1
|
Wu C, Hong YX, Zhang XC, Li JZ, Li YT, Xie J, Wang RY, Wang Y, Li G. SIRT1-dependent regulation of mitochondrial metabolism participates in miR-30a-5p-mediated cardiac remodeling post-myocardial infarction. Free Radic Biol Med 2025; 226:117-128. [PMID: 39557133 DOI: 10.1016/j.freeradbiomed.2024.11.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/25/2024] [Accepted: 11/14/2024] [Indexed: 11/20/2024]
Abstract
Myocardial infarction-triggered myocardial remodeling is fatal for therapies. The miR-30 family is an essential component of several physiological and pathological processes. Previous studies have proved that the miR-30 family may contribute to regulating myocardial infarction. This study aimed to demonstrate that the combination of miR-30a-5p and mitochondrial metabolism recapitulates the critical features for remodeling post-myocardial infarction. Using gain- and loss-of-function of miR-30a-5p in mice, we found miR-30a-5p is highly expressed in the heart and is reduced in infarcted hearts. Further evidence showed that miR-30a-5p acts as a protective molecule to maintain myocardial remodeling, fibrosis, and mitochondrial structure. Mitochondrial function, ATP production, and mitochondrial respiratory chain proteins were positively regulated by miR-30a-5p. Mechanistically, alterations in these properties depend on SIRT1, which modulates miR-30a-5p-regulated mitochondrial metabolism. Remarkably, reactivation of SIRT1 prevented miR-30a-5p deficiency-aggravated myocardial infarction-induced myocardial remodeling. These data identified miR-30a-5p as a critical modulator of mitochondrial function in cardiomyocytes and revealed that the miR-30a-5p-SIRT1-mitochondria network is essential for myocardial infarction-induced cardiac remodeling.
Collapse
Affiliation(s)
- Chan Wu
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361000, China
| | - Yi-Xiang Hong
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361000, China
| | - Xiao-Cheng Zhang
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361000, China
| | - Jing-Zhou Li
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361000, China
| | - Yu-Ting Li
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361000, China
| | - Jun Xie
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Rui-Ying Wang
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361000, China; Xiamen Key Laboratory of Cardiovascular Diseases, Xiamen, Fujian, 361000, China.
| | - Yan Wang
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361000, China; Xiamen Key Laboratory of Cardiovascular Diseases, Xiamen, Fujian, 361000, China.
| | - Gang Li
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361000, China; Xiamen Key Laboratory of Cardiovascular Diseases, Xiamen, Fujian, 361000, China.
| |
Collapse
|
2
|
Hong YX, Wu C, Li JZ, Song F, Hu Y, Han Y, Mao YJ, Wu WY, Wang Y, Li G. SUMOylation of TP53INP1 is involved in miR-30a-5p-regulated heart senescence. Exp Mol Med 2024; 56:2519-2534. [PMID: 39511427 PMCID: PMC11612193 DOI: 10.1038/s12276-024-01347-3] [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: 03/27/2024] [Revised: 08/11/2024] [Accepted: 08/27/2024] [Indexed: 11/15/2024] Open
Abstract
Heart senescence is critical for cardiac function. This study aimed to characterize the role and mechanism of action of miR-30a-5p in cardiac senescence. miR-30a-5p was downregulated in aged mouse hearts and neonatal rat cardiomyocytes (NRCMs). In vivo, using a combination of echocardiography and different molecular biological approaches, we investigated the role of miR-30a-5p knockout or overexpression in natural- or D-galactose-induced heart aging in mice. In vitro, using RNA sequencing and a series of molecular biology methods, the mechanism by which miR-30a-5p regulates cardiac senescence was explored in cardiomyocytes. miR-30a-5p knockout mice showed aggravated natural- or D-galactose-induced heart aging compared to wild-type littermate mice, with significantly decreased heart function, an increased number of γH2AX-positive cells, reduced telomere length, and upregulated p21 and p53 expression. Cardiac-specific knockdown of miR-30a-5p using adeno-associated virus 9 in D-galactose-induced senescent wild-type mice resulted in effects similar to those observed in knockout mice. Notably, the overexpression of miR-30a-5p in wild-type murine hearts alleviated D-galactose-induced heart senescence by improving heart function, increasing telomere length, decreasing the number of γH2AX-positive cells, and downregulating p53 and p21 expression. This was confirmed in D-galactose-treated or naturally aged NRCMs. Mechanistically, TP53INP1 was identified as a target of miR-30a-5p by mediating the SUMOylation of TP53INP1 and its translocation from the cytoplasm to the nucleus to interact with p53. Furthermore, this study demonstrated that cardiac-specific TP53INP1 deficiency ameliorates miR-30a-5p knockout-aggravated cardiac dysfunction and heart senescence. This study identified miR-30a-5p as a crucial modulator of heart senescence and revealed that the miR-30a-5p-TP53INP1-p53 axis is essential for heart and cardiomyocyte aging.
Collapse
Affiliation(s)
- Yi-Xiang Hong
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, Fujian, China
| | - Chan Wu
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, Fujian, China
| | - Jing-Zhou Li
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, Fujian, China
| | - Fei Song
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, Fujian, China
| | - Yu Hu
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, Fujian, China
| | - Yue Han
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, Fujian, China
| | - Yi-Jie Mao
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, Fujian, China
| | - Wei-Yin Wu
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, Fujian, China
- Xiamen Key Laboratory of Cardiovascular Diseases, Xiamen, 361000, Fujian, China
| | - Yan Wang
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, Fujian, China.
- Xiamen Key Laboratory of Cardiovascular Diseases, Xiamen, 361000, Fujian, China.
| | - Gang Li
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, Fujian, China.
- Xiamen Key Laboratory of Cardiovascular Diseases, Xiamen, 361000, Fujian, China.
| |
Collapse
|
3
|
El Khayari A, Hakam SM, Malka G, Rochette L, El Fatimy R. New insights into the cardio-renal benefits of SGLT2 inhibitors and the coordinated role of miR-30 family. Genes Dis 2024; 11:101174. [PMID: 39224109 PMCID: PMC11367061 DOI: 10.1016/j.gendis.2023.101174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 09/15/2023] [Accepted: 10/10/2023] [Indexed: 09/04/2024] Open
Abstract
Sodium-glucose co-transporter inhibitors (SGLTis) are the latest class of anti-hyperglycemic agents. In addition to inhibiting the absorption of glucose by the kidney causing glycosuria, these drugs also demonstrate cardio-renal benefits in diabetic subjects. miR-30 family, one of the most abundant microRNAs in the heart, has recently been linked to a setting of cardiovascular diseases and has been proposed as novel biomarkers in kidney dysfunctions as well; their expression is consistently dysregulated in a variety of cardio-renal dysfunctions. The mechanistic involvement and the potential interplay between miR-30 and SGLT2i effects have yet to be thoroughly elucidated. Recent research has stressed the relevance of this cluster of microRNAs as modulators of several pathological processes in the heart and kidneys, raising the possibility of these small ncRNAs playing a central role in various cardiovascular complications, notably, endothelial dysfunction and pathological remodeling. Here, we review current evidence supporting the pleiotropic effects of SGLT2is in cardiovascular and renal outcomes and investigate the link and the coordinated implication of the miR-30 family in endothelial dysfunction and cardiac remodeling. We also discuss the emerging role of circulating miR-30 as non-invasive biomarkers and attractive therapeutic targets for cardiovascular diseases and kidney diseases. Clinical evidence, as well as metabolic, cellular, and molecular aspects, are comprehensively covered.
