1
|
Diagnostic and Prognostic Significance of microRNA-208a in Acute Myocardial Infarction. DISEASE MARKERS 2022; 2022:7030722. [PMID: 35607440 PMCID: PMC9124095 DOI: 10.1155/2022/7030722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 03/31/2022] [Accepted: 04/04/2022] [Indexed: 11/25/2022]
Abstract
Objective To determine the prognostic and diagnostic significance of microRNA-208a (miR-208a) in acute myocardial infarction (AMI). Methods Totally, 84 AMI patients hospitalized in our hospital between Jan. 2019 and Feb. 2021 were enrolled as the patient group (Pat group), and 50 healthy individuals over same time span as the control group (Con group). qRT-PCR assay was carried out to quantify serum miR-208a in the patients and receiver-operating characteristic (ROC) curves for analysing its diagnostic value in AMI patients and its predictive value in clinical efficacy and adverse events in the patients after therapy. The changes of miR-208a and clinical indexes ((lactate dehydrogenase (LDH), creatine kinase (CK) as well as Creatine kinase-MB (CK-MB)) in the patients before and after therapy were evaluated. Pearson's test was adopted to analyse the associations of miR-208a with clinical indexes. Additionally, the target genes of miR-208a were forecasted. Results The patient group showed a higher miR-208a level than the control group (p < 0.05), and the area under the curve (AUC) of miR-208a in diagnosing AMI was >0.9. After therapy, patients presented notable decreases in serum miR-208a, LDH, CK, and CK-MB (all p < 0.05). Serum miR-208a presented positive associations with LDH, CK, as well as CK-MB both before and after therapy (all p < 0.05). Before therapy, the ineffective group presented a higher miR-208a level than the effective group (p < 0.05), and miR-208a had an AUC of 0.784 in forecasting efficacy. Additionally, the group with adverse events presented a higher miR-208a level than the group without them before therapy (p < 0.05), and miR-208a had an AUC of 0.713 in forecasting adverse events. According to enrichment analysis, the target genes of miR-208a were bound up with signal pathways of cellular senescence, MTOR and Wnt. Conclusion With high expression in AMI cases, miR-208a is a promising potential biomarker for diagnosis and prognosis forecasting of AMI.
Collapse
|
2
|
You H, Zhao Q, Dong M. The Key Genes Underlying Pathophysiology Correlation Between the Acute Myocardial Infarction and COVID-19. Int J Gen Med 2022; 15:2479-2490. [PMID: 35282650 PMCID: PMC8904943 DOI: 10.2147/ijgm.s354885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 02/23/2022] [Indexed: 11/30/2022] Open
Abstract
Introduction Accumulating evidences disclose that COVID-19, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has a marked effect on acute myocardial infarction (AMI). Nevertheless, the underlying pathophysiology correlation between the AMI and COVID-19 remains vague. Materials and Methods Bioinformatics analyses of the altered transcriptional profiling of peripheral blood mononuclear cells (PBMCs) in patients with AMI and COVID-19 were implemented, including identification of differentially expressed genes and common genes between AMI and COVID-19, protein–protein interactions, Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analyses, TF-genes and miRNA coregulatory networks, to explore their biological functions and potential roles in the pathogenesis of COVID-19-related AMI. Conclusion Our bioinformatic analyses of gene expression profiling of PBMCs in patients with AMI and COVID-19 provide us with a unique view regarding underlying pathophysiology correlation between the two vital diseases.
