1
|
Date S, Bhatt LK. Targeting high-mobility-group-box-1-mediated inflammation: a promising therapeutic approach for myocardial infarction. Inflammopharmacology 2025; 33:767-784. [PMID: 39487941 DOI: 10.1007/s10787-024-01586-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 10/11/2024] [Indexed: 11/04/2024]
Abstract
Myocardial ischemia, resulting from coronary artery blockage, precipitates cardiac arrhythmias, myocardial structural changes, and heart failure. The pathophysiology of MI is mainly based on inflammation and cell death, which are essential in aggravating myocardial ischemia and reperfusion injury. Emerging research highlights the functionality of high mobility group box-1, a non-histone nucleoprotein functioning as a chromosomal stabilizer and inflammatory mediator. HMGB1's release into the extracellular compartment during ischemia acts as damage-associated molecular pattern, triggering immune reaction by pattern recognition receptors and exacerbating tissue inflammation. Its involvement in signaling pathways like PI3K/Akt, TLR4/NF-κB, and RAGE/HMGB1 underscores its significance in promoting angiogenesis, apoptosis, and reducing inflammation, which is crucial for MI treatment strategies. This review highlights the complex function of HMGB1 in the pathogenesis of myocardial infarction by summarizing novel findings on the protein in ischemic situations. Understanding the mechanisms underlying HMGB1 could widen the way to specific treatments that minimize the severity of MI and enhance patient outcomes.
Collapse
Affiliation(s)
- Shrutika Date
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai, India
| | - Lokesh Kumar Bhatt
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai, India.
| |
Collapse
|
2
|
Lehto P, Skarp S, Saukko T, Säkkinen H, Syrjälä H, Kerkelä R, Saarimäki S, Bläuer S, Porvari K, Pakanen L, Karhu J, Ala-Kokko T. Postmortem analyses of myocardial microRNA expression in sepsis. Sci Rep 2024; 14:29476. [PMID: 39604475 PMCID: PMC11603066 DOI: 10.1038/s41598-024-81114-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 11/25/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Sepsis can lead to myocardial depression, playing a significant role in sepsis pathophysiology, clinical care, and outcome. To gain more insight into the pathophysiology of the myocardial response in sepsis, we investigated the expression of microRNA in myocardial autopsy specimens in critically ill deceased with sepsis and non-septic controls. MATERIALS AND METHODS In this retrospective observational study, we obtained myocardial tissue samples collected during autopsy from adult patients deceased with sepsis (n = 15) for routine histological examination. We obtained control myocardial tissue specimens (n = 15) from medicolegal autopsies of cadavers whose cause of death was injury or who were found dead at home and the cause of death was coronary artery disease with sudden cardiac arrest. RNA was isolated from formalin-fixed paraffin- embedded (FFPE) cardiac samples using the RecoverAll Total Nucleic Acid Isolation Kit for FFPE (Invitrogen). Differentially expressed miRNAs were identified using edgeR v3.32. MicroRNA was considered up- or down-regulated if the false discovery rate was < 0.05 and logarithmic fold change (log2FC) ≥ 1 for up-regulated or log2FC ≤ -1 for down-regulated miRNAs. The mean difference and 95% confidence interval (CI) were calculated for normalized read counts. Predicted miRNA targets were retrieved using Ingenuity Pathway Analysis (IPA) software, and pathway enrichment and classification were performed using PantherDB. For miRNA - mRNA interaction analysis, differentially expressed genes were analyzed by 3`mRNA sequencing. RESULTS Differential expression analysis identified a total of 32 miRNAs in the myocardial specimens. Eight miRNAs had a significant change in the mean difference based on the 95% CI, with the largest increase in mean counts in septic samples with hsa-miR-12136 and the highest fold change with hsa-miR-146b-5p. The threshold for down-regulated miRNAs in sepsis compared to controls was obtained with hsa-miR-144-5p and hsa-miR-451a, with the latter having the largest decrease in mean counts and fold decrease. The miRNA - mRNA interaction analysis identified eight miRNAs with target genes also differentially expressed in septic hearts. The highest number of potential targets were identified for hsa-miR-363-3p. CONCLUSIONS Several regulatory miRNAs were up-or down-regulated in the myocardial tissue of patients deceased with sepsis compared to non-septic subjects. The predicted target genes of miRNAs and miRNA-mRNA interaction analysis are associated with biological functions related to cardiovascular functions, cell viability, cell adhesion, and regulation of inflammatory and immune response.
