cardiology, Central Hospital Affiliated to Shandong First Medical University
| | - Wei Lv
- Department of Cardiology, Central Hospital Affiliated to Shandong First Medical University
| | - Wenhui Liu
- Department of Cardiology, The Second Hospital of Shandong University
| | - Caihua Fu
- Department of Cardiology, Central Hospital Affiliated to Shandong First Medical University
| |
Collapse
|
11
|
Long L, Yu Z, Chen S, Wu J, Liu Y, Peng J, Qu H, Fu C. Pretreatment of Huoxue Jiedu Formula Ameliorates Myocardial Ischaemia/Reperfusion Injury by Decreasing Autophagy via Activation of the PI3K/AKT/mTOR Pathway. Front Pharmacol 2021; 12:608790. [PMID: 33716739 PMCID: PMC7952439 DOI: 10.3389/fphar.2021.608790] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 01/26/2021] [Indexed: 12/31/2022] Open
Abstract
Background: Myocardial ischaemia/reperfusion (I/R) results in myocardial injury via excessive autophagy. Huoxue Jiedu Formula (HJF) has been widely applied in China for the treatment of ischaemic heart disease. However, the mechanisms of HJF are still poorly understood. Thus, the present experiment was designed to observe the effects of HJF on myocardial I/R injury and explore the possible mechanism. Methods: Myocardial injury in rats subjected to myocardial I/R was reflected by nitrotetrazolium blue chloride staining, thioflavin S staining, serum creatine kinase-MB (CK-MB) and cardiac troponin T (cTnT). Autophagy was determined by electron microscopy, laser confocal microscopy, Q-PCR and western blot. The possible pathway was predicted by network pharmacology and validated in vivo and in vitro. Results: Pretreatment of HJF decreased the no-reflow area, infarcted area, serum CK-MB levels and serum cTnT levels in I/R rat model. In addition, pretreatment of HJF decreased autophagy in heart tissues (decrease in Beclin-1 and LC3-II, and increase in Bcl-2, p62 and ratio of LC3-I/LC3-II). In the vivo study, pretreatment of HJF significantly decreased hypoxia/reoxygenation (H/R)-induced autophagy in H9C2 cells. Network pharmacology was applied to predict the possible mechanism by which HJF affects cardiac autophagy, and the PI3K/AKT/mTOR signalling pathway was the most significantly enriched pathway. And experimental studies demonstrated that pretreatment of HJF increased the phosphorylation of AKT and mTOR, and the effects of HJF on autophagy would be offset by PI3K inhibitor LY294002. Conclusion: Pretreatment of HJF ameliorates myocardial I/R injury by decreasing autophagy through activating PI3K/AKT/mTOR pathway.
Collapse
Affiliation(s)
- Linzi Long
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China.,Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zikai Yu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.,National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - ShengJun Chen
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China.,Jiangyin Tianjiang Pharmaceutical Co., Ltd., Jiangsu, China
| | - Jiarui Wu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yingying Liu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jun Peng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Hua Qu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.,National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Changgeng Fu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.,National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
12
|
Wei F, Ren B, Han W, Guan H, Jing G, Wang M. Investigate the Effect of miR-22 on the Apoptosis of Coronary Heart Disease Cells Through the Wnt-1 Pathway Based on Nano-Silica-Induced Rat Models. JOURNAL OF NANOSCIENCE AND NANOTECHNOLOGY 2021; 21:1338-1344. [PMID: 33183481 DOI: 10.1166/jnn.2021.18637] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
In this paper, by examining the toxicity of nano-silica to coronary heart disease cells, we explored the apoptosis of rat myocardial cells induced by nano-silica, and explored the effect of apoptosis on cells during the process of myocardial cytotoxicity induced by nano-silica. This article selects rat cardiomyocytes as the research object and conducts a group control experiment. A control group is set up with cells that are not stained with nano-silica. Different concentrations of nanosilica suspensions are applied to rat cells and detected by CCK-8 method. Cell survival rate after exposure to different concentrations of cells is used to determine the most stable exposure time and concentration. We used flow cytometry to detect intracellular reactive oxygen species and apoptotic rates, and used Western Blot to detect the expression of proteins that affect apoptosis. Finally, we investigated the effect of the Wnt signaling pathway on coronary heart disease. The Wnt signaling pathway regulates the development of the heart and blood vessels. In the treatment of cardiovascular disease, this pathway will be activated again to play a regulatory role. We conclude that nano-silica can induce cytotoxicity in rat myocardial cells through the Wnt-1 pathway, and nanosilica can induce myocardial cell apoptosis through the Wnt-1 pathway.
Collapse
Affiliation(s)
- Fangjing Wei
- Department of Cardiology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, China
| | - Baojun Ren
- Department of Cardiology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, China
| | - Wei Han
- Department of Cardiology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, China
| | - Hong Guan
- Department of Cardiology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, China
| | - Guoqiang Jing
- Department of Cardiology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, China
| | - Min Wang
- Department of Cardiology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, China
| |
Collapse
|
13
|
Cai X, Wang S, Hong L, Yu S, Li B, Zeng H, Yang X, Zhang P, Shao L. Inhibition of miR-322-5p Protects Cardiac Myoblast Cells Against Hypoxia-Induced Apoptosis and Injury Through Regulating CIAPIN1. J Cardiovasc Pharmacol 2021; 77:200-207. [PMID: 33538533 DOI: 10.1097/fjc.0000000000000949] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/15/2020] [Indexed: 01/11/2023]
Abstract
ABSTRACT Hypoxia leads to insufficient supply of blood and nutrients, which is major incentive for cardiomyocyte injury and apoptosis. Previous studies reported the regulation effects of microRNAs (miRNAs) in myocardial infarction, whereas function and molecular mechanisms of miR-322-5p were still unclear. Therefore, our study focused on the biological role of miR-322-5p in hypoxia-induced cardiac myoblast cells apoptosis and injury. The expression levels of miR-322-5p and cytokine-induced apoptosis inhibitor 1 (CIAPIN1) were measured by real-time quantitative polymerase chain reaction in cardiac myoblast cells. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazol-3-ium bromide (MTT), lactic dehydrogenase, and flow cytometry assays were performed to examine proliferation, injury, and apoptosis of cardiac myoblast cells, respectively. The protein expression levels were evaluated with western blot assay. The relationship between miR-322-5p and CIAPIN1 was confirmed by dual-luciferase reporter analysis. We found that miR-322-5p level was increased in cardiac myoblast cells exposed to hypoxia. In addition, miR-322-5p silencing could weaken injury and apoptosis in cardiac myoblast cells induced by hypoxia; meanwhile, inhibition of miR-322-5p activation of phosphatidylinositol-3 kinases (PI3K)/protein kinase B (AKT) signal pathway. Besides, CIAPIN1 was a target mRNA of miR-322-5p based on bioinformatics prediction. CIAPIN1 knockdown reversed the effects of miR-322-5p silencing on hypoxic cardiac myoblast cells. Suppression of miR-322-5p protected cardiac myoblast cells against hypoxia-induced injury and apoptosis through regulation of CIAPIN1 expression and PI3K/AKT signal pathway.
Collapse
Affiliation(s)
- Xinyong Cai
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, Jiangxi, China
| | - Shu Wang
- Department of Gerontology, The First Affliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Lang Hong
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, Jiangxi, China
| | - Songping Yu
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, Jiangxi, China
| | - Bin Li
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, Jiangxi, China
| | - Hong Zeng
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, Jiangxi, China
| | - Xu Yang
- Shenzhen Realomics (Biotech), Co. Ltd, Shenzhen, China ; and
| | - Ping Zhang
- Department of Neurology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, Jiangxi, China
| | - Liang Shao
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
14
|
Wu S, Zhang H, Chen N, Zhang C, Guo X. Metformin protects cardiomyocytes against oxygen-glucose deprivation injury by promoting autophagic flux through AMPK pathway. J Drug Target 2021; 29:551-561. [PMID: 33355497 DOI: 10.1080/1061186x.2020.1868478] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Metformin has been shown to protect myocardial ischaemia/reperfusion or hypoxia/reoxygenation injury. In our current study, we investigated the effects of metformin on autophagy and its possible underlying mechanisms in in vivo myocardial infarction (MI) model and in vitro oxygen-glucose deprivation (OGD) model. A rat model of MI was made by ligating coronary artery in vivo study. Metformin (200 mg/kg/day) could improve cardiac function, prevent rats from MI-induced injury by reducing myocardial infarct size and apoptosis. Moreover, metformin furtherly promoted autophagy by increasing the protein expression of LC3-II, ATG5, ATG7 and Beclin1, and by involving AMPK pathway during MI. H9c2 cells were treated with metformin (4 mM) in vitro study to assess its effects after exposure to OGD. Metformin increased cell viability and inhibited OGD-induced LDH synthesis and cell apoptosis. Furthermore, metformin increased autophagosome formations as well as expression of autophagy-related proteins, promoted autophagic flux. In addition, metformin augmented the protein level of Bcl-2 and diminished the protein levels of Bax and cleaved caspase-3. Metformin also upregulated p-AMPK expression. Nevertheless, the above-mentioned effects of metformin on H9c2 cells were remarkably eliminated by compound C (an AMPK inhibitor). In summary, we displayed that metformin protected cardiomyocytes against OGD-induced injury and apoptosis by promoting autophagic flux through the AMPK pathway.