Collapse
Affiliation(s)
- Hongjun You
- Department of Cardiovascular Medicine, Shaanxi Provincial People’s Hospital, Xi’an, 710068, Shaanxi, People’s Republic of China
| | - Qianqian Zhao
- Department of Clinical Immunology, The First Affiliated Hospital, Air Force Military Medical University, Xi’an, 710032, Shaanxi, People’s Republic of China
| | - Mengya Dong
- Department of Cardiovascular Medicine, Shaanxi Provincial People’s Hospital, Xi’an, 710068, Shaanxi, People’s Republic of China
- Correspondence: Mengya Dong, Department of Cardiovascular Medicine, Shaanxi Provincial People’s Hospital, 256 West Youyi Road, Xi’an, Shaanxi, 710068, People’s Republic of China, Tel +86–15802943974, Email
| |
Collapse
|
3
|
MicroRNA-489 Promotes the Apoptosis of Cardiac Muscle Cells in Myocardial Ischemia-Reperfusion Based on Smart Healthcare. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:2538769. [PMID: 35035817 PMCID: PMC8759872 DOI: 10.1155/2022/2538769] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/01/2021] [Accepted: 12/09/2021] [Indexed: 01/10/2023]
Abstract
With the development of information technology, the concept of smart healthcare has gradually come to the fore. Smart healthcare uses a new generation of information technologies, such as the Internet of Things (loT), big data, cloud computing, and artificial intelligence, to transform the traditional medical system in an all-around way, making healthcare more efficient, more convenient, and more personalized. miRNAs can regulate the proliferation, differentiation, and apoptosis of human cells. Relevant studies have also shown that miRNAs may play a key role in the occurrence and development of myocardial ischemia-reperfusion injury (MIRI). This study aims to explore the effects of miR-489 in MIRI. In this study, miR-489 expression in a myocardial ischemia-reperfusion animal model and H9C2 cells induced by H/R was detected by qRT-PCR. The release of lactate dehydrogenase (LDH) and the activity of creatine kinase (CK) was detected after miR-489 knockdown in H9C2 cells induced by H/R. The apoptosis of H9C2 cells and animal models were determined by ELISA. The relationship between miR-489 and SPIN1 was verified by a double fluorescence reporter enzyme assay. The expression of the PI3K/AKT pathway-related proteins was detected by Western blot. Experimental results showed that miR-489 was highly expressed in cardiac muscle cells of the animal model and in H9C2 cells induced by H/R of the myocardial infarction group, which was positively associated with the apoptosis of cardiac muscle cells with ischemia-reperfusion. miR-489 knockdown can reduce the apoptosis of cardiac muscle cells caused by ischemia-reperfusion. In downstream targeting studies, it was found that miR-489 promotes the apoptosis of cardiac muscle cells after ischemia-reperfusion by targeting the inhibition of the SPIN1-mediated PI3K/AKT pathway. In conclusion, high expression of miR-489 is associated with increased apoptosis of cardiac muscle cells after ischemia-reperfusion, which can promote the apoptosis after ischemia-reperfusion by targeting the inhibition of the SPIN1-mediated PI3K/AKT pathway. Therefore, miR-489 can be one of the potential therapeutic targets for reducing the apoptosis of cardiac muscle cells after ischemia-reperfusion.
Collapse
|
4
|
Chen J, Liu Z, Ma L, Gao S, Fu H, Wang C, Lu A, Wang B, Gu X. Targeting Epigenetics and Non-coding RNAs in Myocardial Infarction: From Mechanisms to Therapeutics. Front Genet 2022; 12:780649. [PMID: 34987550 PMCID: PMC8721121 DOI: 10.3389/fgene.2021.780649] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 11/30/2021] [Indexed: 12/12/2022] Open
Abstract
Myocardial infarction (MI) is a complicated pathology triggered by numerous environmental and genetic factors. Understanding the effect of epigenetic regulation mechanisms on the cardiovascular disease would advance the field and promote prophylactic methods targeting epigenetic mechanisms. Genetic screening guides individualised MI therapies and surveillance. The present review reported the latest development on the epigenetic regulation of MI in terms of DNA methylation, histone modifications, and microRNA-dependent MI mechanisms and the novel therapies based on epigenetics.