Collapse
Affiliation(s)
- Pasi Lehto
- Research Group of Intensive Care Medicine, Intensive Care Centre, Oulu University Hospital, University of Oulu and Medical Research Center (MRC), PO BOX 29, 90029, Oulu, Finland.
| | - Sini Skarp
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, Oulu, Finland
- Medical Research Centre Oulu, Biocenter Oulu, Oulu University Hospital and University of Oulu, University of Oulu, Oulu, Finland
| | - Taru Saukko
- Research Group of Intensive Care Medicine, Intensive Care Centre, Oulu University Hospital, University of Oulu and Medical Research Center (MRC), PO BOX 29, 90029, Oulu, Finland
| | - Hanna Säkkinen
- Research Group of Intensive Care Medicine, Intensive Care Centre, Oulu University Hospital, University of Oulu and Medical Research Center (MRC), PO BOX 29, 90029, Oulu, Finland
| | - Hannu Syrjälä
- Research Group of Intensive Care Medicine, Intensive Care Centre, Oulu University Hospital, University of Oulu and Medical Research Center (MRC), PO BOX 29, 90029, Oulu, Finland
| | - Risto Kerkelä
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, Oulu, Finland
- Medical Research Centre Oulu, Biocenter Oulu, Oulu University Hospital and University of Oulu, University of Oulu, Oulu, Finland
| | - Samu Saarimäki
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, Oulu, Finland
- Medical Research Centre Oulu, Biocenter Oulu, Oulu University Hospital and University of Oulu, University of Oulu, Oulu, Finland
| | - Sonja Bläuer
- Research Group of Intensive Care Medicine, Intensive Care Centre, Oulu University Hospital, University of Oulu and Medical Research Center (MRC), PO BOX 29, 90029, Oulu, Finland
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, Oulu, Finland
- Medical Research Centre Oulu, Biocenter Oulu, Oulu University Hospital and University of Oulu, University of Oulu, Oulu, Finland
| | - Katja Porvari
- Department of Forensic Medicine, Research Unit of Internal Medicine, Medical Research Center Oulu, University of Oulu, PO Box 5000, 90014, Oulu, Finland
| | - Lasse Pakanen
- Department of Forensic Medicine, Research Unit of Internal Medicine, Medical Research Center Oulu, University of Oulu, PO Box 5000, 90014, Oulu, Finland
- Forensic Medicine Unit, Finnish Institute for Health and Welfare, Oulu, Finland
| | - Jaana Karhu
- Research Group of Intensive Care Medicine, Intensive Care Centre, Oulu University Hospital, University of Oulu and Medical Research Center (MRC), PO BOX 29, 90029, Oulu, Finland
| | - Tero Ala-Kokko
- Research Group of Intensive Care Medicine, Intensive Care Centre, Oulu University Hospital, University of Oulu and Medical Research Center (MRC), PO BOX 29, 90029, Oulu, Finland
| |
Collapse
|
3
|
Hosoya T, Harada K, Kanetake J. β stimulator induces upregulation of miR-27a in the rat heart one hour after the injection. Leg Med (Tokyo) 2024; 70:102475. [PMID: 38924970 DOI: 10.1016/j.legalmed.2024.102475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/10/2024] [Accepted: 06/22/2024] [Indexed: 06/28/2024]
Abstract
MicroRNAs (miRs) are non-coding small RNA containing 18 to 22 nucleotides, that post-transcriptionally regulates mRNA expression. Chronic injection of β stimulator is known to induce cardiac injury and change of miRs expression level in the heart with some pathological changes such as fibrosis, heart failure, myocardial infarction. We investigated the changes in the expression level of miRs in the rat heart one hour after isoproterenol (a β stimulator) injection. Male Sprague-Dawley rats were assigned into three groups and received subcutaneous injection of normal sarin (NS) or 0.1 mg/kg isoproterenol (ISO-0.1) or 10 mg/kg isoproterenol (ISO-10). After one