Collapse
Affiliation(s)
- Abdellatif El Khayari
- Institute of Biological Sciences (ISSB-P), UM6P Faculty of Medical Sciences, Mohammed VI Polytechnic University (UM6P), Ben-Guerir 43150, Morocco
| | - Soukaina Miya Hakam
- Institute of Biological Sciences (ISSB-P), UM6P Faculty of Medical Sciences, Mohammed VI Polytechnic University (UM6P), Ben-Guerir 43150, Morocco
| | - Gabriel Malka
- Institute of Biological Sciences (ISSB-P), UM6P Faculty of Medical Sciences, Mohammed VI Polytechnic University (UM6P), Ben-Guerir 43150, Morocco
| | - Luc Rochette
- Equipe d'Accueil (EA 7460): Physiopathologie et Epidémiologie Cérébro-Cardiovasculaires (PEC2), Université de Bourgogne – Franche Comté, Faculté des Sciences de Santé, 7 Bd Jeanne d'Arc, Dijon 21000, France
| | - Rachid El Fatimy
- Institute of Biological Sciences (ISSB-P), UM6P Faculty of Medical Sciences, Mohammed VI Polytechnic University (UM6P), Ben-Guerir 43150, Morocco
| |
Collapse
|
4
|
Wang R, Chen RL, Wu C, Zhang XC, Wu WY, Dai C, Wang Y, Li G. The gut microbiotas with metabolites regulate the protective role of miR-30a-5p in myocardial infarction. J Adv Res 2024:S2090-1232(24)00472-7. [PMID: 39442873 DOI: 10.1016/j.jare.2024.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/21/2024] [Accepted: 10/16/2024] [Indexed: 10/25/2024] Open
Abstract
INTRODUCTION Gut microbial homeostasis is closely associated with myocardial infarction (MI). However, little is known about how gut microbiota influences miRNAs-regulated MI. OBJECTIVES This study aims to elucidate the connections between miR-30a-5p, MI, gut microbiota, and gut microbial metabolite-related pathways, to explore potential strategy for preventing and treating MI. METHODS We evaluated the effects of knocking out (KO) or overexpressing (OE) miR-30a-5p on MI by assessing cardiac structure and function, myocardial enzyme levels, and apoptosis. Then, we applied 16S rDNA sequencing and metabolomics to explore how intestinal microecology and its microorganisms affect miR-30a-5p-regulated MI. RESULTS The results showed that KO exacerbated MI, whereas OE improved MI damage, compared to the wild-type (WT) mice. KO exacerbated intestinal barrier structure deterioration and further downregulated the expression of Cloudin-1, Occludin, and ZO-1 in MI mice. 16S rDNA sequencing-analyzed gut microbiome of KO and WT mice found that KO mainly reduced g_Lactobacillus. Transplanting fecal microorganisms from KO mice aggravated MI damage in WT mice. However, administering probiotics (mainly containing Lactobacillus) helped neutralize these damages. Intriguingly, fecal microbiota transplantation from OE mice reduced MI damage. Analysis of intestinal microbial metabolites in KO and WT mice found that KO may mainly affect ABC transporters. ABCC1 was identified as the target of KO-aggravated MI. Furthermore, fecal transplantation microorganisms of MI patients aggravated MI injury in mice and miR-30a-5p and ABCC1 were involved in the process. CONCLUSIONS Our findings demonstrate that miR-30a-5p regulates MI by affecting intestinal microbiota homeostasis and targeting ABCC1. This highlights the critical importance of maintaining a healthy gut microbiota homeostasis in MI management.
Collapse
Affiliation(s)
- Ruiying Wang
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361000, China; Xiamen Key Laboratory of Cardiovascular Diseases, Xiamen, Fujian 361000, China
| | - Ruo-Lan Chen
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361000, China
| | - Chan Wu
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361000, China
| | - Xiao-Cheng Zhang
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361000, China
| | - Wei-Yin Wu
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361000, China; Xiamen Key Laboratory of Cardiovascular Diseases, Xiamen, Fujian 361000, China
| | - Cuilian Dai
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361000, China; Xiamen Key Laboratory of Cardiovascular Diseases, Xiamen, Fujian 361000, China
| | - Yan Wang
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361000, China; Xiamen Key Laboratory of Cardiovascular Diseases, Xiamen, Fujian 361000, China.
| | - Gang Li
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361000, China; Xiamen Key Laboratory of Cardiovascular Diseases, Xiamen, Fujian 361000, China.
| |
Collapse
|
5
|
Wang R, Zhang X, Wang Y, Lin Y, Zhou Y, Wang Y, Li G. Gut microbiota of miR-30a-5p-deleted mice aggravate high-fat diet-induced hepatic steatosis by regulating arachidonic acid metabolic pathway. Clin Transl Med 2024; 14:e70035. [PMID: 39360667 PMCID: PMC11447637 DOI: 10.1002/ctm2.70035] [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: 06/11/2024] [Revised: 09/08/2024] [Accepted: 09/15/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Patients with non-alcoholic fatty liver disease (NAFLD) often exhibit hepatic steatosis and dyslipidemia. Studies have shown that intestinal microorganisms are closely related to the occurrence of NAFLD and atherosclerosis. Our previous study has underscored the protective role of microRNA-30a-5p (miR-30a-5p) against atherosclerosis. METHODS AND RESULTS In the present study, we aimed to elucidate the effect and underlying mechanism of the intestinal microorganisms of miR-30a-5p knockout (KO) mice on NAFLD. Our findings demonstrated that KO exacerbated high-fat diet (HFD)-induced hepatic steatosis and disrupted liver function, as evidenced by elevated levels of total cholesterol, low-density lipoprotein, alanine aminotransferase, aspartate transaminase, and total bile acids in serum. Fecal microbiota from HFD-fed KO mice induced hepatic steatosis, dyslipidemia, and higher levels of enzymes indicative of liver damage in wild-type mice. Remarkably, KO mice significantly intensified the above effects. 16s rDNA sequencing and metabolomics of the intestinal microbiota in the HFD-treated KO and WT mice showed that the loss of miR-30a-5p resulted in intestinal microbiota imbalance and was highly related to the arachidonic acid metabolic pathway. Targeted metabolomic in the liver tissues unveiled upregulation of COX-related (PGF2a, 8-iso-PGF2a and PGF2) and LOX-related (LTB4, LTD4, 12S-HETE and 15S-HETE) factors in HFD-treated KO mice. Immunohistochemistry and transcriptional analyses showed that miR-30a-5p affected arachidonic acid metabolism through the LOX/COX pathways. Besides, COX/LOX pathways and hepatic steatosis were reversed after reintroducing miR-30a-5p in HFD-treated KO mice. CONCLUSIONS This study reveals the pivotal mechanism by which miR-30a-5p and intestinal microbes regulate hepatic steatosis and abnormal lipid metabolism, offering promising avenues for NAFLD and atherosclerosis therapeutics. HIGHLIGHTS MiR-30a-5p deletion aggravated hepatic steatosis and lipid disorder induced by an HFD in mice. Gut microbiota participated in the regulation of hepatic steatosis in the context of miR-30a-5p. Gut microbiota metabolism-related arachidonic acid metabolic pathway contributed to miR-30a-5p-regulated hepatic steatosis and lipid disorder. Reintroducing miR-30a-5p reversed hepatic steatosis and arachidonic acid metabolism disorder caused by HFD and miR-30a-5p deletion.
Collapse
Affiliation(s)
- Ruiying Wang
- Xiamen Cardiovascular Hospital of Xiamen UniversitySchool of Medicine, Xiamen UniversityXiamenFujianP. R. China
- Xiamen Key Laboratory of Cardiovascular DiseasesXiamen Cardiovascular Hospital of Xiamen UniversityXiamenFujianP. R. China
| | - Xiaocheng Zhang
- Xiamen Cardiovascular Hospital of Xiamen UniversitySchool of Medicine, Xiamen UniversityXiamenFujianP. R. China
| | - Yutian Wang
- Department of CardiologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdongP. R. China
| | - Yijun Lin
- Xiamen Cardiovascular Hospital of Xiamen UniversitySchool of Medicine, Xiamen UniversityXiamenFujianP. R. China
- Xiamen Key Laboratory of Cardiovascular DiseasesXiamen Cardiovascular Hospital of Xiamen UniversityXiamenFujianP. R. China
| | - Yuling Zhou
- Xiamen Cardiovascular Hospital of Xiamen UniversitySchool of Medicine, Xiamen UniversityXiamenFujianP. R. China
- Xiamen Key Laboratory of Cardiovascular DiseasesXiamen Cardiovascular Hospital of Xiamen UniversityXiamenFujianP. R. China
| | - Yan Wang
- Xiamen Cardiovascular Hospital of Xiamen UniversitySchool of Medicine, Xiamen UniversityXiamenFujianP. R. China
- Xiamen Key Laboratory of Cardiovascular DiseasesXiamen Cardiovascular Hospital of Xiamen UniversityXiamenFujianP. R. China
| | - Gang Li
- Xiamen Cardiovascular Hospital of Xiamen UniversitySchool of Medicine, Xiamen UniversityXiamenFujianP. R. China
- Xiamen Key Laboratory of Cardiovascular DiseasesXiamen Cardiovascular Hospital of Xiamen UniversityXiamenFujianP. R. China
| |
Collapse
|
6
|
Liang G, Guo C, Tang H, Zhang M. miR-30a-5p attenuates hypoxia/reoxygenation-induced cardiomyocyte apoptosis by regulating PTEN protein expression and activating PI3K/Akt signaling pathway. BMC Cardiovasc Disord 2024; 24:236. [PMID: 38705985 PMCID: PMC11070099 DOI: 10.1186/s12872-024-03900-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 04/24/2024] [Indexed: 05/07/2024] Open
Abstract
BACKGROUND This study was designed to investigate the mechanism by which miR-30a-5p mediates cardiomyocyte apoptosis after acute myocardial infarction (AMI) induced by hypoxia/reoxygenation (H/R). METHODS Differentially expressed miRNAs were analyzed by RNA high-throughput sequencing in acute myocardial infarction (ST-elevation myocardial infarction) patients versus healthy individuals (controls). The H/R model was used to assess the regulatory mechanism of miRNAs in AMI. Lentivirus-associated vectors were used to overexpress or knock down miR-30a-5p in cellular models. The pathological mechanisms of miR-30a-5p regulating the development of acute myocardial infarction were serially explored by qPCR, bioinformatics, target gene prediction, dual luciferase, enzyme-linked immunosorbent assays (ELISAs) and Western blotting. RESULTS The results showed that the expression of miR-30a-5p was significantly increased in AMI patients and H9C2 cells. Hypoxia decreased cardiomyocyte survival over time, and reoxygenation further reduced cell survival. Bax and Phosphatase and tensin homolog (PTEN)were suppressed, while Bcl-2 was upregulated. Additionally, miR-30a-5p specifically targeted the PTEN gene. According to the GO and KEGG analyses, miR-30a-5p may participate in apoptosis by interacting with PTEN. The miR-30a-5p mimic decreased the expression of apoptosis-related proteins and the levels of the proinflammatory markers IL-1β, IL-6, and TNF-α by activating the PTEN/PI3K/Akt signaling pathway. Conversely, anti-miR-30a-5p treatment attenuated these effects. Additionally, silencing PTEN and anti-miR-30a-5p had opposite effects on H/R-induced cell apoptosis. CONCLUSIONS miR-30a-5p plays a crucial role in cardiomyocyte apoptosis after hypoxia-induced acute myocardial infarction. Our findings provide translational evidence that miR-30a-5p is a novel potential therapeutic target for AMI.