Collapse
Affiliation(s)
- Shiyong Wu
- Department of Vascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hairong Zhang
- The First Clinical College, Chongqing Medical University, Chongqing, China
| | - Ningheng Chen
- Department of Vascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chuang Zhang
- Department of Vascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xueli Guo
- Department of Vascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
15
|
Therapeutic Value of miRNAs in Coronary Artery Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8853748. [PMID: 33953838 PMCID: PMC8057887 DOI: 10.1155/2021/8853748] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 01/25/2021] [Accepted: 03/29/2021] [Indexed: 12/14/2022]
Abstract
Atherosclerotic ischemic coronary artery disease (CAD) is a significant community health challenge and the principal cause of morbidity and mortality in both developed and developing countries for all ethnic groups. The progressive chronic coronary atherosclerosis is the main underlying cause of CAD. Although enormous progress occurred in the last three decades in the management of cardiovascular diseases, the prevalence of CAD continues to increase worldwide, indicating the need for discovery of deeper molecular insights of CAD mechanisms, biomarkers, and innovative therapeutic targets. Recently, several research groups established that microRNAs essentially regulate various cardiovascular development and functions, and a deregulated cardiac enriched microRNA profile plays a vital role in the pathogenesis of CAD and its biological aging. Numerous studies established that over- or downregulation of a single miRNA gene by ago-miRNA or anti-miRNA is enough to modify the CAD disease process, significantly prevent age-dependent cardiac cell death, and markedly improve cardiac function. In the light of more recent experimental and clinical evidences, we briefly reviewed and discussed the involvement of miRNAs in CAD and their possible diagnostic/therapeutic values. Moreover, we also focused on the role of miRNAs in the initiation and progression of the atherosclerosis plaque as the strongest risk factor for CAD.
Collapse
|
16
|
miR-126-5p regulates H9c2 cell proliferation and apoptosis under hypoxic conditions by targeting IL-17A. Exp Ther Med 2020; 21:67. [PMID: 33365067 DOI: 10.3892/etm.2020.9499] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 06/19/2020] [Indexed: 12/12/2022] Open
Abstract
Accumulating evidence has indicated that microRNAs (miRNAs/miRs) regulate the occurrence and development of various diseases, including diabetes, osteoporosis and cardiovascular conditions. However, the role of miRNAs in acute myocardial infarction (AMI) is not completely understood. The present study aimed to evaluate the therapeutic efficacy and mechanisms underlying the effects of miR-126-5p on H9c2 cell proliferation and apoptosis by targeting interleukin (IL)-17A. A total of 40 patients with AMI and 40 healthy volunteers were recruited in the present study and the expression levels of serum miR-126-5p and IL-17A were determined. Following confirmation that IL-17A was a target of miR-126-5p via a dual-luciferase reporter assay, H9c2 cells were exposed to hypoxic conditions. H9c2 cell viability and apoptosis were subsequently assessed. Additionally, the protein expression levels of apoptosis-associated proteins were detected following transfection. Compared with healthy individuals, miR-126-5p expression was significantly decreased in the serum samples of patients with AMI, whereas IL-17A, the target of miR-126-5p, was significantly increased. Following hypoxic treatment, miR-126-5p overexpression enhanced H9c2 cell viability compared with the NC group, which was subsequently reversed following co-transfection with pcDNA3.1-IL-17A. Additionally, the results indicated that hypoxia-induced H9c2 cell apoptosis was significantly reduced following transfection with miR-126-5p mimics via the PI3K/AKT signaling pathway compared with the NC group. The present study indicated that miR-126-5p may serve as a novel miRNA that regulates H9c2 cell viability and apoptosis by targeting IL-17A under hypoxic conditions. Therefore, miR-126-5p may serve as a crucial biomarker for the diagnosis of AMI.
Collapse
|
17
|
Gao J, Chen X, Shan C, Wang Y, Li P, Shao K. Autophagy in cardiovascular diseases: role of noncoding RNAs. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 23:101-118. [PMID: 33335796 PMCID: PMC7732971 DOI: 10.1016/j.omtn.2020.10.039] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cardiovascular diseases (CVDs) remain the world's leading cause of death. Cardiomyocyte autophagy helps maintain normal metabolism and functioning of the heart. Importantly, mounting evidence has revealed that autophagy plays a dual role in CVD pathology. Under physiological conditions, moderate autophagy maintains cell metabolic balance by degrading and recycling damaged organelles and proteins, and it promotes myocardial survival, but excessive or insufficient autophagy is equally deleterious and contributes to disease progression. Noncoding RNAs (ncRNAs) are a class of RNAs transcribed from the genome, but most ncRNAs do not code for functional proteins. In recent years, increasingly, various ncRNAs have been identified, and they play important regulatory roles in the physiological and pathological processes of organisms, as well as in autophagy. Thus, determining whether ncRNA-regulated autophagy plays a protective role in CVDs or promotes their progression can help us to develop ncRNAs as therapeutic targets in autophagy-related CVDs. In this review, we briefly summarize the regulatory roles of several important ncRNAs, including microRNAs (miRNAs), long ncRNAs (lncRNAs), and circular RNAs (circRNAs), in the autophagy of various CVDs to provide a theoretical basis for the etiology and pathogenesis of CVDs and develop novel therapies to treat CVDs.
Collapse
Affiliation(s)
- Jinning Gao
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Xiatian Chen
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Chan Shan
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Yin Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Peifeng Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Kai Shao
- Department of Central Laboratory, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, Shandong 266035, China
| |
Collapse
|
18
|
Si Q, Shi Y, Huang D, Zhang N. Diosmetin alleviates hypoxia‑induced myocardial apoptosis by inducing autophagy through AMPK activation. Mol Med Rep 2020; 22:1335-1341. [PMID: 32627001 PMCID: PMC7339627 DOI: 10.3892/mmr.2020.11241] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Accepted: 08/08/2019] [Indexed: 12/12/2022] Open
Abstract
Diosmetin has shown great potential in the control of several diseases. The aim of the present study was to evaluate the role of diosmetin as a candidate agent for the treatment of myocardial infarction which was mainly caused by hypoxia. The model of hypoxia‑injured myocardial cells was established using the H9c2 cell line. Cell viability was determined using Cell Counting Kit‑8, cell apoptosis was determined by Annexin V‑FITC Apoptosis Detection Kit and cleaved caspase‑3 level was assessed by western blot analysis. Autophagy was monitored using a commercial kit, and a well‑established reporter system was used to confirm the role of diosmetin in autophagy. The activity of adenosine 5'‑monophosphate‑activated protein kinase (AMPK) signaling was detected by western blot analysis. Cell viability assay indicated that diosmetin alleviated hypoxia‑induced cell death of H9c2 cells in a dose‑dependent manner. Data of the apoptosis assay revealed that diosmetin reduced the proportion of apoptotic cells in the hypoxia‑injured H9c2 cells. It was also found that the occurrence of autophagy was promoted when hypoxia‑injured cells were treated with diosmetin alone, and results of the western blot analysis revealed that AMPK signaling was activated by diosmetin. Administration of diosmetin together with an inhibitor of autophagy (3‑methyladenine, 3‑MA) or AMPK (Compound C) was able to decrease the diosmetin‑induced autophagy as well as the cytoprotective effects in the hypoxia‑injured cells. Our study concluded that diosmetin exhibits a cytoprotective effect on hypoxia‑injured myocardial cells by inducing autophagy and alleviating apoptosis. AMPK was demonstrated to regulate the observed effects caused by diosmetin. This investigation confirmed diosmetin as a promising drug candidate for myocardial infarction treatment. The present findings regarding the inherent molecular mechanisms involved in the protective effects of diosmetin promote the clinical application of diosmetin.