hour, we collected their heart and plasma. Total RNA was extracted from the left ventricle and used for deep miRNA sequencing. Based on the results of miRNA sequencing, we performed real-time polymerase chain reaction (RT-PCR) using 8 miR primers. Cardiac injury was evaluated by hematoxylin and eosin, and phosphotungstic acid-hematoxylin staining and measuring troponin-I levels in plasma. Troponin-I was significantly increased in ISO-0.1 and ISO-10 groups, but histological observation did not show any cardiac necrosis. miRNA sequencing identified 14 upregulated miRs and 12 downregulated miRs. Of the 26 miRs, RT-PCR confirmed miR-144-3p/5p and miR-451-5p were decreased, and that 5 miRs (miR-27a-5p, miR-30b-3p, miR-92a-1-5p, miR-132-5p, miR-582-3p) were upregulated. This study showed that β stimulus causes downregulation of miR-144/451 cluster and increases expression of five 5 miRs in the heart, especially 6.5-fold upregulation of miR-27a-5p as early as one hour after isoproterenol injection. Therefore, these miRs might be good biomarkers for cardiac injury.
Collapse
Affiliation(s)
- Tadashi Hosoya
- Department of Forensic Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan.
| | - Kazuki Harada
- Department of Forensic Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan; Department of Legal Medicine, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Jun Kanetake
- Department of Forensic Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan
| |
Collapse
|
4
|
Gao Y, Gao Y, Niu Z, Liu J, Feng H, Sun J, Wang L, Pan L. CCCTC-binding factor-mediated microRNA-340-5p suppression aggravates myocardial injury in rats with severe acute pancreatitis through activation of the HMGB1/TLR4 axis. Immunopharmacol Immunotoxicol 2022; 44:306-315. [PMID: 35238277 DOI: 10.1080/08923973.2022.2043898] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 02/13/2022] [Indexed: 02/08/2023]
Abstract
BACKGROUND Severe acute pancreatitis (SAP) is a life-threatening disorder associated with multisystem organ failure. This study aimed to investigate the function of high mobility group box 1 (HMGB1) in SAP-induced myocardial injury. METHODS A rat model with SAP was induced. The pathological changes in rat pancreatic and cardiac tissues were examined by HE staining. Cardiomyocyte apoptosis in rat cardiac tissues, and the serum levels of myocardial injury markers and pro-inflammatory cytokines were examined. Rat primary cardiomyocytes were treated with H2O2 for in vitro experiments. The regulatory molecules of HMGB1 were predicted by bioinformatics analysis. Altered expression of HMGB1, microRNA (miR)-340-5p and CCCTC-binding factor (CTCF) was introduced in rats or cells to investigate their roles in myocardial injury. RESULTS CTCF and HMGB1 were highly expressed but miR-340-5p was poorly expressed in cardiac tissues of rats with SAP. HMGB1 silencing reduced toll-like receptor 4 (TLR4) expression to promote proliferation and reduce apoptosis of H2O2-treated cardiomyocytes. miR-340-5p targeted HMGB1 mRNA, while CTCF suppressed miR-340-5p transcription. CTCF upregulation or miR-340-5p downregulation blocked the effects of HMGB1 silencing on cardiomyocytes. In vivo, CTCF silencing alleviated injury in rat pancreatic and cardiac tissues and reduced the expression of creatine kinase-MB (CK-MB), lactic dehydrogenase, interleukin (IL)-1β, IL-6 and tumor necrosis factor-α (TNF-α) in rat serum. But further overexpression of HMGB1 or inhibition of miR-340-5p aggravated the symptoms in rats. CONCLUSION This study demonstrated that CTCF reduces transcription of miR-340-5p to promote HMGB1 expression, which activates TLR4 expression and promotes myocardial injury in rats with SAP.