Collapse
Affiliation(s)
- Guoxin Liang
- Department of Laboratory Medicine, Nanjing BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
- Graduate School, North China University of Science and Technology, Tangshan, Hebei, 063210, China
- Clinical Medicine Research Center, Hebei Key Laboratory of Metabolic Diseases, Hebei General Hospital, 348#, Hepingxi Road, PO Box: 050051, Shijiazhuang, 050051, China
| | - Chang Guo
- Graduate School, North China University of Science and Technology, Tangshan, Hebei, 063210, China
- Clinical Medicine Research Center, Hebei Key Laboratory of Metabolic Diseases, Hebei General Hospital, 348#, Hepingxi Road, PO Box: 050051, Shijiazhuang, 050051, China
| | - Hongyue Tang
- Clinical Medicine Research Center, Hebei Key Laboratory of Metabolic Diseases, Hebei General Hospital, 348#, Hepingxi Road, PO Box: 050051, Shijiazhuang, 050051, China
- School of Clinical Medicine, Graduate School of Hebei North College, Zhangjiakou, Hebei, 075000, China
| | - Mingming Zhang
- Clinical Medicine Research Center, Hebei Key Laboratory of Metabolic Diseases, Hebei General Hospital, 348#, Hepingxi Road, PO Box: 050051, Shijiazhuang, 050051, China.
| |
Collapse
|
7
|
Tobaruela EDC, Brasili E, Zeraik L, Milenkovic D, Hassimotto NMA, Lajolo FM. Plasma proteome profiling reveals molecular mechanisms underlying the effects of daily consumption of 'Bahia' and 'Cara Cara' orange juices. Food Funct 2024; 15:1031-1049. [PMID: 38193367 DOI: 10.1039/d3fo04091g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2024]
Abstract
Orange juice is an important food source of bioactive compounds, mainly the flavanones hesperidin and narirutin. This study aimed to investigate the underlying molecular mechanisms of action of orange juice's health properties by analyzing changes in the plasma proteome of healthy Brazilian volunteers after consuming juices made from 'Bahia' (BOJ-source of flavanones) and 'Cara Cara' (CCOJ-source of flavanones and carotenoids) oranges cultivated in Brazil. We used an untargeted proteomic approach, with a particular emphasis on the juices' effects on blood coagulant activity. We identified 247 differentially expressed proteins, of which 170 significantly increased or decreased after BOJ consumption and 145 after CCOJ. These proteins are involved in 105 processes that can significantly regulate cell adhesion, cell signaling, cell metabolism, inflammation, or others. Bioinformatic analysis evidenced proteins with major cellular regulatory capacity (e.g., FN1 and GAPDH) and predicted transcription factors (TFs) (e.g., SP1 and CEBPA) and miRNAs (e.g., miR-1-3p and miR-615-3p) that could be involved in the regulation of differentially expressed proteins. In-silico docking analyses between flavanone metabolites and TFs evidenced the higher binding capacity of narirutin phase II metabolites with akt1 and p38, interactions that suggest how the expression of genes of differentially expressed proteins were activated or inhibited. Moreover, the study shed light on proteins of coagulation cascade that presented expression modulated by both juices, proposing the modulation of blood coagulant activity as a potential benefit of OJ (mainly CCOJ) consumption. Taken together, this study revealed that BOJ and CCOJ consumption affected plasma proteome in healthy individuals, suggesting potential molecular targets and mechanisms of OJ bioactive compounds in humans.
Collapse
Affiliation(s)
- Eric de Castro Tobaruela
- Food Research Center (FoRC), Department of Food and Experimental Nutrition, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil.
| | - Elisa Brasili
- Department of Environmental Biology, Sapienza University of Rome, Rome, Italy
| | - Laila Zeraik
- Food Research Center (FoRC), Department of Food and Experimental Nutrition, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil.
| | - Dragan Milenkovic
- Department of Nutrition, University of California Davis, 95616 Davis, CA, USA
| | - Neuza Mariko Aymoto Hassimotto
- Food Research Center (FoRC), Department of Food and Experimental Nutrition, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil.
| | - Franco Maria Lajolo
- Food Research Center (FoRC), Department of Food and Experimental Nutrition, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil.
| |
Collapse
|
8
|
Gehris J, Ervin C, Hawkins C, Womack S, Churillo AM, Doyle J, Sinusas AJ, Spinale FG. Fibroblast activation protein: Pivoting cancer/chemotherapeutic insight towards heart failure. Biochem Pharmacol 2024; 219:115914. [PMID: 37956895 PMCID: PMC10824141 DOI: 10.1016/j.bcp.2023.115914] [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: 08/25/2023] [Revised: 11/06/2023] [Accepted: 11/07/2023] [Indexed: 11/21/2023]
Abstract
An important mechanism for cancer progression is degradation of the extracellular matrix (ECM) which is accompanied by the emergence and proliferation of an activated fibroblast, termed the cancer associated fibroblast (CAF). More specifically, an enzyme pathway identified to be amplified with local cancer progression and proliferation of the CAF, is fibroblast activation protein (FAP). The development and progression of heart failure (HF) irrespective of the etiology is associated with left ventricular (LV) remodeling and changes in ECM structure and function. As with cancer, HF progression is associated with a change in LV myocardial fibroblast growth and function, and expresses a protein signature not dissimilar to the CAF. The overall goal of this review is to put forward the postulate that scientific discoveries regarding FAP in cancer as well as the development of specific chemotherapeutics could be pivoted to target the emergence of FAP in the activated fibroblast subtype and thus hold translationally relevant diagnostic and therapeutic targets in HF.