Collapse
Affiliation(s)
- Qijun Si
- Clinical Laboratory, Zhuji People's Hospital, Zhuji, Zhejiang 311800, P.R. China
| | - Yujie Shi
- Cardiovascular Disease Institute, PLA Army General Hospital, Beijing 100000, P.R. China
| | - Dandan Huang
- Preclinical School, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Na Zhang
- Department of Internal Medicine, The Hospital of Wuhan University, Wuhan, Hubei 430000, P.R. China
| |
Collapse
|
19
|
Identification of a miRNA Based-Signature Associated with Acute Coronary Syndrome: Evidence from the FLORINF Study. J Clin Med 2020; 9:jcm9061674. [PMID: 32492915 PMCID: PMC7356017 DOI: 10.3390/jcm9061674] [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: 05/11/2020] [Revised: 05/27/2020] [Accepted: 05/27/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The discovery of novel biomarkers that improve risk prediction models of acute coronary syndrome (ACS) is needed to better identify and stratify very high-risk patients. MicroRNAs (miRNAs) are essential non-coding modulators of gene expression. Circulating miRNAs recently emerged as important regulators and fine-tuners of physiological and pathological cardiovascular processes; therefore, specific miRNAs expression profiles may represent new risk biomarkers. The aims of the present study were: i) to assess the changes in circulating miRNAs levels associated with ACS and ii) to evaluate the incremental value of adding circulating miRNAs to a clinical predictive risk model. METHODS AND RESULTS The study population included ACS patients (n = 99) and control subjects (n = 103) at high to very high cardiovascular risk but without known coronary event. Based on a miRNA profiling in a matched derivation case (n = -6) control (n = 6) cohort, 21 miRNAs were selected for validation. Comparing ACS cases versus controls, seven miRNAs were significantly differentially expressed. Multivariate logistic regression analyses demonstrated that among the seven miRNAs tested, five were independently associated with the occurrence of ACS. A receiver operating characteristic curve analysis revealed that the addition of miR-122 + miR-150 + miR-195 + miR-16 to the clinical model provided the best performance with an increased area under the curve (AUC) from 0.882 to 0.924 (95% CI 0.885-0.933, p = 0.003). CONCLUSIONS Our study identified a powerful signature of circulating miRNAs providing additive value to traditional risk markers for ACS.
Collapse
|
20
|
Li W, Ren Y, Meng T, Yang W, Zhang W. miR‐129‐5p attenuates hypoxia‐induced apoptosis in rat H9c2 cardiomyocytes by activating autophagy. J Gene Med 2020; 22:e3200. [PMID: 32298509 DOI: 10.1002/jgm.3200] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 04/06/2020] [Accepted: 04/06/2020] [Indexed: 12/17/2022] Open
Affiliation(s)
- Wenjia Li
- Department of Geriatric MedicineThe First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Shaanxi China
| | - Yanping Ren
- Department of Geriatric MedicineThe First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Shaanxi China
| | - Tianyu Meng
- Department of Geriatric MedicineThe First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Shaanxi China
| | - Wei Yang
- Department of Geriatric MedicineThe First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Shaanxi China
| | - Wei Zhang
- Department of Geriatric MedicineThe First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Shaanxi China
| |
Collapse
|
21
|
Hao Y, Yuan H, Yu H. Downregulation of miR-483-5p decreases hypoxia-induced injury in human cardiomyocytes by targeting MAPK3. Cell Mol Biol Lett 2020; 25:20. [PMID: 32206064 PMCID: PMC7079484 DOI: 10.1186/s11658-020-00213-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 02/26/2020] [Indexed: 12/16/2022] Open
Abstract
Background MiR-483-5p was recently identified as a risk factor in the early stages of acute myocardial infarction (AMI) patients. Here, we further investigated how miR-483-5p affects cardiomyocyte apoptosis and oxidative stress under hypoxic conditions. Methods Plasma samples were collected from AMI patients and healthy volunteers. The expression of miR-483-5p was determined using quantitative real-time PCR. An in vitro hypoxic model was constructed to mimic AMI in AC16 cells. Cell viability, apoptosis and oxidative stress biomarker levels (MDA, SOD and CAT) were respectively determined using CCK-8, flow cytometry and commercial assay kits. Results The expression levels of miR-483-5p were significantly higher in AMI patients than in control subjects. Circulating levels of miR-483-5p positively correlated with creatine kinase MB isoform (CK-MB) and cardiac troponin I (cTnI) levels. The in vitro experiments showed that the expression levels of miR-483-5p were also upregulated in hypoxia-induced AC16 cell injury. MiR-483-5p overexpression significantly increased hypoxia-induced cardiomyocyte apoptosis and oxidative stress, while knockdown attenuated these effects. Mechanistically, miR-483-5p directly targets MAPK3 in AC16 cells. Furthermore, the protective effects of miR-483-5p knockdown against hypoxia-induced cardiomyocyte injury are partially dependent on MAPK3. Conclusions MiR-483-5p, which targets MAPK3, might be a potential therapeutic target for the diagnosis and prevention of hypoxia-induced myocardial injury.
Collapse
Affiliation(s)
- Yan Hao
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, 324 Jingwu Road, Jinan, 250021 Shandong China
| | - Haitao Yuan
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, 324 Jingwu Road, Jinan, 250021 Shandong China
| | - Houzhi Yu
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, 324 Jingwu Road, Jinan, 250021 Shandong China
| |
Collapse
|
22
|
Wang Y, Zhao H, Guo M, Fei D, Zhang L, Xing M. Targeting the miR-122/PKM2 autophagy axis relieves arsenic stress. JOURNAL OF HAZARDOUS MATERIALS 2020; 383:121217. [PMID: 31546213 DOI: 10.1016/j.jhazmat.2019.121217] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 08/12/2019] [Accepted: 09/11/2019] [Indexed: 06/10/2023]
Abstract
Arsenic (As) is a natural hepatotoxicity inducer that is found ubiquitously in foods and environmental media. We found that arsenite exposure elicits autophagy in vivo and vitro, the specific role and regulatory mechanism of which are yet clear. MicroRNAs (miRNAs) are short noncoding RNAs that function in the posttranscriptional regulation of gene expression. Here, we report that miR-122, the most enriched constitutive miRNA in the liver, induced cell protective autophagy in arsenite-exposed hepatocytes. Arsenite exposure elevated miRNA-122 level and decreased the level of its target gene, PKM2. Under arsenic stress, overexpression of miR-122 significantly induced cell protective autophagy, characterized by lipidation of LC3-II and a corresponding consumption of p62. Conversely, autophagy inhibition by miR-122 knockdown was reversed by si-PKM2 cotransfection. We also found that miR-122 knockdown positively regulated the PI3K/Akt/mTOR pathway, and this phenomenon was reversed by cotransfecting cells with si-PKM2. Taken together, our findings show that the miR-122/PKM2 autophagy axis protects hepatocytes from arsenite stress via the PI3K/Akt/mTOR pathway; thus, miR-122 may be a potential candidate in the treatment of arseniasis.
Collapse
Affiliation(s)
- Yu Wang
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, Heilongjiang, PR China.
| | - Hongjing Zhao
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, Heilongjiang, PR China.
| | - Menghao Guo
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, Heilongjiang, PR China
| | - Dongxue Fei
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, Heilongjiang, PR China
| | - Lina Zhang
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, Heilongjiang, PR China
| | - Mingwei Xing
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, Heilongjiang, PR China.
| |
Collapse
|
23
|
Spotlight on epigenetic reprogramming in cardiac regeneration. Semin Cell Dev Biol 2020; 97:26-37. [PMID: 31002867 DOI: 10.1016/j.semcdb.2019.04.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 04/02/2019] [Accepted: 04/15/2019] [Indexed: 02/06/2023]
|
24
|
Gong L, Chang H, Xu H. RETRACTED: LncRNA MALAT1 knockdown alleviates oxygen-glucose deprivation and reperfusion induced cardiomyocyte apoptotic death by regulating miR-122. Exp Mol Pathol 2019; 111:104325. [PMID: 31669130 DOI: 10.1016/j.yexmp.2019.104325] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 09/05/2019] [Accepted: 10/22/2019] [Indexed: 01/01/2023]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (https://www.elsevier.com/about/our-business/policies/article-withdrawal). This article has been retracted at the request of the Editor-in-Chief. The journal was initially contacted by the corresponding author to request the retraction of the article. Given the comments of Dr Elisabeth Bik regarding this article “… the Western blot bands in all 400+ papers are all very regularly spaced and have a smooth appearance in the shape of a dumbbell or tadpole, without any of the usual smudges or stains. All bands are placed on similar looking backgrounds, suggesting they were copy/pasted from other sources, or computer generated”, the journal requested the authors to provide the raw data. However, the authors were not able to fulfil this request and therefore the Editor-in-Chief decided to retract the article.