Collapse
Affiliation(s)
- Yazhou Gao
- Department of Emergency Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Yanxia Gao
- Department of Emergency Medicine, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Zequn Niu
- Department of Emergency Medicine, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Jie Liu
- Department of Emergency Medicine, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Hui Feng
- Department of Emergency Medicine, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Jiangli Sun
- Department of Emergency Medicine, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Liming Wang
- Department of Emergency Medicine, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Longfei Pan
- Department of Emergency Medicine, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| |
Collapse
|
5
|
Jia S, He D, Liang X, Cheng P, Liu J, Chen M, Wang C, Zhang H, Meng C. Corilagin induces apoptosis and inhibits autophagy of HL‑60 cells by regulating miR‑451/HMGB1 axis. Mol Med Rep 2021; 25:34. [PMID: 34850958 PMCID: PMC8669704 DOI: 10.3892/mmr.2021.12550] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 08/12/2021] [Indexed: 11/17/2022] Open
Abstract
Corilagin is the primary active component of the Euphorbia phyllanthus plant and has significant anti-cancer properties. However, the biological effects and mechanisms of corilagin on acute myeloid leukemia (AML) have not been clarified. The Cell Counting Kit-8 and Carboxyfluorescein Diacetate Succinimidyl Ester assay results showed that corilagin significantly inhibited proliferation of the AML cell line HL-60 in a time- and dose-dependent manner. Western blotting and flow cytometry analysis were performed to determine the levels of apoptosis in HL-60 cells. The protein levels of cleaved caspase-3 and Bak were upregulated, while Bcl-xl was downregulated in cells treated with corilagin. The percentage of early- and late-stage apoptotic cells increased following corilagin treatment in a dose-dependent manner, indicating that the intrinsic mitochondrial apoptosis pathway was activated by corilagin. Simultaneously, western blotting and immunofluorescence results revealed that autophagy was suppressed; this was accompanied by a decrease in light chain 3-II (LC3-II) conversion and autophagosomes. MicroRNA (miRNA/miR) profile analysis showed that corilagin elevated the expression of the tumor suppressor miR-451, while the mRNA and protein levels of high mobility group protein B1 (HMGB1), the target of miR-451, decreased following exposure to corilagin. Knockdown of miR-451 decreased the downregulation of HMGB1 caused by corilagin, indicating negative regulation of HMGB1 by miR-451 during corilagin treatment. Furthermore, knockdown of miR-451 also attenuated corilagin-induced proliferation inhibition of HL-60 cells, implying that miR-451 was essential for the proliferation inhibitory effect of corilagin. In conclusion, these results indicated that corilagin induced apoptosis and inhibited autophagy in HL-60 cells by regulating the miR-451/HMGB1 axis, and corilagin may be a novel therapeutic drug for the treatment of AML.