Collapse
Affiliation(s)
- John Gehris
- Cell Biology and Anatomy and Cardiovascular Research Center, University of South Carolina School of Medicine and the Columbia VA Health Care System, Columbia, SC, United States
| | - Charlie Ervin
- Cell Biology and Anatomy and Cardiovascular Research Center, University of South Carolina School of Medicine and the Columbia VA Health Care System, Columbia, SC, United States
| | - Charlotte Hawkins
- Cell Biology and Anatomy and Cardiovascular Research Center, University of South Carolina School of Medicine and the Columbia VA Health Care System, Columbia, SC, United States
| | - Sydney Womack
- Cell Biology and Anatomy and Cardiovascular Research Center, University of South Carolina School of Medicine and the Columbia VA Health Care System, Columbia, SC, United States
| | - Amelia M Churillo
- Cell Biology and Anatomy and Cardiovascular Research Center, University of South Carolina School of Medicine and the Columbia VA Health Care System, Columbia, SC, United States
| | - Jonathan Doyle
- Cell Biology and Anatomy and Cardiovascular Research Center, University of South Carolina School of Medicine and the Columbia VA Health Care System, Columbia, SC, United States
| | - Albert J Sinusas
- Yale University Cardiovascular Imaging Center, New Haven CT, United States
| | - Francis G Spinale
- Cell Biology and Anatomy and Cardiovascular Research Center, University of South Carolina School of Medicine and the Columbia VA Health Care System, Columbia, SC, United States.
| |
Collapse
|
9
|
Wu YX, Xu RY, Jiang L, Chen XY, Xiao XJ. MicroRNA-30a-5p Promotes Chronic Heart Failure in Rats by Targeting Sirtuin-1 to Activate the Nuclear Factor-κB/NOD-Like Receptor 3 Signaling Pathway. Cardiovasc Drugs Ther 2023; 37:1065-1076. [PMID: 35488974 DOI: 10.1007/s10557-021-07304-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/14/2021] [Indexed: 11/03/2022]
Abstract
OBJECTIVE MicroRNA-30a-5p (miR-30a-5p) has been identified as a marker of heart failure; however, its functional mechanisms in chronic heart failure (CHF) remain unknown. We aim to investigate the role of miR-30a-5p targeting sirtuin-1 (SIRT1) in myocardial remodeling in CHF via the nuclear factor-κB/NOD-like receptor 3 (NF-κB/NLRP3) signaling pathway. METHODS CHF rat models were established using aortic coarctation. The expression of miR-30a-5p, SIRT1, and the NF-κB/NLRP3 signaling pathway-related factors in CHF rats was determined. The CHF rats were then respectively treated with altered miR-30a-5p or SIRT1 to explore their roles in cardiac function, myocardial function, inflammatory response, pathological changes, and cardiomyocyte apoptosis. The binding relation between miR-30a-5p and SIRT1 was confirmed. RESULTS MiR-30a-5p was upregulated whereas SIRT1 was downregulated in myocardial tissues of CHF rats. MiR-30a-5p inhibition or SIRT1 overexpression improved cardiac and myocardial function, and suppressed the inflammatory response, alleviated pathological changes and inhibited cardiomyocyte apoptosis in CHF rats. MiR-30a-5p targeted SIRT1 to regulate the NF-κB/NLRP3 signaling pathway. In CHF rats, downregulated miR-30a-5p and silenced SIRT1 could reverse the beneficial effects of downregulated miR-30a-5p. CONCLUSION Inhibited miR-30a-5p inhibits CHF progression via the SIRT1-mediated NF-κB/NLRP3 signaling pathway.
Collapse
Affiliation(s)
- Yu-Xian Wu
- Department of Critical Care Medicine, Quanzhou First Hospital, Quanzhou, 362000, Fujian Province, China
| | - Rong-Yu Xu
- Department of Thoracic Surgery, Quanzhou First Hospital, Quanzhou, 362000, Fujian Province, China
| | - Ling Jiang
- Department of Cardiovascular Medicine, The First Hospital of Nanping, Nanping, 353000, Fujian Province, China
| | - Xiang-Yan Chen
- Department of Critical Care Medicine, Quanzhou First Hospital, Quanzhou, 362000, Fujian Province, China
| | - Xiong-Jian Xiao
- Department of Critical Care Medicine, The First Affiliated Hospital of Fujian Medical University, No. 20 Chazhong Road, Fuzhou, 350000, Fujian, China.
| |
Collapse
|
10
|
Liu X, Dou B, Tang W, Yang H, Chen K, Wang Y, Qin J, Yang F. Cardioprotective effects of circ_0002612 in myocardial ischemia/reperfusion injury correlate with disruption of miR-30a-5p-dependent Ppargc1a inhibition. Int Immunopharmacol 2023; 117:110006. [PMID: 37012879 DOI: 10.1016/j.intimp.2023.110006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 02/19/2023] [Accepted: 03/06/2023] [Indexed: 03/14/2023]
Abstract
INTRODUCTION Novel mechanistic insights into the effects of circular RNAs (circRNAs) on the physiology and pathology of cardiovascular diseases are under increasingly active investigation. This study defined the cardioprotective role and mechanistic actions of circ_0002612 in myocardial ischemia/reperfusion injury (MI/RI). METHODS MI/RI was induced in mice by ligation of the left anterior descending (LAD) artery followed by reperfusion, and the in vitro model was established in cultured cardiomyocytes under hypoxia/reoxygenation (H/R) conditions. Interaction among circ_0002612, miR-30a-5p, Ppargc1a, and NLRP3 was predicted by bioinformatics analysis and further experimentally identified. Gain- and loss-of-function experiments were performed to evaluate the effect of the circ_0002612/miR-30a-5p/Ppargc1a/NLRP3 axis on the cardiac function and myocardial infarction of I/R-injured mice, as well as viability and apoptosis of H/R-challenged cardiomyocytes. RESULTS In the myocardial tissues of MI/RI mice, miR-30a-5p was negatively correlated with circ_0002612 or Ppargc1a, but circ_0002612 was positively correlated with the expression of Ppargc1a. circ_0002612 competitively bound to miR-30a-5p to release expression of its target gene Ppargc1a. circ_0002612 promoted cardiomyocyte viability while suppressing the apoptosis by impairing the miR-30a-5p-mediated inhibition of Ppargc1a. Additionally, Ppargc1a inhibited the expression of NLRP3 and consequently facilitated cardiomyocyte proliferation while suppressing cell apoptosis. By inhibiting the expression of NLRP3, circ_0002612 protected mice from MI/RI. CONCLUSION Overall, this study demonstrates the cardioprotective role of circ_0002612 against MI/RI, which may be a viable target for MI/RI.
Collapse
|
11
|
Chen X, He X, Yang YY, Wang Y. Amyotrophic Lateral Sclerosis-Associated Mutants of SOD1 Modulate miRNA Biogenesis through Aberrant Interactions with Exportin 5. ACS Chem Biol 2022; 17:3450-3457. [PMID: 36475596 PMCID: PMC9867941 DOI: 10.1021/acschembio.2c00591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mutations in the SOD1 (superoxide dismutase 1) gene are associated with amyotrophic lateral sclerosis (ALS), a fatal neurodegenerative disease. By employing ascorbate peroxidase-based proximity labeling, coupled with LC-MS/MS analysis, we uncovered 43 and 24 proteins exhibiting higher abundance in the proximity proteomes of SOD1G85R and SOD1G93A, respectively, than that of wild-type SOD1. Immunoprecipitation followed by western blot analysis indicated the preferential binding of one of these proteins, exportin 5 (XPO5), toward the two mutants of SOD1 over the wild-type counterpart. In line with the established function of XPO5 in pre-miRNA transport, we observed diminished nucleocytoplasmic transport of pre-miRNAs in cells with ectopic expression of the two SOD1 mutants over those expressing the wild-type protein. On the other hand, RT-qPCR results revealed significant elevations in mature miRNA in cells expressing the two SOD1 mutants, which are attributed to the diminished inhibitory effect of XPO5 on Dicer-mediated cleavage of pre-miRNA to mature miRNA. Together, our chemoproteomic approach led to the revelation of a novel mechanism through which ALS-associated mutants of SOD1 perturb miRNA biogenesis, that is, through aberrant binding toward XPO5.
Collapse
Affiliation(s)
- Xingyuan Chen
- Environmental Toxicology Graduate Program, University of California, Riverside, California 92502, United States
| | - Xiaomei He
- Department of Chemistry, University of California, Riverside, California 92502, United States
| | - Yen-Yu Yang
- Department of Chemistry, University of California, Riverside, California 92502, United States
| | - Yinsheng Wang
- Environmental Toxicology Graduate Program, University of California, Riverside, California 92502, United States
- Department of Chemistry, University of California, Riverside, California 92502, United States
| |
Collapse
|
12
|
Kabłak-Ziembicka A, Badacz R, Przewłocki T. Clinical Application of Serum microRNAs in Atherosclerotic Coronary Artery Disease. J Clin Med 2022; 11:6849. [PMID: 36431326 PMCID: PMC9698927 DOI: 10.3390/jcm11226849] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/15/2022] [Accepted: 11/18/2022] [Indexed: 11/22/2022] Open
Abstract
MicroRNAs (miRs) are promising diagnostic, prognostic and therapeutic biomolecules for atherosclerotic cardiovascular disease. Atherosclerotic occlusive disease concerns a large population of patients, carrying the highest incidence of fatal and non-fatal adverse events, such as myocardial infarction, ischemic stroke, and limb ischemia, worldwide. Consistently, miRs are involved in regulation and pathogenesis of atherosclerotic coronary artery disease (CAD), acute coronary syndromes (ACS), both with ST-segment (STEMI) and non-ST segment elevation myocardial infarctions (NSTEMI), as well as cardiac remodeling and fibrosis following ACS. However, the genetic and molecular mechanisms underlying adverse outcomes in CAD are multifactorial, and sometimes difficult to interpret for clinicians. Therefore, in the present review paper we have focused on the clinical meaning and the interpretation of various miRs findings, and their potential application in routine clinical practice.