Collapse
Affiliation(s)
- Licheng Gong
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Hong Chang
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Haiming Xu
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun 130033, China.
| |
Collapse
|
25
|
Song G, Zhu L, Ruan Z, Wang R, Shen Y. MicroRNA-122 promotes cardiomyocyte hypertrophy via targeting FoxO3. Biochem Biophys Res Commun 2019; 519:682-688. [PMID: 31543343 DOI: 10.1016/j.bbrc.2019.09.035] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 09/09/2019] [Indexed: 12/12/2022]
Abstract
OBJECTIVE -microRNAs (miRNAs) have emerged as novel regulators for cardiac hypertrophy. MiR-122 is well recognized as a promising therapeutic target in liver disease, whereas recently plays important roles in cardiovascular diseases. The current study aimed to explore the effect of miR-122 on the pathogenesis of cardiomyocyte hypertrophy. METHODS AND RESULTS -The cardiomyocytes isolated from the neonatal rat ventricular cardiomyocytes (NRVMs) were collected and performed to Angiotensin II (Ang II) administration. We observed a dramatically increased miR-122 expression in hypertrophic cardiomyocytes. The NRVMs transfected with miR-122 mimic or negative control were utilized for the functional analysis. Overexpression of miR-122 increased the morphology size of cardiomyocytes and promoted the pro-hypertrophic genes expression, whereas downregulated the anti-hypertrophic genes upon Ang II stimulation. The bioinformatics analysis and luciferase reporter assays exhibited that miR-122 directly targeted FoxO3 and attenuated its gene level in hypertrophic cardiomyocytes. Moreover, miR-122 negatively regulated FoxO3 but promoted calcineurin signaling pathway activation. Importantly, FoxO3 overexpression significantly reversed the effect of miR-122 on cardiomyocyte hypertrophy. CONCLUSION -Collected, our finding demonstrated that miR-122 accelerated the development of cardiomyocytes hypertrophy partially via directly regulation of FoxO3-calcineurin pathway.
Collapse
Affiliation(s)
- Guixian Song
- Department of Cardiology, Taizhou People's Hospital, Fifth Affiliated Hospital of Nantong University, Jiangsu Province, 225300, China
| | - Li Zhu
- Department of Cardiology, Taizhou People's Hospital, Fifth Affiliated Hospital of Nantong University, Jiangsu Province, 225300, China
| | - Zhongbao Ruan
- Department of Cardiology, Taizhou People's Hospital, Fifth Affiliated Hospital of Nantong University, Jiangsu Province, 225300, China
| | - Ruzhu Wang
- Department of Cardiology, Taizhou People's Hospital, Fifth Affiliated Hospital of Nantong University, Jiangsu Province, 225300, China
| | - Yahui Shen
- Department of Respiratory and Critical Care Medicine, Taizhou People's Hospital, Fifth Affiliated Hospital of Nantong University, Jiangsu Province, 225300, China.
| |
Collapse
|
26
|
Wu L, Chen Y, Chen Y, Yang W, Han Y, Lu L, Yang K, Cao J. Effect of HIF-1α/miR-10b-5p/PTEN on Hypoxia-Induced Cardiomyocyte Apoptosis. J Am Heart Assoc 2019; 8:e011948. [PMID: 31480879 PMCID: PMC6818010 DOI: 10.1161/jaha.119.011948] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background Few reports have addressed the mechanism by which microRNA miR-10b-5p regulates post-myocardial infarction (post-MI) cardiomyocyte apoptosis under hypoxic conditions. Methods and Results C57BL/6 mice underwent surgical ligation of the left anterior descending artery to create an MI or ischemia/reperfusion animal model. The expression of miR-10b-5p, PTEN (phosphatase and tensin homolog), and HIF-1α (hypoxia-inducible factor 1α) was detected in infarct border zone tissues at various time points. After precordial injections of the negative control or miR-10b-5p, overexpression lentiviruses were made in the areas surrounding the MI sites at 1 week, and myocardial infarct size, cardiac function, and cardiomyocyte apoptosis were examined. A miR-10b-5p mimic was transfected into primary mouse cardiomyocytes to analyze its effects on cardiomyocyte apoptosis and PTEN expression. Meanwhile, PTEN as a target of miR-10b-5p was verified via luciferase reporter gene assays. Cotransfection of miR-10b-5 and PTEN verified the relationship between miR-10b-5 and PTEN. Under hypoxic stress, the expression of HIF-1α and miR-10b-5p was examined. The results showed that miR-10b-5p expression was markedly reduced in the infarct border zone. Overexpression of miR-10b-5p in the murine model of MI significantly reduced MI size, improved cardiac function, and inhibited apoptosis. Overexpression of miR-10b-5p in vitro antagonized hypoxia-induced cardiomyocyte apoptosis and specifically inhibited the expression of the apoptosis-related gene PTEN, but overexpression of PTEN weakened these effects. We also found that hypoxia-induced accumulation of HIF-1α resulted in decreased expression of miR-10b-5p. Interfering with the activation of the HIF-1α signaling pathway promoted Pri-miR-10b and miR-10b-5p expression and inhibited PTEN expression. Conclusions MicroRNA miR-10b-5p antagonizes hypoxia-induced cardiomyocyte apoptosis, indicating that miR-10b-5p may serve as a potential future clinical target for the treatment of MI.
Collapse
Affiliation(s)
- Liping Wu
- Department of Geratology Ruijin Hospital Shanghai Jiaotong University School of Medicine Shanghai China
| | - Yafen Chen
- Department of Geratology Ruijin Hospital Shanghai Jiaotong University School of Medicine Shanghai China
| | - Yuanyuan Chen
- Institute of Cardiovascular Disease Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Wenbo Yang
- Institute of Cardiovascular Disease Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Yanxin Han
- Institute of Cardiovascular Disease Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Lin Lu
- Institute of Cardiovascular Disease Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Ke Yang
- Institute of Cardiovascular Disease Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Jiumei Cao
- Department of Geratology Ruijin Hospital Shanghai Jiaotong University School of Medicine Shanghai China
| |
Collapse
|
27
|
Jin Q, Chen Y. Silencing circular RNA circ_0010729 protects human cardiomyocytes from oxygen–glucose deprivation-induced injury by up-regulating microRNA-145-5p. Mol Cell Biochem 2019; 462:185-194. [DOI: 10.1007/s11010-019-03621-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 08/24/2019] [Indexed: 01/12/2023]
|
28
|
Zhang Z, Li H, Liu M, He J, Zhang X, Chen Y. Skullcapflavone I protects cardiomyocytes from hypoxia-caused injury through up-regulation of lincRNA-ROR. Int J Immunopathol Pharmacol 2019; 33:2058738419857537. [PMID: 31220954 PMCID: PMC6589964 DOI: 10.1177/2058738419857537] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Myocardial infarction (MI) is a serious heart disease in which cardiomyocytes are damaged, caused by hypoxia. This study explored the possible protective activity of Skullcapflavone I (SF I), a flavonoid isolated from the root of Scutellaria baicalensis Georgi, on hypoxia-stimulated cardiomyocytes cell injury in vitro. Viability and apoptosis of H9c2 cells and primary cardiomyocytes were tested using cell counting kit–8 (CCK-8) assay and Guava Nexin Reagent, respectively. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) was used to measure the long non-coding RNA regulator of reprogramming (lincRNA-ROR) expression. si-ROR was transfected to knockdown lincRNA-ROR. Western blotting was conducted to assess the protein levels of key molecules related to cell proliferation, apoptosis, and mitogen-activated protein kinase/extracellular signal–regulated kinase (MEK/ERK) pathway. We discovered that hypoxia stimulation obviously reduced H9c2 cell and primary cardiomyocytes’ viability and proliferation, but promoted cell apoptosis. SF I treatment mitigated the cell viability and proliferation inhibition, as well as cell apoptosis caused by hypoxia. Moreover, SF I promoted the hypoxia-caused up-regulation of lincRNA-ROR in H9c2 cells and primary cardiomyocytes. Knockdown of lincRNA-ROR reversed the influence of SF I on hypoxia-stimulated H9c2 cells and primary cardiomyocytes. Besides, SF I activated MEK/ERK pathway in H9c2 cells and primary cardiomyocytes via up-regulating lincRNA-ROR. To sum up, our research verified the beneficial activity of SF I on hypoxia-caused cardiomyocytes injury. SF I protected cardiomyocytes from hypoxia-caused injury through up-regulation of lincRNA-ROR and activation of MEK/ERK pathway.
Collapse
Affiliation(s)
- Zhenxiao Zhang
- 1 Department of Emergency, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hui Li
- 1 Department of Emergency, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Mingyang Liu
- 1 Department of Emergency, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jianshuai He
- 2 Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaotian Zhang
- 2 Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yuehua Chen
- 3 Department of Intensive Care Unit, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
29
|
Wu D, Zhang K, Hu P. The Role of Autophagy in Acute Myocardial Infarction. Front Pharmacol 2019; 10:551. [PMID: 31214022 PMCID: PMC6554699 DOI: 10.3389/fphar.2019.00551] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 05/01/2019] [Indexed: 12/14/2022] Open
Abstract
Acute myocardial infarction refers to a sudden death of cardiomyocytes, which leads to a large mortality worldwide. To attenuate acute myocardial infarction, strategies should be made to increase cardiomyocyte survival, improve postinfarcted cardiac function, and reverse the process of cardiac remodeling. Autophagy, a pivotal cellular response, has been widely studied and is known to be involved in various kinds of diseases. In the recent few years, the role of autophagy in diseases has been drawn increasing attention to by researchers. Here in this review, we mainly focus on the discussion of the effect of autophagy on the pathogenesis and progression of acute myocardial infarction under ischemic and ischemia/reperfusion injuries. Furthermore, several popular therapeutic agents and strategies taking advantage of autophagy will be described.