Collapse
Affiliation(s)
- Jinhong Chen
- Department of TCM, Tianjin University of TCM, Tianjin, China
| | - Zhichao Liu
- Department of TCM, Tianjin University of TCM, Tianjin, China
| | - Li Ma
- Department of TCM, Tianjin University of TCM, Tianjin, China
| | - Shengwei Gao
- Department of TCM, Tianjin University of TCM, Tianjin, China
| | - Huanjie Fu
- Department of TCM, Tianjin University of TCM, Tianjin, China
| | - Can Wang
- Acupuncture Department, The First Affiliated Hospital of Tianjin University of TCM, Tianjin, China
| | - Anmin Lu
- Department of TCM, Tianjin University of TCM, Tianjin, China
| | - Baohe Wang
- Department of Cardiology, The Second Affiliated Hospital of Tianjin University of TCM, Tianjin, China
| | - Xufang Gu
- Department of Cardiology, The Second Affiliated Hospital of Tianjin University of TCM, Tianjin, China
| |
Collapse
|
5
|
Pan J, Alexan B, Dennis D, Bettina C, Christoph LIM, Tang Y. microRNA-193-3p attenuates myocardial injury of mice with sepsis via STAT3/HMGB1 axis. J Transl Med 2021; 19:386. [PMID: 34503521 PMCID: PMC8428118 DOI: 10.1186/s12967-021-03022-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 08/04/2021] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE Little is known regarding the functional role of microRNA-193-3p (miR-193-3p) in sepsis. Hence, the aim of the present study was to investigate the effect of miR-193-3p on myocardial injury in mice with sepsis and its mechanism through the regulation of signal transducers and activators of transcription 3 (STAT3). METHODS The mice model of sepsis was established by cecal ligation and puncture (CLP), septic mice were injected with miR-193-3p agomir, miR-193-3p antagomir or siRNA-STAT3. The expression of miR-193-3p, STAT3 and HMGB1 in the myocardial tissue of septic mice were detected. Cardiac ultrasound, hemodynamics, myocardial injury markers, inflammatory factors and cardiomyocyte apoptosis in septic mice were measured. RESULTS MiR-193-3p expression was reduced while STAT3 expression was increased in septic mice. Down-regulated STAT3 or up-regulated miR-193-3p improved cardiac function, attenuated myocardial injury, inflammation and cardiomyocyte apoptosis in septic mice. Knockdown STAT3 reversed the role of inhibited miR-193-3p for mice with sepsis. miR-193-3p targeted STAT3, thereby inhibiting HMGB1 expression. CONCLUSION This study provides evidence that miR-193-3p targets STAT3 expression to reduce HMGB1 expression, thereby reducing septic myocardial damage. MiR-193-3p might be a potential candidate marker and therapeutic target for sepsis.
Collapse
Affiliation(s)
- Jianyuan Pan
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, 230001, Anhui, China
- Institute of Experimental Cardiology, Internal Medicine VIII, Heidelberg University, Heidelberg, Germany
| | - Buse Alexan
- Institute of Experimental Cardiology, Internal Medicine VIII, Heidelberg University, Heidelberg, Germany
| | - Dorn Dennis
- Institute of Experimental Cardiology, Internal Medicine VIII, Heidelberg University, Heidelberg, Germany
- Anatomy and Developmental Biology, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Chiristine Bettina
- Institute of Experimental Cardiology, Internal Medicine VIII, Heidelberg University, Heidelberg, Germany
- Anatomy and Developmental Biology, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Laeuf Ilona Mariya Christoph
- Anatomy and Developmental Biology, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Yongqin Tang
- Anatomy and Developmental Biology, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany.
- Department of General surgery, Chuzhou Hospital affiliated to Anhui Medical University, 230001, Anhui, China.
| |
Collapse
|
6
|
Zou B, Huang T, Wu D, Hu X, Xiao L, Wang C, Zhang H, Xiang J, Hu C, Wu Q, Wu T. Knockdown of ZFAS1 improved the cardiac function of myocardial infarction rats via regulating Wnt/β-catenin signaling pathway. Aging (Albany NY) 2021; 13:12919-12928. [PMID: 33952724 PMCID: PMC8148456 DOI: 10.18632/aging.202961] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 03/14/2021] [Indexed: 01/21/2023]
Abstract
Myocardial infarction (MI) is a big health threat in the world, and it is characterized by high morbidity and mortality. However, current treatments are not effective enough, and novel therapeutic strategies need to be explored. ZFAS1 has been proved to be involved in the regulation of MI, but the specific mechanism remains unclear. MI rats were constructed through left anterior descending artery ligation, and hypoxia cell model was also established. The proliferation, invasion, and migration of cells were detected via CCK8, traswell, and wound healing methods. Immunohistochemistry staining, western blotting, and qRT-PCR were used to detect the levels of molecules. Knockdown of ZFAS1 significantly increased the proliferation, migration, and invasion of cardiac fibroblasts. Knockdown of ZFAS1 remarkably improved cardiac function via decreasing infarction ratio and increasing vWF expression, left ventricular ejection fraction, and left ventricular fractional shortening compared with group MI. Knockdown of ZFAS1 also suppressed Wnt/β-catenin pathway in vivo. The inhibition of Wnt/β-catenin remarkably reversed the influence of shZFAS1 on cardiac function and cardiac fibroblasts viability. Therefore, Knockdown of ZFAS1 could improve the cardiac function of myocardial infarction rats via regulating Wnt/β-catenin signaling pathway. The present study might provide new thoughts for the prevention and treatment of MI damage.