Collapse
Affiliation(s)
- Anna Kabłak-Ziembicka
- Department of Interventional Cardiology, Institute of Cardiology, Jagiellonian University Medical College, św. Anny 12, 31-007 Kraków, Poland
- Noninvasive Cardiovascular Laboratory, The John Paul II Hospital, Prądnicka 80, 31-202 Kraków, Poland
| | - Rafał Badacz
- Department of Interventional Cardiology, Institute of Cardiology, Jagiellonian University Medical College, św. Anny 12, 31-007 Kraków, Poland
- Department of Interventional Cardiology, The John Paul II Hospital, Prądnicka 80, 31-202 Kraków, Poland
| | - Tadeusz Przewłocki
- Department of Interventional Cardiology, The John Paul II Hospital, Prądnicka 80, 31-202 Kraków, Poland
- Department of Cardiac and Vascular Diseases, Institute of Cardiology, Jagiellonian University Medical College, św. Anny 12, 31-007 Kraków, Poland
| |
Collapse
|
13
|
TREML4 polymorphisms increase the mRNA in blood leukocytes in the progression of atherosclerosis. Sci Rep 2022; 12:18612. [PMID: 36329152 PMCID: PMC9633690 DOI: 10.1038/s41598-022-22040-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 10/07/2022] [Indexed: 11/06/2022] Open
Abstract
TREML4 and other members of the triggering receptor expressed in the myeloid cell family are associated with a risk of atherosclerosis and progression in coronary artery disease, acute coronary syndrome, and coronary artery calcification. Herein, the relationship between TREML4 expression and its polymorphisms (rs2803495 and rs280396) was evaluated in patients with subclinical atherosclerosis (n = 340) and heart failure post-acute myocardial infarction (MI) (n = 68) for the first time. TREML4 variants rs2803495 (A > G) and rs2803496 (T > C) and leukocyte mRNA expression was analyzed by qRT-PCR. The rs2803495 G allele was associated with TREML4 expression (OR 8.01, CI 3.78-16.99, p < 0.001). Patients carrying the rs2803496 C minor allele (TC/CC genotypes) were more likely to express TREML4 than those without the C allele (OR 10.42, CI 4.76-22.78, p < 0.001), as well as having higher levels of TREML4 expression (OR 4.88, CI 2.35-10.12, p < 0.001). Thus, we report for the first time that TREML4 is not associated with the early stages of atherosclerotic plaque formation and later stages after MI. In conclusion, TREML4 mRNA expression in blood leukocytes is influenced by minor alleles (G and C) and may regulate differently during the atherosclerosis progression stages, but not in asymptomatic atherosclerosis disease and post-MI.
Collapse
|
14
|
Venugopal P, George M, Kandadai SD, Balakrishnan K, Uppugunduri CRS. Prioritization of microRNA biomarkers for a prospective evaluation in a cohort of myocardial infarction patients based on their mechanistic role using public datasets. Front Cardiovasc Med 2022; 9:981335. [PMID: 36407428 PMCID: PMC9668885 DOI: 10.3389/fcvm.2022.981335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022] Open
Abstract
Background MicroRNAs (miR) have proven to be promising biomarkers for several diseases due to their diverse functions, stability and tissue/organ-specific nature. Identification of new markers with high sensitivity and specificity will help in risk reduction in acute myocardial infarction (AMI) patients with chest pain and also prevent future adverse outcomes. Hence the aim of this study was to perform a detailed in silico analysis for identifying the mechanistic role of miRs involved in the pathogenesis/prognosis of AMI for prospective evaluation in AMI patients. Methods miR profiling data was extracted from GSE148153 and GSE24591 datasets using the GEO2R gene expression omnibus repository and analyzed using limma algorithm. Differentially expressed miRs were obtained by comparing MI patients with corresponding controls after multiple testing corrections. Data mining for identifying candidate miRs from published literature was also performed. Target prediction and gene enrichment was done using standard bioinformatics tools. Disease specific analysis was performed to identify target genes specific for AMI using open targets platform. Protein-protein interaction and pathway analysis was done using STRING database and Cytoscape platform. Results and conclusion The analysis revealed significant miRs like let-7b-5p, let-7c-5p, miR-4505, and miR-342-3p in important functions/pathways including phosphatidylinositol-3-kinase/AKT and the mammalian target of rapamycin, advanced glycation end products and its receptor and renin–angiotensin–aldosterone system by directly targeting angiotensin II receptor type 1, forkhead box protein O1, etc. With this approach we were able to prioritize the miR candidates for a prospective clinical association study in AMI patients of south Indian origin.
Collapse
Affiliation(s)
| | - Melvin George
- Clinical Research Department, Hindu Mission Hospital, Chennai, India
| | | | | | - Chakradhara Rao S. Uppugunduri
- CANSEARCH Research Platform in Pediatric Oncology and Hematology, Department of Pediatrics, Gynecology and Obstetrics, University of Geneva, Geneva, Switzerland
- *Correspondence: Chakradhara Rao S. Uppugunduri,
| |
Collapse
|
15
|
Rincón LM, Rodríguez-Serrano M, Conde E, Lanza VF, Sanmartín M, González-Portilla P, Paz-García M, Del Rey JM, Menacho M, García Bermejo ML, Zamorano JL. Serum microRNAs are key predictors of long-term heart failure and cardiovascular death after myocardial infarction. ESC Heart Fail 2022; 9:3367-3379. [PMID: 35837763 DOI: 10.1002/ehf2.13919] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 02/09/2022] [Accepted: 03/21/2022] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Patients with acute myocardial infarction (MI) are at high risk of upcoming events, in particular heart failure (HF), but reliable stratification methods are lacking. Our goal was to evaluate the potential role of circulating miRNAs as prognostic biomarkers in patients presenting with MI. METHODS AND RESULTS We conducted a prospective study among 311 consecutive patients hospitalized with MI (65% ST-segment elevation MI & median age of 55 years) with long-term follow-up. An initial screening was conducted to select candidate miRNAs, with subsequent study of 14 candidate miRNAs. The primary outcome was the composite of hospital admission for HF or cardiovascular death. During a mean follow-up of 2.1 years miR-21-5p, miR-23a-3p, miR27b-3p, miR-122-5p, miR210-3p, and miR-221-3p reliably predicted the primary outcome. Multivariate Cox regression analyses highlighted that miR-210-3p [hazard ratio (HR) 2.65 per 1 SD increase, P < 0.001], miR-23a-3p (HR 2.11 per 1 SD increase, P < 0.001), and miR-221-3p (HR 2.03 per 1 SD increase, P < 0.001) were able to accurately predict the primary outcome, as well as cardiovascular death, HF hospitalizations, and long-term New York Heart Association (NYHA) functional class. These three miRNAs clearly improved the performance of multivariate clinical models: ΔC-statistic = 0.10 [95% confidence interval (CI), 0.03-0.17], continuous net reclassification index = 34.8% (95%CI, 5.8-57.4%), and integrated discrimination improvement (P < 0.001). CONCLUSIONS This is the largest study evaluating the prognostic value of circulating miRNAs for HF-related events among patients with MI. We show that several miRNAs predict HF hospitalizations, cardiovascular mortality, and poor long-term NYHA status and improve current risk prediction methods.