Collapse
Affiliation(s)
- Du Wu
- Department of Internal Medicine, The WuYun Mountain Sanatorium of Hangzhou, Hangzhou, China
| | - Kangfeng Zhang
- Department of Internal Medicine, The WuYun Mountain Sanatorium of Hangzhou, Hangzhou, China
| | - Pengfei Hu
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
30
|
Shi HJ, Wang MW, Sun JT, Wang H, Li YF, Chen BR, Fan Y, Wang SB, Wang ZM, Wang QM, Wang LS. A novel long noncoding RNA FAF inhibits apoptosis via upregulating FGF9 through PI3K/AKT signaling pathway in ischemia-hypoxia cardiomyocytes. J Cell Physiol 2019; 234:21973-21987. [PMID: 31093967 DOI: 10.1002/jcp.28760] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 04/03/2019] [Accepted: 04/10/2019] [Indexed: 01/03/2023]
Abstract
Long noncoding RNAs (lncRNAs) have been increasingly considered to play an important role in the pathological process of various cardiovascular diseases, which often bind to the proximal promoters of the protein-coding gene to regulate the protein expression. However, the functions and mechanisms of lncRNAs in cardiomyocytes have not been fully elucidated. High-throughput RNA sequencing was performed to identify the differently expressed lncRNAs and messenger RNAs (mRNAs) between acute myocardial infarction (AMI) rats and healthy controls. One novel lncRNA FGF9-associated factor (termed FAF) and mRNAs in AMI rats were verified by bioinformatics, real-time polymerase chain reaction or western blot. Moreover, RNA fluorescence in situ hybridization was performed to determine the location of lncRNA. Subsequently, a series of in vitro assays were used to observe the functions of lncRNA FAF in cardiomyocytes. The expression of lncRNA FAF and FGF9 were remarkably decreased in ischemia-hypoxia cardiomyocytes and heart tissues of AMI rats. Overexpression of FAF could significantly inhibit cardiomyocytes apoptosis induced by ischemia and hypoxia. Conversely, knockdown of lncRNA FAF could promote apoptosis in ischemia-hypoxia cardiomyocytes. Moreover, overexpression of lncRNA FAF could also increase the expression of FGF9. Knockdown of the FGF9 expression could promote apoptosis in cardiomyocytes with the insult of ischemia and hypoxia, which was consistent with the effect of lncRNA FAF overexpression on cardiomyocyte apoptosis. Mechanistically, FGF9 inhibited cardiomyocytes apoptosis through activating signaling tyrosine kinase FGFR2 via phosphoinositide 3-kinase/protein kinase B signaling pathway. Thus, lncRNA FAF plays a protective role in ischemia-hypoxia cardiomyocytes and may serve as a treatment target for AMI.
Collapse
Affiliation(s)
- Hao-Jie Shi
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ming-Wei Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jia-Teng Sun
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hao Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ya-Fei Li
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Bing-Rui Chen
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yi Fan
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Si-Bo Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zi-Mu Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qi-Ming Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lian-Sheng Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
31
|
Shi Y, Han Y, Niu L, Li J, Chen Y. MiR-499 inhibited hypoxia/reoxygenation induced cardiomyocytes injury by targeting SOX6. Biotechnol Lett 2019; 41:837-847. [PMID: 31076992 PMCID: PMC6551346 DOI: 10.1007/s10529-019-02685-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Accepted: 05/02/2019] [Indexed: 01/12/2023]
Abstract
Objective MiR-499 has been reported to be expressed only in cardiomyocytes, and its expression would increase after acute myocardial infarction (AMI). miR-499 plays a role in the process of cardiomyocytes injury induced by hypoxia/reoxygenation (H/R), however, it still remains unclear. Results Hypoxia inhibited miR-499-5p expression and H/R induced apoptosis. SOX6 was a target gene of miR-499-5p, and high expression of miR-499-5p inhibited the expression of SOX6. MiR-499-5p reduced H9c2 cells injury by inhibiting the expression of SOX6, overexpression of which could reverse the effect of miR-499-5p on H9c2 cells. MiR-499-5p inhibited the levels of LDH and MDA, while overexpression of miR-499-5p inhibited H/R-induced cell apoptosis. MiR-499-5p could up-regulate the level of Bcl-2 and down-regulate the expression levels of Bax and caspase-3. However, SOX6 partially reversed these effects of miR-499-5p. Conclusion We proved that miR-499-5p inhibited H/R-induced cardiomyocytes injury by targeting SOX6. Our results suggested that miR-499-5p/SOX6 pathway may present a potential therapeutic target for the treatment of AMI.
Collapse
Affiliation(s)
- Yujie Shi
- Department of Cardiology, Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, China
| | - Yunfeng Han
- Cardiovascular Disease Institute, PLA Army General Hospital, Beijing, China
| | - Lili Niu
- Cardiovascular Disease Institute, PLA Army General Hospital, Beijing, China
| | - Junxia Li
- Cardiovascular Disease Institute, PLA Army General Hospital, Beijing, China
| | - Yundai Chen
- Department of Cardiology, Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, China.
| |
Collapse
|
32
|
Wong LL, Saw EL, Lim JY, Zhou Y, Richards AM, Wang P. MicroRNA Let-7d-3p Contributes to Cardiac Protection via Targeting HMGA2. Int J Mol Sci 2019; 20:ijms20071522. [PMID: 30934671 PMCID: PMC6480063 DOI: 10.3390/ijms20071522] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 03/21/2019] [Accepted: 03/25/2019] [Indexed: 01/01/2023] Open
Abstract
We tested the hypothesis that Let-7d-3p contributes to cardiac cell protection during hypoxic challenge. Myoblast H9c2 cells and primary neonatal rat ventricular cardiomyocytes (NRVM) were transfected with five selected miRNA mimics. Both cell lines were subjected to 0.2% oxygen hypoxia. The protective effects of these miRNAs were determined by assessment of cell metabolic activity by CCK8 assay and measurement of lactate dehydrogenase (LDH) release as a marker of cell injury. Apoptosis and autophagy flux were assessed by Annexin V/7-AAD double staining and the ratio of LC3 II/I with Baf-A1 treatment, an autophagy flux inhibitor, respectively. Luciferase-reporter assay, RT-qPCR and Western blots were performed to identify the changes of relevant gene targets. Among five miRNA mimic transfections, Let-7d-3p increased CCK8 activity, and decreased LDH release in both H9c2 and NRVM during hypoxia. Apoptosis was significantly reduced in H9c2 cells transfected with Let-7d-3p mimic. Autophagy and autophagy flux were not affected. In silico, mRNAs of HMGA2, YY1, KLF9, KLF12, and MEX3C are predicted targets for Let-7d-3p. Luciferase-reporter assay confirmed that Let-7d-3p bound directly to the 3’-UTR region of HMGA2, MEX3C, and YY1, the down-regulations of these mRNAs were verified in both H9c2 and NRVM. The protein expression of HMGA2, but not others, was downregulated in H9c2 and NRVM. It is known that HMGA2 is a strong apoptosis trigger through the blocking of DNA repair. Thus, we speculate that the anti-apoptotic effects of Let-7d-3p mimic during hypoxia challenge are due to direct targeting of HMGA2.
Collapse
Affiliation(s)
- Lee Lee Wong
- Cardiovascular Research Institute, National University Heart Centre, Singapore 117599, Singapore.
- Department of Medicine, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore.
| | - Eng Leng Saw
- Cardiovascular Research Institute, National University Heart Centre, Singapore 117599, Singapore.
- Department of Medicine, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore.
| | - Jia Yuen Lim
- Cardiovascular Research Institute, National University Heart Centre, Singapore 117599, Singapore.
- Department of Medicine, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore.
| | - Yue Zhou
- Cardiovascular Research Institute, National University Heart Centre, Singapore 117599, Singapore.
- Department of Medicine, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore.
| | - Arthur Mark Richards
- Cardiovascular Research Institute, National University Heart Centre, Singapore 117599, Singapore.
- Department of Medicine, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore.
- Christchurch Heart Institute, Department of Medicine, University of Otago, Christchurch 8014, New Zealand.
| | - Peipei Wang
- Cardiovascular Research Institute, National University Heart Centre, Singapore 117599, Singapore.