Collapse
Affiliation(s)
- Shu Jia
- Medical Research Center, Affiliated Hospital of Jining Medical University, Jining, Shandong 272029, P.R. China
| | - Dongye He
- Medical Research Center, Affiliated Hospital of Jining Medical University, Jining, Shandong 272029, P.R. China
| | - Xiao Liang
- Department of Spine Surgery, Affiliated Hospital of Jining Medical University, Jining, Shandong 272029, P.R. China
| | - Panpan Cheng
- Department of Hematology, Affiliated Hospital of Jining Medical University, Jining, Shandong 272029, P.R. China
| | - Jilan Liu
- Medical Research Center, Affiliated Hospital of Jining Medical University, Jining, Shandong 272029, P.R. China
| | - Mingtai Chen
- Medical Research Center, Affiliated Hospital of Jining Medical University, Jining, Shandong 272029, P.R. China
| | - Cuiling Wang
- Department of Hematology, Affiliated Hospital of Jining Medical University, Jining, Shandong 272029, P.R. China
| | - Hao Zhang
- Department of Hematology, Affiliated Hospital of Jining Medical University, Jining, Shandong 272029, P.R. China
| | - Chunyang Meng
- Department of Spine Surgery, Affiliated Hospital of Jining Medical University, Jining, Shandong 272029, P.R. China
| |
Collapse
|
6
|
Zhang D, Wang Q, Qiu X, Chen Y, Yang X, Guan Y. Remifentanil protects heart from myocardial ischaemia/reperfusion (I/R) injury via miR-206-3p/TLR4/NF-κB signalling axis. J Pharm Pharmacol 2021; 74:282-291. [PMID: 34850055 DOI: 10.1093/jpp/rgab151] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 10/15/2021] [Indexed: 01/30/2023]
Abstract
OBJECTIVES Myocardial I/R injury is one of the most serious complications after reperfusion therapy in patients with myocardial infarction. Remifentanil has been found to protect the heart against I/R injury. However, its underlying mechanism remains uncertain in myocardial I/R injury. METHODS The myocardial I/R injury rat model was established by 30 min of ischaemia followed by 24 h of reperfusion. The animal model was evaluated by the levels of TC, ALT and AST and H&E staining. The binding of miR-206-3p and TLR4 was predicted and verified using TargetScan software, luciferase reporter and RNA pull-down assays. The functional role and mechanism of remifentanil were identified by ultrasonic echocardiography, oxidative stress markers, H&E, Masson and TUNEL staining and western blot. KEY FINDINGS The rat myocardial I/R injury model displayed a significantly high level of TC, ALT, AST, TLR4, p-IκBα and p-p65 and the presence of disorganized cells and inflammatory cell infiltration. The model also showed increased levels of LVEDD, LVESD, MDA, fibrosis and apoptosis and decreased levels of EF, FS, SOD and GSH, which were reversed with remifentanil treatment. Knockdown of miR-206-3p damaged cardiac function and aggravated oxidative stress. miR-206-3p could directly bind to TLR4. TLR4 overexpression destroyed cardiac function, exacerbated oxidative stress, increased levels of p-IκBα and p-p65 and aggravated pathology manifestation affected by remifentanil. CONCLUSIONS Our results elucidated that remifentanil alleviated myocardial I/R injury by miR-206-3p/TLR4/NF-κB signalling axis.