Collapse
Affiliation(s)
- Luis M Rincón
- Department of Cardiology, Hospital Universitario Ramón y Cajal, Universidad de Alcalá, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, 28034, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Universidad de Salamanca, Salamanca, Spain
| | - Macarena Rodríguez-Serrano
- Biomarkers and Therapeutic Targets Laboratory and Core Facility, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Spanish Renal Research Network (REDinREN), Ctra. Colmenar Km 9100, Madrid, 28034, Spain
| | - Elisa Conde
- Biomarkers and Therapeutic Targets Laboratory and Core Facility, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Spanish Renal Research Network (REDinREN), Ctra. Colmenar Km 9100, Madrid, 28034, Spain
| | - Val F Lanza
- Bioinformatics Core Facility, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Marcelo Sanmartín
- Department of Cardiology, Hospital Universitario Ramón y Cajal, Universidad de Alcalá, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, 28034, Spain
| | - Paz González-Portilla
- Department of Cardiology, Hospital Universitario Ramón y Cajal, Universidad de Alcalá, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, 28034, Spain
| | - Marta Paz-García
- Biomarkers and Therapeutic Targets Laboratory and Core Facility, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Spanish Renal Research Network (REDinREN), Ctra. Colmenar Km 9100, Madrid, 28034, Spain
| | - José Manuel Del Rey
- Department of Biochemistry, Hospital Universitario Ramón y Cajal, Universidad de Alcalá, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Miriam Menacho
- Department of Biochemistry, Hospital Universitario Ramón y Cajal, Universidad de Alcalá, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - María-Laura García Bermejo
- Biomarkers and Therapeutic Targets Laboratory and Core Facility, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Spanish Renal Research Network (REDinREN), Ctra. Colmenar Km 9100, Madrid, 28034, Spain
| | - José L Zamorano
- Department of Cardiology, Hospital Universitario Ramón y Cajal, Universidad de Alcalá, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, 28034, Spain
- Hospital La Zarzuela, Madrid, Spain
| |
Collapse
|
16
|
Qian L, Zhao Q, Yu P, Lü J, Guo Y, Gong X, Ding Y, Yu S, Fan L, Fan H, Zhang Y, Liu Z, Sheng H, Yu Z. Diagnostic potential of a circulating miRNA model associated with therapeutic effect in heart failure. Lab Invest 2022; 20:267. [PMID: 35690861 PMCID: PMC9188190 DOI: 10.1186/s12967-022-03465-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 05/30/2022] [Indexed: 11/10/2022]
Abstract
Heart failure (HF), as the leading cause of death, is continuing to increase along with the aging of the general population all over the world. Identification of diagnostic biomarkers for early detection of HF is considered as the most effective way to reduce the risk and mortality. Herein, we collected plasma samples from HF patients (n = 40) before and after medical therapy to determine the change of circulating miRNAs through a quantitative real-time PCR (QRT-PCR)-based miRNA screening analysis. miR-30a-5p and miR-654-5p were identified as the most significantly changed miRNAs in the plasma of patients upon treatment. In consistence, miR-30a-5p showed upregulation and miR-654-5p showed downregulation in the circulation of 30 HF patients, compared to 15 normal controls in the training phase, from which a two-circulating miRNA model was developed for HF diagnosis. Next, we performed the model validation using an independent cohort including 50 HF patients and 30 controls. As high as 98.75% of sensitivity and 95.00% of specificity were achieved. A comparison between the miRNA model and NT-pro BNP in diagnostic accuracy of HF indicated an upward trend of the miRNA model. Moreover, change of the two miRNAs was further verified in association with the therapeutic effect of HF patients, in which miR-30a-5p showed decrease while miR-654-5p showed increase in the plasma of patients after LVAD implantation. In conclusion, the current study not only identified circulating miR-654-5p for the first time as a novel biomarker of HF, but also developed a novel 2-circulating miRNA model with promising potentials for diagnosis and prognosis of HF patients, and in association with therapeutic effects as well.
Collapse
Affiliation(s)
- Lu Qian
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Heart Failure Institute, Shanghai East Hospital, Jinzhou Medical University & Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Qian Zhao
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Heart Failure Institute, Shanghai East Hospital, Jinzhou Medical University & Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Ping Yu
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Heart Failure Institute, Shanghai East Hospital, Jinzhou Medical University & Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Jinhui Lü
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Heart Failure Institute, Shanghai East Hospital, Jinzhou Medical University & Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Yuefan Guo
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Heart Failure Institute, Shanghai East Hospital, Jinzhou Medical University & Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Xin Gong
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Heart Failure Institute, Shanghai East Hospital, Jinzhou Medical University & Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Yuanyuan Ding
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Heart Failure Institute, Shanghai East Hospital, Jinzhou Medical University & Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Shanshan Yu
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Heart Failure Institute, Shanghai East Hospital, Jinzhou Medical University & Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Lieying Fan
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Heart Failure Institute, Shanghai East Hospital, Jinzhou Medical University & Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Huimin Fan
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Heart Failure Institute, Shanghai East Hospital, Jinzhou Medical University & Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Yuzhen Zhang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Heart Failure Institute, Shanghai East Hospital, Jinzhou Medical University & Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Zhongmin Liu
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Heart Failure Institute, Shanghai East Hospital, Jinzhou Medical University & Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China.
| | - Hongzhuan Sheng
- Affiliated Hospital of Nantong University, Nantong, Jiangsu, China.
| | - Zuoren Yu
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Heart Failure Institute, Shanghai East Hospital, Jinzhou Medical University & Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China.
| |
Collapse
|
17
|
Venugopal P, Balakrishnan K, Damal Kandadai S, George M. Usefullness of MicroRNAs in Predicting the Clinical Outcome of Patients with Acute Myocardial Infarction During Follow-Up: A Systematic Review. Genet Test Mol Biomarkers 2022; 26:277-289. [PMID: 35638909 DOI: 10.1089/gtmb.2021.0283] [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] [Indexed: 11/12/2022] Open
Abstract
Background: Myocardial infarction (MI) is reported as the leading cause of mortality and morbidity worldwide. It is associated with a 30% mortality rate. Echocardiography, coronary angiography, and biomarkers like cardiac troponins are employed as prognostic tests. Although these biomarkers are the gold standard for the diagnosis of MI, they are not accurate as prognostic markers due to their lack of specificity. Studies have suggested that dysregulation of specific microRNAs (miRNAs) influences post-MI complications during follow-up. However, the findings of these studies have several inconsistencies. This systematic review was performed to investigate the potential of miRNAs to predict clinical outcomes post-MI. Methodology: Pubmed and Google Scholar databases were used for identifying research articles published from inception till August 2021; the search terms included "microRNAs" AND "prognosis" AND "myocardial infarction" or "acute coronary syndrome." All the articles included were critically analyzed using STROBE guidelines. Results and Conclusion: Several miRNAs were elevated in MI patients, including miR-208b, miR-499, and miR-375. Association of these miRNA levels with the outcome of MI, such as all-cause mortality and major adverse cardiovascular events during follow-up, were also reported. However, none of the studies included in this systematic review exhibited promising evidence that these miRNAs can be utilized as ideal biomarkers for prognosis post-MI. Understanding the molecular mechanisms involved in the pathogenesis and progression of MI is crucial. Hence, these findings can be used as a guide when performing further experimental studies to identify useful post-MI prognostic markers.
Collapse
Affiliation(s)
| | | | | | - Melvin George
- Department of Clinical Research, Hindu Mission Hospital, Chennai, India
| |
Collapse
|
18
|
miRNA Expression Profiling in Subcutaneous Adipose Tissue of Monozygotic Twins Discordant for HIV Infection: Validation of Differentially Expressed miRNA and Bioinformatic Analysis. Int J Mol Sci 2022; 23:ijms23073486. [PMID: 35408847 PMCID: PMC8998861 DOI: 10.3390/ijms23073486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/21/2022] [Accepted: 03/21/2022] [Indexed: 02/04/2023] Open
Abstract
Combined AntiRetroviral Treatments (cARTs) used for HIV infection may result in varied metabolic complications, which in some cases, may be related to patient genetic factors, particularly microRNAs. The use of monozygotic twins, differing only for HIV infection, presents a unique and powerful model for the controlled analysis of potential alterations of miRNAs regulation consequent to cART treatment. Profiling of 2578 mature miRNA in the subcutaneous (SC) adipose tissue and plasma of monozygotic twins was investigated by the GeneChip® miRNA 4.1 array. Real-time PCR and ddPCR experiments were performed in order to validate differentially expressed miRNAs. Target genes of deregulated miRNAs were predicted by the miRDB database (prediction score > 70) and enrichment analysis was carried out with g:Profiler. Processes in SC adipose tissue most greatly affected by miRNA up-regulation included (i) macromolecular metabolic processes, (ii) regulation of neurogenesis, and (iii) protein phosphorylation. Furthermore, KEGG analysis revealed miRNA up-regulation involvement in (i) insulin signaling pathways, (ii) neurotrophin signaling pathways, and (iii) pancreatic cancer. By contrast, miRNA up-regulation in plasma was involved in (i) melanoma, (ii) p53 signaling pathways, and (iii) focal adhesion. Our findings suggest a mechanism that may increase the predisposition of HIV+ patients to insulin resistance and cancer.