- Department of Medicine, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore.
| |
Collapse
|
33
|
Effects of microRNA-195 on the Prognosis of Glioma Patients and the Proliferation and Apoptosis of Human Glioma Cells. Pathol Oncol Res 2019; 26:753-763. [PMID: 30806889 DOI: 10.1007/s12253-019-00622-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 02/19/2019] [Indexed: 02/07/2023]
Abstract
Glioma is the most common and aggressive intracranial malignant tumor with poor prognosis. Acts as a tumor suppressor, microRNA-195 (miR-195) plays important roles in a variety of cancers. However, the expression of miR-195 and role of miR-195 in glioma are still not well understood. 186 patients with glioma were enrolled and the follow-up period ranges from 1 to 69 months. MiR-195 was exogenously transfected into human glioma U87 cell line. The cell proliferation assay (CCK-8), colony formation assay, cell cycle analysis and cell apoptosis analysis were examined to investigate miR-195 effect on U87 cells. MiR-195 levels were reversely correlated with pathological grades (r = -0.487, p = 0.003). For patients with low miR-195 levels, their median survival time was 15 months, whereas the median survival time in patients with high miR-195 levels was 56.53 months. Multi-factor Cox regression analysis showed that high level of miR-195 (Odds ratio (OR): 0.347, 95% CI: 0.121-0.992) was associated with decreased mortality risk of patients. Moreover, overexpression of miR-195 inhibits proliferation and colony formation, and induces apoptosis of U87 cells. MiR-195 could block the glioma cells in G0/G1 phase, reducing S phase cells and regulating apoptosis related proteins (Caspase-3, Caspase-8, Caspase-9 and Bcl-2). Downregulation of miR-195 was associated with poor prognosis in human glioma. MiR-195 acted as tumor suppressor through inhibiting cell proliferation and promoting cell apoptosis via blockade of cell cycle and regulation of apoptosis related proteins.
Collapse
|
34
|
Dai M, Li L, Qin X. Clinical value of miRNA-122 in the diagnosis and prognosis of various types of cancer. Oncol Lett 2019; 17:3919-3929. [PMID: 30881509 PMCID: PMC6403504 DOI: 10.3892/ol.2019.10024] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 02/06/2019] [Indexed: 02/06/2023] Open
Abstract
The present study aimed to systematically analyze the value of microRNA-122 (miRNA-122) in the diagnosis and prognosis of hepatocellular carcinoma (HCC) and other types of cancer. First, the reverse transcription-quantitative polymerase chain reaction method was used to detect the expression levels of miRNA-122 in the serum samples of patients with HCC, benign lesions and healthy volunteers. Next, miRNA-seq data of miRNA-122 from The Cancer Genome Atlas database were used to analyze the differential expression and overall survival rate associated with a variety of types of cancer. Meanwhile, the target gene prediction of miRNA-122 was performed using four different software programs. Finally, 353 significant target genes were identified for Gene Ontology and Kyoto Encyclopedia of Genes and Genomes functional enrichment analysis. Finally, it was demonstrated that the expression levels of miRNA-122 in the HCC group were increased compared with the healthy group (P<0.001), but decreased with respect to the benign group (P<0.001). In addition, the combination of the miRNA-122 and a fetoprotein may further improve the diagnostic accuracy between the HCC and healthy groups (area under the curve, 0.980; 95% confidence interval, 0.958–1.000). It was also demonstrated that miRNA-122 exhibited significantly differential expression and the overall survival rate was predicted for various other types of cancer, including colorectal cancer, renal carcinoma, cholangiocarcinoma, prostate cancer and thyroid carcinoma. Functional enrichment analysis demonstrated that the target genes of miRNA-122 may contribute to the composition of the nucleus and cytoplasm, and regulate a variety of biological processes, including cardiac muscle cell differentiation and glucose metabolic processes via protein biosynthesis, estrogen and glucagon associated signaling pathways. These results revealed that miRNA-122 may be an indispensable biomarker for the diagnosis, prognostic evaluation and targeted therapy in pan-cancer.
Collapse
Affiliation(s)
- Meiyu Dai
- Department of Clinical Laboratory, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Limin Li
- Department of Clinical Laboratory, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Xue Qin
- Department of Clinical Laboratory, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
35
|
Corsetti G, Chen-Scarabelli C, Romano C, Pasini E, Dioguardi FS, Onorati F, Knight R, Patel H, Saravolatz L, Faggian G, Scarabelli TM. Autophagy and Oncosis/Necroptosis Are Enhanced in Cardiomyocytes from Heart Failure Patients. Med Sci Monit Basic Res 2019; 25:33-44. [PMID: 30713336 PMCID: PMC6373236 DOI: 10.12659/msmbr.913436] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background Although originally described as a survival mechanism, it is unknown whether and to what extent autophagy is implicated in the terminal stages of heart failure. Here, we studied magnitude and evolution of autophagy in patients with intractable heart failure. Material/Methods Myocardial samples were obtained from 22 patients with ischemic cardiomyopathy and idiopathic dilated cardiomyopathy who were undergoing cardiac transplantation. Hearts from 11 patients who died from non-cardiac causes were used as control samples. Autophagy was evaluated by immunostaining with a monoclonal microtubule associated protein light chain 3 (LC3)-II antibody, while the relationship of autophagy with apoptosis and oncosis was assessed by double staining with TUNEL (terminal deoxynucleotidyl transferase – mediated deoxyuridine triphosphate nick end labeling) assay and complement 9 (C9) immunological staining, respectively. In addition, several necroptotic markers, including RIP1 and RIP3 (receptor interacting protein kinase 1 and 3), anti-C3 (cleaved-caspase-3), and anti-NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) were assessed by immunohistochemistry. Results Anti-LC3-II staining was detected in 8.7±1.6% of the heart failure patient heart samples and in 1.2±0.3% of control patient heart samples. Vacuole formation started at one nuclear pole, before becoming bipolar and involving the cytosol. Subsequently, the autophagic process extended also to the nuclei, which underwent a progressive vacuolization and disintegration, assuming a peculiar “strawberry like appearance”. Myocytes with extensive vacuole formation exhibited nuclear degeneration, which was associated with TUNEL, C3, C9, RIP1, and RIP3 positive staining. Conversely, myocytes with less extensive vacuole formation showed RIP1 and NF-κB positive staining, though not positivity for other cell death markers. Conclusions Autophagy was extensively detected in end-stage heart failure and its progression, resulted in secondary cell death, with occurrence of oncosis and necroptosis exceeding that of apoptosis. Conversely, activation of the RIP1/NF-κB pathway was associated with cell survival.
Collapse
Affiliation(s)
- Giovanni Corsetti
- Division of Human Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Carol Chen-Scarabelli
- Center for Heart and Vessel Preclinical Studies, Department of Internal Medicine, St. John Hospital and Medical Center, Wayne State University, Detroit, MI, USA
| | - Claudia Romano
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Evasio Pasini
- Scientific Clinical Institutes Maugeri, Cardiac Rehabilitation Lumezzane Institute, Brescia, Italy
| | | | - Francesco Onorati
- Division of Cardiovascular Surgery, Verona University Hospital, Verona, Italy
| | - Richard Knight
- Medical Research Council (MRC) Toxicology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Hemang Patel
- General Medical Education, Department of Internal Medicine, Ascension St. John Hospital, Detroit, MI, USA
| | - Louis Saravolatz
- Department of Medicine, Ascension St John Hospital and Wayne State University School of Medicine, Detroit, MI, USA
| | - Giuseppe Faggian
- Division of Cardiovascular Surgery, Verona University Hospital, Verona, Italy
| | - Tiziano M Scarabelli
- Center for Heart and Vessel Preclinical Studies, Department of Internal Medicine, St. John Hospital and Medical Center, Wayne State University, Detroit, MI, USA
| |
Collapse
|
36
|
Di Ciaula A, Wang DQH, Portincasa P. Cholesterol cholelithiasis: part of a systemic metabolic disease, prone to primary prevention. Expert Rev Gastroenterol Hepatol 2019; 13:157-171. [PMID: 30791781 DOI: 10.1080/17474124.2019.1549988] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cholesterol gallstone disease have relationships with various conditions linked with insulin resistance, but also with heart disease, atherosclerosis, and cancer. These associations derive from mechanisms active at a local (i.e. gallbladder, bile) and a systemic level and are involved in inflammation, hormones, nuclear receptors, signaling molecules, epigenetic modulation of gene expression, and gut microbiota. Despite advanced knowledge of these pathways, the available therapeutic options for symptomatic gallstone patients remain limited. Therapy includes oral litholysis by the bile acid ursodeoxycholic acid (UDCA) in a small subgroup of patients at high risk of postdissolution recurrence, or laparoscopic cholecystectomy, which is the therapeutic radical gold standard treatment. Cholecystectomy, however, may not be a neutral event, and potentially generates health problems, including the metabolic syndrome. Areas covered: Several studies on risk factors and pathogenesis of cholesterol gallstone disease, acting at a systemic level have been reviewed through a PubMed search. Authors have focused on primary prevention and novel potential therapeutic strategies. Expert commentary: The ultimate goal appears to target the manageable systemic mechanisms responsible for gallstone occurrence, pointing to primary prevention measures. Changes must target lifestyles, as well as experimenting innovative pharmacological tools in subgroups of patients at high risk of developing gallstones.