Collapse
Affiliation(s)
- Bing Zou
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Tieqiu Huang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Dan Wu
- Jiangxi Health Vocational College, Nanchang, Jiangxi 330052, China
| | - Xiaomin Hu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Linghui Xiao
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Chenxi Wang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Hongzhou Zhang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Jian Xiang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Chenkai Hu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Qinghua Wu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Tao Wu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| |
Collapse
|
7
|
Effect of Interventions in WNT Signaling on Healing of Cardiac Injury: A Systematic Review. Cells 2021; 10:cells10020207. [PMID: 33494313 PMCID: PMC7912185 DOI: 10.3390/cells10020207] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/08/2021] [Accepted: 01/14/2021] [Indexed: 12/12/2022] Open
Abstract
The wound healing that follows myocardial infarction is a complex process involving multiple mechanisms, such as inflammation, angiogenesis and fibrosis. In the last two decades, the involvement of WNT signaling has been extensively studied and effects on virtually all aspects of this wound healing have been reported. However, as often is the case in a newly emerging field, inconsistent and sometimes even contradictory findings have been reported. The aim of this systematic review is to provide a comprehensive overview of studies in which the effect of interventions in WNT signaling were investigated in in vivo models of cardiac injury. To this end, we used different search engines to perform a systematic search of the literature using the key words "WNT and myocardial and infarction". We categorized the interventions according to their place in the WNT signaling pathway (ligand, receptor, destruction complex or nuclear level). The most consistent improvements of the wound healing response were observed in studies in which the acylation of WNT proteins was inhibited by administering porcupine inhibitors, by inhibiting of the downstream glycogen synthase kinase-3β (GSK3β) and by intervening in the β-catenin-mediated gene transcription. Interestingly, in several of these studies, evidence was presented for activation of cardiomyocyte proliferation around the infarct area. These findings indicate that inhibition of WNT signaling can play a valuable role in the repair of cardiac injury, thereby improving cardiac function and preventing the development of heart failure.
Collapse
|
8
|
Zhu G, Yang S, Wang R, Lei J, Ji P, Wang J, Tao K, Yang C, Ge S, Wang L. P53/miR-154 Pathway Regulates the Epithelial-Mesenchymal Transition in Glioblastoma Multiforme Cells by Targeting TCF12. Neuropsychiatr Dis Treat 2021; 17:681-693. [PMID: 33664574 PMCID: PMC7924251 DOI: 10.2147/ndt.s273578] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 01/18/2021] [Indexed: 02/06/2023] Open
Abstract
PURPOSE Glioblastoma multiforme (GBM) is an aggressive brain tumor with a rather short survival time. Mutation of p53 has been observed and reported to play critical roles in the progression of GBM. However, the pathological mechanisms are still unclear. This study was designed to identify the role of miR-154 in mediating the biological functions of p53 in glioblastoma multiforme. METHODS In the current study, the expression of miR-154 in GBM tissue samples and cell lines with wt-p53 or mutant p53 was evaluated. The functions of miR-154 in tumor migration, invasion and epithelial-mesenchymal transition were analyzed in vitro. A luciferase reporter assay was used to identify the target of miR-154. RESULTS We found that expression of miR-154 was much lower in patient tissues with mutant p53. Further study revealed that p53 was a transcription factor of miR-154 and that the R273H mutation led to its inactivation. In addition, overexpression of miR-154 remarkably suppressed cell migration, invasion and EMT in vitro and tumor growth in vivo. Moreover, TCF12 was proven to be a direct target of miR-154, and the tumor suppressive effect of miR-154 was reversed by TCF12. CONCLUSION Overall, miR-154, which was regulated by wt-p53, inhibited migration, invasion and EMT of GBM cells by targeting TCF12, indicating that miR-154 may act as a biomarker and that the p53/miR-154/TCF12 pathway could be a potential therapeutic target for GBM.
Collapse
Affiliation(s)
- Gang Zhu
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Shirong Yang
- Department of General Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Ronglin Wang
- Department of Oncology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Jie Lei
- Department of Neurosurgery, Wuhan General Hospital of PLA, Wuhan, Hubei, People's Republic of China
| | - Peigang Ji
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Jiancai Wang
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Kai Tao
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Chen Yang
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Shunnan Ge
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Liang Wang
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| |
Collapse
|