Collapse
|
19
|
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: 1.0] [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
|
20
|
The Diagnostic and Therapeutic Potential of Galectin-3 in Cardiovascular Diseases. Biomolecules 2021; 12:biom12010046. [PMID: 35053194 PMCID: PMC8774137 DOI: 10.3390/biom12010046] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/25/2021] [Accepted: 12/27/2021] [Indexed: 12/25/2022] Open
Abstract
Galectin-3 plays a prominent role in chronic inflammation and has been implicated in the development of many disease conditions, including heart disease. Galectin-3, a regulatory protein, is elevated in both acute and chronic heart failure and is involved in the inflammatory pathway after injury leading to myocardial tissue remodelling. We discussed the potential utility of galectin-3 as a diagnostic and disease severity/prognostic biomarker in different cardio/cerebrovascular diseases, such as acute ischemic stroke, acute coronary syndromes, heart failure and arrhythmogenic cardiomyopathy. Over the last decade there has been a marked increase in the understanding the role of galectin-3 in myocardial fibrosis and inflammation and as a therapeutic target for the treatment of heart failure and myocardial infarction.
Collapse
|
21
|
Tanase DM, Gosav EM, Ouatu A, Badescu MC, Dima N, Ganceanu-Rusu AR, Popescu D, Floria M, Rezus E, Rezus C. Current Knowledge of MicroRNAs (miRNAs) in Acute Coronary Syndrome (ACS): ST-Elevation Myocardial Infarction (STEMI). Life (Basel) 2021; 11:life11101057. [PMID: 34685428 PMCID: PMC8541211 DOI: 10.3390/life11101057] [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: 09/13/2021] [Revised: 10/02/2021] [Accepted: 10/06/2021] [Indexed: 02/06/2023] Open
Abstract
Regardless of the newly diagnostic and therapeutic advances, coronary artery disease (CAD) and more explicitly, ST-elevation myocardial infarction (STEMI), remains one of the leading causes of morbidity and mortality worldwide. Thus, early and prompt diagnosis of cardiac dysfunction is pivotal in STEMI patients for a better prognosis and outcome. In recent years, microRNAs (miRNAs) gained attention as potential biomarkers in myocardial infarction (MI) and acute coronary syndromes (ACS), as they have key roles in heart development, various cardiac processes, and act as indicators of cardiac damage. In this review, we describe the current available knowledge about cardiac miRNAs and their functions, and focus mainly on their potential use as novel circulating diagnostic and prognostic biomarkers in STEMI.
Collapse
Affiliation(s)
- Daniela Maria Tanase
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (A.O.); (M.C.B.); (N.D.); (A.R.G.-R.); (D.P.); (C.R.)
- Internal Medicine Clinic, “Sf. Spiridon” County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| | - Evelina Maria Gosav
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (A.O.); (M.C.B.); (N.D.); (A.R.G.-R.); (D.P.); (C.R.)
- Internal Medicine Clinic, “Sf. Spiridon” County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
- Correspondence: (E.M.G.); (M.F.); (E.R.)
| | - Anca Ouatu
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (A.O.); (M.C.B.); (N.D.); (A.R.G.-R.); (D.P.); (C.R.)
- Internal Medicine Clinic, “Sf. Spiridon” County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| | - Minerva Codruta Badescu
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (A.O.); (M.C.B.); (N.D.); (A.R.G.-R.); (D.P.); (C.R.)
- Internal Medicine Clinic, “Sf. Spiridon” County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| | - Nicoleta Dima
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (A.O.); (M.C.B.); (N.D.); (A.R.G.-R.); (D.P.); (C.R.)
- Internal Medicine Clinic, “Sf. Spiridon” County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| | - Ana Roxana Ganceanu-Rusu
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (A.O.); (M.C.B.); (N.D.); (A.R.G.-R.); (D.P.); (C.R.)
- Internal Medicine Clinic, “Sf. Spiridon” County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| | - Diana Popescu
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (A.O.); (M.C.B.); (N.D.); (A.R.G.-R.); (D.P.); (C.R.)
- Internal Medicine Clinic, “Sf. Spiridon” County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| | - Mariana Floria
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (A.O.); (M.C.B.); (N.D.); (A.R.G.-R.); (D.P.); (C.R.)
- Internal Medicine Clinic, Emergency Military Clinical Hospital Iasi, 700483 Iasi, Romania
- Correspondence: (E.M.G.); (M.F.); (E.R.)
| | - Elena Rezus
- Department of Rheumatology and Physiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- I Rheumatology Clinic, Clinical Rehabilitation Hospital, 700661 Iasi, Romania
- Correspondence: (E.M.G.); (M.F.); (E.R.)
| | - Ciprian Rezus
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (A.O.); (M.C.B.); (N.D.); (A.R.G.-R.); (D.P.); (C.R.)
- Internal Medicine Clinic, “Sf. Spiridon” County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| |
Collapse
|
22
|
Weale CJ, Matshazi DM, Davids SFG, Raghubeer S, Erasmus RT, Kengne AP, Davison GM, Matsha TE. Expression Profiles of Circulating microRNAs in South African Type 2 Diabetic Individuals on Treatment. Front Genet 2021; 12:702410. [PMID: 34567065 PMCID: PMC8456082 DOI: 10.3389/fgene.2021.702410] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 08/13/2021] [Indexed: 12/30/2022] Open
Abstract
Aim: The influence of disease duration and anti-diabetic treatment on epigenetic processes has been described, with limited focus on interactions with microRNAs (miRNAs). miRNAs have been found to play key roles in the regulation of pathways associated with type 2 diabetes mellitus (T2DM), and expression patterns in response to treatment may further promote their use as therapeutic targets in T2DM and its associated complications. We therefore aimed to investigate the expressions of circulating miRNAs (miR-30a-5p, miR-1299, miR-182-5p, miR-30e-3p and miR-126-3p) in newly diagnosed and known diabetics on treatment, in South Africa. Methods: A total of 1254 participants with an average age of 53.8years were included in the study and classified according to glycaemic status (974 normotolerant, 92 screen-detected diabetes and 188 known diabetes). Whole blood levels of miR-30a-5p, miR-1299, miR-182-5p, miR-30e-3p and miR-126-3p were quantitated using RT-qPCR. Expression analysis was performed and compared across groups. Results: All miRNAs were significantly overexpressed in subjects with known diabetes when compared to normotolerant individuals, as well as known diabetics vs. screen-detected (p<0.001). Upon performing regression analysis, of all miRNAs, only miR-182-5p remained associated with the duration of the disease after adjustment for type of treatment (OR: 0.127, CI: 0.018–0.236, p=0.023). Conclusion: Our findings revealed important associations and altered expression patterns of miR-30a-5p, miR-1299, miR-182-5p, miR-30e-3p and miR-126-3p in known diabetics on anti-diabetic treatment compared to newly diagnosed individuals. Additionally, miR-182-5p expression decreased with increasing duration of T2DM. Further studies are, however, recommended to shed light on the involvement of the miRNA in insulin signalling and glucose homeostasis, to endorse its use as a therapeutic target in DM and its associated complications.
Collapse
Affiliation(s)
- Cecil J Weale
- SAMRC/CPUT/Cardiometabolic Health Research Unit, Department of Biomedical Sciences, Faculty of Health and Wellness Science, Cape Peninsula University of Technology, Cape Town, South Africa
| | - Don M Matshazi
- SAMRC/CPUT/Cardiometabolic Health Research Unit, Department of Biomedical Sciences, Faculty of Health and Wellness Science, Cape Peninsula University of Technology, Cape Town, South Africa
| | - Saarah F G Davids
- SAMRC/CPUT/Cardiometabolic Health Research Unit, Department of Biomedical Sciences, Faculty of Health and Wellness Science, Cape Peninsula University of Technology, Cape Town, South Africa
| | - Shanel Raghubeer
- SAMRC/CPUT/Cardiometabolic Health Research Unit, Department of Biomedical Sciences, Faculty of Health and Wellness Science, Cape Peninsula University of Technology, Cape Town, South Africa
| | - Rajiv T Erasmus
- National Health Laboratory Service (NHLS), Division of Chemical Pathology, Faculty of Health Sciences, University of Stellenbosch, Cape Town, South Africa
| | - Andre P Kengne
- Non-Communicable Diseases Research Unit, South African Medical Research Council, Cape Town, South Africa.,Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Glenda M Davison
- SAMRC/CPUT/Cardiometabolic Health Research Unit, Department of Biomedical Sciences, Faculty of Health and Wellness Science, Cape Peninsula University of Technology, Cape Town, South Africa
| | - Tandi E Matsha
- SAMRC/CPUT/Cardiometabolic Health Research Unit, Department of Biomedical Sciences, Faculty of Health and Wellness Science, Cape Peninsula University of Technology, Cape Town, South Africa
| |
Collapse
|
23
|
MicroRNAs-The Heart of Post-Myocardial Infarction Remodeling. Diagnostics (Basel) 2021; 11:diagnostics11091675. [PMID: 34574016 PMCID: PMC8469128 DOI: 10.3390/diagnostics11091675] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 09/02/2021] [Accepted: 09/09/2021] [Indexed: 12/20/2022] Open
Abstract
Myocardial infarction (MI) is one of the most frequent cardiac emergencies, with significant potential for mortality. One of the major challenges of the post-MI healing response is that replacement fibrosis could lead to left ventricular remodeling (LVR) and heart failure (HF). This process involves canonical and non-canonical transforming growth factor-beta (TGF-β) signaling pathways translating into an intricate activation of cardiac fibroblasts and disproportionate collagen synthesis. Accumulating evidence has indicated that microRNAs (miRNAs) significantly contribute to the modulation of these signaling pathways. This review summarizes the recent updates regarding the molecular mechanisms underlying the role of the over 30 miRNAs involved in post-MI LVR. In addition, we compare the contradictory roles of several multifunctional miRNAs and highlight their potential use in pressure overload and ischemia-induced fibrosis. Finally, we discuss their attractive role as prognostic biomarkers for HF, highlighting the most relevant human trials involving these miRNAs.