Collapse
Affiliation(s)
- Agostino Di Ciaula
- a Division of Internal Medicine , Hospital of Bisceglie , Bisceglie , Italy
| | - David Q-H Wang
- b Department of Medicine, Division of Gastroenterology and Liver Diseases , Marion Bessin Liver Research Center, Albert Einstein College of Medicine , Bronx , NY , USA
| | - Piero Portincasa
- c Department of Biomedical Sciences and Human Oncology, Clinica Medica "A. Murri" , University of Bari Medical School , Bari , Italy
| |
Collapse
|
37
|
Li Q, Zhang Z, Li H, Pan X, Chen S, Cui Z, Ma J, Zhou Z, Xing B. Lycium barbarum polysaccharides protects H9c2 cells from hypoxia-induced injury by down-regulation of miR-122. Biomed Pharmacother 2019; 110:20-28. [DOI: 10.1016/j.biopha.2018.11.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 10/22/2018] [Accepted: 11/02/2018] [Indexed: 02/07/2023] Open
|
38
|
Serum miR-122 levels correlate with diabetic retinopathy. Clin Exp Med 2019; 19:255-260. [PMID: 30673918 DOI: 10.1007/s10238-019-00546-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Accepted: 01/16/2019] [Indexed: 12/15/2022]
Abstract
Diabetic retinopathy is the most severe ocular complication of diabetes and may lead to visual disability and blindness. Proliferative diabetic retinopathy (PDR) is characterized by ischemia-induced neovascularization with associated complications. An association was established between the presence of PDR, cardiovascular disease, and mortality among patients with type 1 diabetes mellitus and type 2 diabetes mellitus in epidemiological studies. However, the mechanism underlying increased cardiovascular risk in patients with PDR is still unknown. In recent years, a group of miRNAs has been linked to the pathology of diabetes mellitus. Besides, miRNAs in biofluids such as serum have been suggested as potential minimally invasive biomarkers of diabetes and vascular complications. This was a prospective study that recruited 40 human subjects: 10 healthy subjects, 10 with diabetes but without retinopathy (NDR), 10 with diabetic non-proliferative retinopathy (NPDR), and 10 with proliferative diabetic retinopathy (PDR). To examine whether serum miRNAs show altered levels at different stages of diabetic retinopathy, seven specific miRNA candidates (miR-126-3p, miR-130a-3p, miR-21-1, let-7f-5p, miR-122, miR-30c and miR-451a) were measured by qRT-PCR in RNA isolated from sera of all subjects. miR-122 levels increased in parallel with retinopathy severity: from healthy controls to NDR and from NDR to NPDR. However, when the disease progressed to PDR a marked decrease in miR-122 level was noted. This decrease was significant both compared to NPDR samples (p = 0.016) and to all non-PDR samples (p = 0.0002). Additionally, a positive trend was observed comparing miR-122 levels and the number of endothelial progenitor cells in the sera of all subjects. A significant increase in miR-122 was found in patients with diabetic retinopathy that may be related to its role in preventing angiogenesis and proliferation. The dramatic decline in patients with PDR may represent an inhibition or exhaustion of the anti-angiogenic anti-proliferative defense system. Further studies are needed to understand whether miRNA-122 has a role in the pathogenesis of diabetic retinopathy.
Collapse
|
39
|
Chen Z, Liu R, Niu Q, Wang H, Yang Z, Bao Y. Morphine Postconditioning alleviates autophage in ischemia-reperfusion induced cardiac injury through up-regulating lncRNA UCA1. Biomed Pharmacother 2018; 108:1357-1364. [DOI: 10.1016/j.biopha.2018.09.119] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 09/18/2018] [Accepted: 09/19/2018] [Indexed: 01/18/2023] Open
|
40
|
Chang L, Chai X, Chen P, Cao J, Xie H, Zhu J. miR-181b-5p suppresses starvation-induced cardiomyocyte autophagy by targeting Hspa5. Int J Mol Med 2018; 43:143-154. [PMID: 30431062 PMCID: PMC6257845 DOI: 10.3892/ijmm.2018.3988] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 10/16/2018] [Indexed: 12/25/2022] Open
Abstract
This study aimed to investigate the role of microRNA-181b-5p (miR-181b-5p) in starvation-induced cardiomyocyte autophagy by targeting heat shock protein family A member 5 (Hspa5). For this purpose, H9c2 cardiomyocytes and neonatal rat ventricular myocytes (NRVMs) were glucose-starved in Earle's Balanced Salt Solution (EBSS) for different periods of time (0, 2, 4, 6 and 8 h). RT-qPCR analysis was performed to examine the expression of miR-181b-5p in the different groups. Immunofluorescence was performed to detect the expression of LC3. In addition, the H9c2 cardiomyo-cytes and NRVMs were transfected with miR-181b-5p mimic, miR-181b-5p inhibitor, siHspa5 or their respective controls. An MTT assay was performed to measure cell proliferation in the different groups. Western blot analysis was performed to determine the expression of Beclin-1, Hspa5, phosphorylated phosphoinositide 3-kinase PI3K (p-PI3K), phosphorylated Akt (p-Akt), phosphorylated mammalian target of rapamycin (p-mTOR), Bcl-2, Bax and cleaved caspase-3. Flow cytometry was performed to assess cell apoptosis. A luciferase reporter assay was performed to determine whether Hspa5 is a direct target of miR-181b-5p. The results revealed that the down-regulation of miR-181b-5p promoted cell autophagy in the cardiomyocytes. Moreover, miR-181b-5p negatively regulated Beclin-1 and Hspa5. Beclin-1 is a well-known autophagy- and apoptosis-related protein. In addition, cell apoptosis was attenuated by the decreased expression of miR-181b-5p in the cardiomyocytes. Bcl-2 prevented apoptosis and autophagy by binding to Bax and Bcl-2, respectively. The upregulation of miR-181b-5p inhibited autophagy and promoted apoptosis via Hspa5. miR-181b-5p inhibition promoted p-mTOR, p-Akt and p-PI3K expression via Hspa5. The results of luciferase reporter assay also confirmed that Hspa5 is a direct target of miR-181b-5p. On the whole, the findings of this study suggest that miR-181b-5p contributes to starvation-induced autophagy and apoptosis in cardiomyocytes by directly targeting Hspa5 via the PI3K/Akt/mTOR signaling pathway.
Collapse
Affiliation(s)
- Liuhui Chang
- Department of Anesthesiology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Xiaoming Chai
- Department of Anesthesiology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Peiming Chen
- Department of Anesthesiology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Jianfang Cao
- Department of Anesthesiology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Hong Xie
- Department of Anesthesiology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Jiang Zhu
- Department of Anesthesiology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| |
Collapse
|
41
|
Wu S, Chang G, Gao L, Jiang D, Wang L, Li G, Luo X, Qin S, Guo X, Zhang D. Trimetazidine protects against myocardial ischemia/reperfusion injury by inhibiting excessive autophagy. J Mol Med (Berl) 2018; 96:791-806. [PMID: 29955901 DOI: 10.1007/s00109-018-1664-3] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 06/16/2018] [Accepted: 06/21/2018] [Indexed: 12/25/2022]
Abstract
Trimetazidine (TMZ) has been demonstrated to have protective effects against myocardial ischemia/reperfusion (MI/R) injury. In the present study, we investigated the effects and the underlying mechanisms of TMZ on autophagy during MI/R in vivo and in vitro. In the in vivo study, an animal model of MI/R was induced by coronary occlusion. TMZ (20 mg/kg/day) protected the rat hearts from MI/R-induced heart failure by increasing ejection fraction and fractional shortening and decreasing end-systolic volume, end-diastolic volume, left ventricular (LV) internal diameter at systole, and LV internal diameter at diastole; it alleviated myocardial injury and oxidative stress by decreasing LDH, creatine kinase MB isoenzyme, ROS, and MDA levels and increasing SOD and glutathione peroxidase levels in plasma. TMZ also reduced myocardial infarct size and apoptosis. Moreover, TMZ markedly inhibited MI/R-induced autophagy by decreasing the protein and messenger RNA levels of LC3-II, Beclin1, ATG5, and ATG7 and the number of autophagosomes and by involving the AKT/mTOR pathway. Further, in the in vitro experiments, H9c2 cells were incubated with TMZ (40 μM) to explore the direct effects of TMZ following exposure to hypoxia and reoxygenation (H/R). TMZ increased cell viability and the concentration of intracellular SOD and inhibited H/R-induced cell apoptosis and ROS production. Moreover, TMZ decreased the number of autophagosomes and autophagy-related protein expression; it also upregulated p-AKT and p-mTOR expression. In addition, TMZ augmented Bcl-2 protein expression and diminished Bax protein expression, the Bax/Bcl-2 rate, and cleaved caspase-3 level. However, these effects on H9c2 cells were notably abolished by the PI3K inhibitor LY294002. In conclusion, our results showed that TMZ inhibited I/R-induced excessive autophagy and apoptosis, which was, at least partly, mediated by activating the AKT/mTOR pathway. KEY MESSAGES TMZ improved cardiac function, alleviated myocardial injury and oxidative stress, and reduced the myocardial infarct area and apoptosis. TMZ inhibited MI/R-induced myocardial autophagy, H/R-induced H9c2 cell apoptosis, and autophagy flux. The effect of TMZ on autophagy was repressed by LY294002. TMZ protected against MI/R injury by inhibiting excessive autophagy via activating the AKT/mTOR pathway.