Collapse
Affiliation(s)
- Dongyun Zhang
- Department of Anesthesiology, Binhaiwan Central Hospital of Dongguan, Dongguan City, China
| | - Qun Wang
- Department of Anesthesiology, Binhaiwan Central Hospital of Dongguan, Dongguan City, China
| | - Xunbin Qiu
- Department of Anesthesiology, Binhaiwan Central Hospital of Dongguan, Dongguan City, China
| | - Yiguan Chen
- School of Medicine, Jinan University, Guangzhou City, China
| | - Xiaoli Yang
- Department of Anesthesiology, Binhaiwan Central Hospital of Dongguan, Dongguan City, China
| | - Yujian Guan
- Department of Anesthesiology, Binhaiwan Central Hospital of Dongguan, Dongguan City, China
| |
Collapse
|
7
|
Wang S, Cheng Z, Chen X, Lu G, Zhu X, Xu G. CircUBXN7 mitigates H/R-induced cell apoptosis and inflammatory response through the miR-622-MCL1 axis. Am J Transl Res 2021; 13:8711-8727. [PMID: 34539989 PMCID: PMC8430130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 06/09/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Hypoxia/reoxygenation (H/R)-mediated apoptosis and inflammation are major causes of tissue injury in acute myocardial infarction (AMI). Exploring the underlying mechanisms of cardiomyocyte injury induced by H/R is important for AMI treatment. Circular RNAs have been demonstrated to paly vital roles in the pathogenesis of AMI. Our study aimed to explore the function of circular RNA UBXN7 (circUBXN7) in regulating H/R-induced cardiomyocyte injury. METHODS H/R-treated H9c2 cells and a mouse model of AMI were used to investigate the function of circUBXN7 in H/R damage and AMI. The expressions of circUNXN7, miR-622 and MCL1 were analyzed by RT-qPCR. CCK-8 was used for examining cell viability. Cell apoptosis was evaluated with caspase 3 activity and Annexin V/PI staining. MCL1, Bax, Bcl-2 and cleaved-caspase 3 were examined with western blot. ELISA was used to examine the secretion of IL-6, TNF-α and IL-1β. RESULTS CircUBXN7 was downregulated in patients and mice with AMI, as well as in H/R-treated cells. Overexpression of circUBXN7 mitigated H/R-mediated apoptosis and secretion of inflammatory factors including IL-6, TNF-α and IL-1β. CircUBXN7 suppressed cell apoptosis and inflammatory reaction induced by H/R via targeting miR-622. MiR-622 targeted MCL1 to restrain its expression in H9c2 cells. Knockdown of MCL1 abrogated circUBXN7-mediated alleviation of apoptosis and inflammation after H/R treatment. CONCLUSION CircUBXN7 mitigates cardiomyocyte apoptosis and inflammatory reaction in H/R injury by targeting miR-622 and maintaining MCL1 expression. Our study provides novel potential therapeutic targets for AMI treatment.
Collapse
Affiliation(s)
- Sheng Wang
- Department of Cardiovascular Surgery, Fuwai Central China Cardiovascular Hospital, Heart Center of He'nan Provincial People's Hospital Zhengzhou, He'nan Province, China
| | - Zhaoyun Cheng
- Department of Cardiovascular Surgery, Fuwai Central China Cardiovascular Hospital, Heart Center of He'nan Provincial People's Hospital Zhengzhou, He'nan Province, China
| | - Xianjie Chen
- Department of Cardiovascular Surgery, Fuwai Central China Cardiovascular Hospital, Heart Center of He'nan Provincial People's Hospital Zhengzhou, He'nan Province, China
| | - Guoqing Lu
- Department of Cardiovascular Surgery, Fuwai Central China Cardiovascular Hospital, Heart Center of He'nan Provincial People's Hospital Zhengzhou, He'nan Province, China
| | - Xiliang Zhu
- Department of Cardiovascular Surgery, Fuwai Central China Cardiovascular Hospital, Heart Center of He'nan Provincial People's Hospital Zhengzhou, He'nan Province, China
| | - Gaojun Xu
- Department of Cardiovascular Surgery, Fuwai Central China Cardiovascular Hospital, Heart Center of He'nan Provincial People's Hospital Zhengzhou, He'nan Province, China
| |
Collapse
|
8
|
Moukette B, Barupala NP, Aonuma T, Sepulveda M, Kawaguchi S, Kim IM. Interactions between noncoding RNAs as epigenetic regulatory mechanisms in cardiovascular diseases. Methods Cell Biol 2021; 166:309-348. [PMID: 34752338 DOI: 10.1016/bs.mcb.2021.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Cardiovascular diseases (CVDs) represent the foremost cause of mortality in the United States and worldwide. It is estimated that CVDs account for approximately 17.8 million deaths each year. Despite the advances made in understanding cellular mechanisms and gene mutations governing the pathophysiology of CVDs, they remain a significant cause of mortality and morbidity. A major segment of mammalian genomes encodes for genes that are not further translated into proteins. The roles of the majority of such noncoding ribonucleic acids (RNAs) have been puzzling for a long time. However, it is becoming increasingly clear that noncoding RNAs (ncRNAs) are dynamically expressed in different cell types and have a comprehensive selection of regulatory roles at almost every step involved in DNAs, RNAs and proteins. Indeed, ncRNAs regulate gene expression through epigenetic interactions, through direct binding to target sequences, or by acting as competing endogenous RNAs. The profusion of ncRNAs in the cardiovascular system suggests that they may modulate complex regulatory networks that govern cardiac physiology and pathology. In this review, we summarize various functions of ncRNAs and highlight the recent literature on interactions between ncRNAs with an emphasis on cardiovascular disease regulation. Furthermore, as the broad-spectrum of ncRNAs potentially establishes new avenues for therapeutic development targeting CVDs, we discuss the innovative prospects of ncRNAs as therapeutic targets for CVDs.