Collapse
Affiliation(s)
- Shiyong Wu
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Guanglei Chang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Lei Gao
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Dan Jiang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Liyou Wang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Guoxing Li
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xuexiu Luo
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Shu Qin
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xueli Guo
- Department of Vascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan Province, China
| | - Dongying Zhang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
42
|
Liu J, Jiang M, Deng S, Lu J, Huang H, Zhang Y, Gong P, Shen X, Ruan H, Jin M, Wang H. miR-93-5p-Containing Exosomes Treatment Attenuates Acute Myocardial Infarction-Induced Myocardial Damage. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 11:103-115. [PMID: 29858047 PMCID: PMC5852413 DOI: 10.1016/j.omtn.2018.01.010] [Citation(s) in RCA: 148] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 01/23/2018] [Accepted: 01/24/2018] [Indexed: 02/07/2023]
Abstract
Adipose-derived stromal cells (ADSCs) have been considered as an attractive therapeutic tool. Accumulating evidence indicates that the healing effects of ADSCs are mainly related to paracrine action rather than transdifferentiation. Data show that the expression of miR-93-5p has a cardio-protective effect after acute myocardial infarction (AMI). To identify whether miR-93-5p-encapsulating exosomes that form ADSCs have a better cardio-protective effect, we investigated the inflammatory factors and miR-30d-5p expression in clinical levels. A rat model of AMI and an in vitro model of hypoxic H9c2 cells were established to study the protective mechanism of miR-93-5p in ischemia-induced cardiac injury. The results show that the expression of inflammatory cytokines and miR-93-5p were increased following AMI in both patients and animal models. Moreover, treatment with ADSC-derived miR-93-5p-containing exosomes has a greater protective effect on infarction-induced myocardial damage than simple exosome processing. Furthermore, in vitro experiments confirmed that the expression of miR-93-5p can significantly suppress hypoxia-induced autophagy and inflammatory cytokine expression by targeting Atg7 and Toll-like receptor 4 (TLR4), respectively, and was confirmed with Atg7 or TLR4 overexpression. The results also show that autophagy activation can promote inflammatory cytokine expression indirectly. Taken together, these results suggest that the miR-93-5p-enhanced ADSC-derived exosomes prevent cardiac injury by inhibiting autophagy and the inflammatory response.
Collapse
Affiliation(s)
- Jiwen Liu
- Department of Cardiology, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai 200135, China
| | - Mei Jiang
- Department of Neurology, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai 200135, China
| | - Shengqiong Deng
- Department of Clinical Laboratory, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai 200135, China
| | - Jide Lu
- Department of Cardiology, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai 200135, China
| | - Hui Huang
- Department of Cardiology, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai 200135, China
| | - Yu Zhang
- Department of Cardiology, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai 200135, China
| | - Peihua Gong
- Department of Cardiology, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai 200135, China
| | - Xumin Shen
- Department of Cardiology, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai 200135, China
| | - Huanjun Ruan
- Department of Cardiology, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai 200135, China
| | - Mingming Jin
- Department of Central Laboratory, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai 200135, China.
| | - Hairong Wang
- Department of Cardiology, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai 200135, China.
| |
Collapse
|
43
|
Feng L, Liu W, Yang J, Wang Q, Wen S. Effect of Hexadecyl Azelaoyl Phosphatidylcholine on Cardiomyocyte Apoptosis in Myocardial Ischemia-Reperfusion Injury: A Hypothesis. Med Sci Monit 2018; 24:2661-2667. [PMID: 29706617 PMCID: PMC5949054 DOI: 10.12659/msm.907578] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Reperfusion after myocardial ischemia can induce cardiomyocyte death, known as myocardial reperfusion injury. The pathophysiology of the process of reperfusion suggests the confluence multiple pathways. Recent studies have focused on the inflammatory response, which is considered to be the main mechanism during the process of myocardial ischemia-reperfusion injury and can cause cardiomyocyte apoptosis. Peroxisome proliferator-activated receptors gamma activated by endogenous ligands and exogenous ligand can decrease the inflammatory response in cardiomyocytes. Thiazolidinediones are synthetic, high-affinity, selective ligands for peroxisome proliferator-activated receptors gamma, and can inhibit the inflammatory response, decrease myocardial infarct size, and protect cardiac function. However, thiazolidinediones, including rosiglitazone and pioglitazone, can also contribute to adverse cardiovascular events such as congestive heart failure. Therefore, there are some limitations to the use of thiazolidinediones. Most endogenous ligands were of low affinity until hexadecyl azelaoyl phosphatidylcholine was identified as a high-affinity ligand and agonist for peroxisome proliferator-activated receptors gamma. Hexadecyl azelaoyl phosphatidylcholine binds recombinant peroxisome proliferator-activated receptors with an affinity (Kd(app) ≈40 nM) which is equivalent to rosiglitazone. Therefore, hexadecyl azelaoyl phosphatidylcholine is a specific peroxisome proliferator-activated receptors gamma agonist. Given these findings, we hypothesized that the use of hexadecyl azelaoyl phosphatidylcholine can activate the peroxisome proliferator-activated receptors gamma signal pathways and prevent the inflammatory response process of myocardial ischemia-reperfusion injury, with reduced cardiomyocyte apoptosis and death.
Collapse
Affiliation(s)
- Limin Feng
- Department of Cardiology, The Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China (mainland)
| | - Wennan Liu
- Department of Cardiology, Tianjin Medical University General Hospital, Tianjin, China (mainland)
| | - Jianzhou Yang
- Department of Preventive Medicine, Changzhi Medical College, Changzhi, Shanxi, China (mainland)
| | - Qing Wang
- Department of Cardiology, Tianjin Medical University General Hospital, Tianjin, China (mainland)
| | - Shiwu Wen
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, Hunan, China (mainland)
| |
Collapse
|
44
|
Wang X, Yang C, Liu X, Yang P. The impact of microRNA-122 and its target gene Sestrin-2 on the protective effect of ghrelin in angiotensin II-induced cardiomyocyte apoptosis. RSC Adv 2018; 8:10107-10114. [PMID: 35540851 PMCID: PMC9078835 DOI: 10.1039/c7ra13028g] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 03/06/2018] [Indexed: 12/15/2022] Open
Abstract
Ghrelin with n-octanoylated serine 3 residue is a peptide hormone with well-known cardioprotective properties. MicroRNA-122 is associated with the pathogenesis of many cardiovascular diseases, including apoptosis and was found highly increased in our previous rat model of post-myocardial infarction heart failure. In this study, we aimed to identify the target gene of microRNA-122 and to evaluate their impacts on the protective effect of acylated ghrelin in angiotensin II-induced apoptosis. The results showed that microRNA-122 was upregulated in the angiotensin II administration group accompanied by increased cell apoptosis, which were both reversed by ghrelin. Furthermore, microRNA-122 mimics upregulated numerous pro-apoptotic genes and increased apoptosis. The luciferase activity assay revealed Sestrin-2 as a direct target of microRNA-122. The expression of Sestrin-2 was downregulated by angiotensin II and upregulated by co-treatment with ghrelin. Inhibition of microRNA-122 and overexpression of Sestrin-2 alleviated apoptosis which was further reduced upon administered of ghrelin. Together, these results indicated that Sestrin-2 expression is inhibited by microRNA-122 and that this inhibition is involved in the protective effect of ghrelin and angiotensin II-induced apoptosis. We also found that microRNA-122 influenced several apoptosis pathways including the caspase cascade reaction and death receptor-mediated pathways. Collectively, our data reveal that microRNA-122 and its target gene Sestrin-2, under the regulation of angiotensin II and ghrelin, are important players in cardiomyocyte apoptosis. We therefore believe that microRNA-122 and Sestrin-2 can be developed as potential therapeutic targets against apoptosis in cardiovascular diseases. Inhibition of microRNA-122 and overexpression of Sestrin-2 alleviated angiotensin II-induced cardiomyocyte apoptosis and enhanced the protective effect of ghrelin.![]()
Collapse
Affiliation(s)
- Xiaotong Wang
- The Department of Cardiology, China-Japan Union Hospital, Jilin University Changchun 130011 Jilin China
| | - Chunyan Yang
- The Department of Cardiology, China-Japan Union Hospital, Jilin University Changchun 130011 Jilin China
| | - Xueyan Liu
- The Department of Cardiology, China-Japan Union Hospital, Jilin University Changchun 130011 Jilin China
| | - Ping Yang
- The Department of Cardiology, China-Japan Union Hospital, Jilin University Changchun 130011 Jilin China
| |
Collapse
|