Collapse
Affiliation(s)
- Bruno Moukette
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Nipuni P Barupala
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Tatsuya Aonuma
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Marisa Sepulveda
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Satoshi Kawaguchi
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Il-Man Kim
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, United States; Krannert Institute of Cardiology, Indiana University School of Medicine, Indianapolis, IN, United States; Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States.
| |
Collapse
|
9
|
Mo Y, Wu H, Zheng X, Xu L, Liu L, Liu Z. LncRNA CHRF aggravates myocardial ischemia/reperfusion injury by enhancing autophagy via modulation of the miR-182-5p/ATG7 pathway. J Biochem Mol Toxicol 2021; 35:e22709. [PMID: 33491285 DOI: 10.1002/jbt.22709] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 12/01/2020] [Accepted: 01/08/2021] [Indexed: 11/08/2022]
Abstract
Myocardial ischemia/reperfusion (I/R) injury is a very frequent cardiovascular disease and one of the leading causes of death. Abundant evidence has shown that long noncoding RNAs (lncRNAs) are crucial players in myocardial I/R injury. LncRNA cardiac hypertrophy-related factor (CHRF) has been revealed as an important modulator in cardiac disease. However, the function of CHRF in myocardial I/R injury is unclear. In our current work, we found that the expression of CHRF was upregulated in myocardial I/R injury models. Suppression of CHRF relieved myocardial I/R injury in vivo. In addition, in vitro silencing of CHRF enhanced cell viability and attenuated lactate dehydrogenase activity (LDH) as well as apoptosis in H9C2 cells treated with hypoxia/reoxygenation injury. Autophagy has been studied to play an important role in myocardial I/R injury. Thus, experiments related to autophagy were done, and the results showed that CHRF knockdown decreased autophagy. For the exploration of the regulatory mechanism, we found that CHRF sequestered and negatively regulated miR-182-5p to release its inhibition on ATG7. Findings from rescue assays revealed that ATG7 overexpression could suppress the effects of CHRF silence on cell viability, LDH level, apoptosis, and autophagy. To sum up, our results suggested that CHRF exacerbated myocardial I/R injury by enhancing autophagy via modulation of the miR-182-5p/ATG7 pathway. Therefore, this competing endogenous RNA axis may be a potential therapeutic biomarker for myocardial I/R injury.
Collapse
Affiliation(s)
- Yipeng Mo
- Department of Cardiovascular Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Hairuo Wu
- Department of Cardiovascular Medicine, Lianyungang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Lianyungang, Jiangsu Province, China
| | - Xiaojun Zheng
- Department of Cardiovascular Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Lin Xu
- Department of Cardiovascular Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Liangliang Liu
- Department of Cardiovascular Medicine, Lianyungang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Lianyungang, Jiangsu Province, China
| | - Zhen Liu
- Department of Cardiovascular Medicine, Lianyungang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Lianyungang, Jiangsu Province, China
| |
Collapse
|