Collapse
|
24
|
Bär C, Chatterjee S, Falcão Pires I, Rodrigues P, Sluijter JPG, Boon RA, Nevado RM, Andrés V, Sansonetti M, de Windt L, Ciccarelli M, Hamdani N, Heymans S, Figuinha Videira R, Tocchetti CG, Giacca M, Zacchigna S, Engelhardt S, Dimmeler S, Madonna R, Thum T. Non-coding RNAs: update on mechanisms and therapeutic targets from the ESC Working Groups of Myocardial Function and Cellular Biology of the Heart. Cardiovasc Res 2021; 116:1805-1819. [PMID: 32638021 DOI: 10.1093/cvr/cvaa195] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/15/2020] [Accepted: 06/30/2020] [Indexed: 02/06/2023] Open
Abstract
Vast parts of mammalian genomes are actively transcribed, predominantly giving rise to non-coding RNA (ncRNA) transcripts including microRNAs, long ncRNAs, and circular RNAs among others. Contrary to previous opinions that most of these RNAs are non-functional molecules, they are now recognized as critical regulators of many physiological and pathological processes including those of the cardiovascular system. The discovery of functional ncRNAs has opened up new research avenues aiming at understanding ncRNA-related disease mechanisms as well as exploiting them as novel therapeutics in cardiovascular therapy. In this review, we give an update on the current progress in ncRNA research, particularly focusing on cardiovascular physiological and disease processes, which are under current investigation at the ESC Working Groups of Myocardial Function and Cellular Biology of the Heart. This includes a range of topics such as extracellular vesicle-mediated communication, neurohormonal regulation, inflammation, cardiac remodelling, cardio-oncology as well as cardiac development and regeneration, collectively highlighting the wide-spread involvement and importance of ncRNAs in the cardiovascular system.
Collapse
Affiliation(s)
- Christian Bär
- Institute for Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany.,REBIRTH Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Shambhabi Chatterjee
- Institute for Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany.,REBIRTH Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Inês Falcão Pires
- Cardiovascular Research and Development Center, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Patrícia Rodrigues
- Cardiovascular Research and Development Center, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Joost P G Sluijter
- Experimental Cardiology Laboratory, UMC Utrecht Regenerative Medicine Center, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Reinier A Boon
- Department of Physiology, Amsterdam Cardiovascular Sciences (ACS), Amsterdam UMC, VU University Medical Center, Amsterdam, The Netherlands.,Institute for Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany.,Partner site Rhein/Main, German Center for Cardiovascular Research (DZHK), Frankfurt am Main, Germany
| | - Rosa M Nevado
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain
| | - Vicente Andrés
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain
| | - Marida Sansonetti
- Institute for Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany.,REBIRTH Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany.,Department of Molecular Genetics, Faculty of Science and Engineering, Maastricht University, Maastricht, The Netherlands.,Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Leon de Windt
- Department of Molecular Genetics, Faculty of Science and Engineering, Maastricht University, Maastricht, The Netherlands.,Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Michele Ciccarelli
- Department of Medicine, Surgery and Dentistry, University of Salerno, Italy
| | - Nazha Hamdani
- Department of Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany.,Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany
| | - Stephane Heymans
- Department of Cardiology, Maastricht University Medical Centre, University Hospital Maastricht, The Netherlands.,Center for Heart Failure Research, Cardiovascular Research Institute Maastricht (CARIM), University Hospital Maastricht, The Netherlands
| | - Raquel Figuinha Videira
- Cardiovascular Research and Development Center, Faculty of Medicine, University of Porto, Porto, Portugal.,Department of Molecular Genetics, Faculty of Science and Engineering, Maastricht University, Maastricht, The Netherlands.,Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Carlo G Tocchetti
- Department of Translational Medical Sciences and Interdepartmental Center of Clinical and Translational Research (CIRCET), Federico II University, Naples, Italy
| | - Mauro Giacca
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy.,School of Cardiovascular Medicine & Sciences, King's College London, London, UK.,Department of Medicine, Surgery and Health Sciences, University of Trieste, Italy
| | - Serena Zacchigna
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy.,Department of Medicine, Surgery and Health Sciences, University of Trieste, Italy
| | - Stefan Engelhardt
- Institute of Pharmacology and Toxicology, Technische Universität München, Biedersteiner Str. 29, Munich 80802, Germany.,DZHK (German Center for Cardiovascular Research), Partner site Munich Heart Alliance, Biedersteiner Str. 29, Munich 80802, Germany
| | - Stefanie Dimmeler
- Institute for Cardiovascular Regeneration, Goethe University, Germany.,German Center for Cardiovascular Research (DZHK), Frankfurt, Germany.,Cardio-Pulmonary Institute (CPI), Frankfurt, Germany
| | - Rosalinda Madonna
- Institute of Cardiology, University of Pisa, Pisa, Italy.,Department of Internal Medicine, University of Texas Medical School, Houston, TX, USA
| | - Thomas Thum
- Institute for Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany.,REBIRTH Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| |
Collapse
|
25
|
Xu L, Zhang H, Wang Y, Guo W, Gu L, Yang A, Ma S, Yang Y, Wu K, Jiang Y. H3K14 hyperacetylation‑mediated c‑Myc binding to the miR‑30a‑5p gene promoter under hypoxia postconditioning protects senescent cardiomyocytes from hypoxia/reoxygenation injury. Mol Med Rep 2021; 23:468. [PMID: 33880587 PMCID: PMC8097758 DOI: 10.3892/mmr.2021.12107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 02/22/2021] [Indexed: 11/25/2022] Open
Abstract
Our previous study reported that microRNA (miR)‑30a‑5p upregulation under hypoxia postconditioning (HPostC) exert a protective effect on aged H9C2 cells against hypoxia/reoxygenation injury via DNA methyltransferase 3B‑induced DNA hypomethylation at the miR‑30a‑5p gene promoter. This suggests that miR‑30a‑5p may be a potential preventative and therapeutic target for ischemic heart disease in aged myocardium. The present study aimed to investigate the underlying mechanisms of miR‑30a‑5p transcription in aged myocardium in ischemic heart disease. Cardiomyocytes were treated with 8 mg/ml D‑galactose for 9 days, and then exposed to hypoxic conditions. Cell viability was determined using a cell viability assay. Expression levels of histone deacetylase 2 (HDAC2), LC3B‑II/I, beclin‑1 and p62 were detected via reverse transcription‑quantitative PCR and western blotting. Chromatin immunoprecipitation‑PCR and luciferase reporter assays were performed to evaluate the effect of c‑Myc binding and activity on the miR‑30a‑5p promoter in senescent cardiomyocytes following HPostC. It was found that HPostC enhanced the acetylation levels of H3K14 at the miR‑30a‑5p gene promoter in senescent cardiomyocytes, which attributed to the decreased expression of HDAC2. In addition, c‑Myc could positively regulate miR‑30a‑5p transcription to inhibit senescent cardiomyocyte autophagy. Mechanically, it was observed that increased H3K14 acetylation level exposed to romidepsin facilitated c‑Myc binding to the miR‑30a‑5p gene promoter region, which led to the increased transcription of miR‑30a‑5p. Taken together, these results demonstrated that HDAC2‑mediated H3K14 hyperacetylation promoted c‑Myc binding to the miR‑30a‑5p gene promoter, which contributed to HPostC senescent cardioprotection.
Collapse
Affiliation(s)
- Lingbo Xu
- Department of Pathophysiology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Huiping Zhang
- Department of Prenatal Diagnosis Center, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Yanhua Wang
- Department of Gynecology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Wei Guo
- Department of Pathophysiology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Lingyu Gu
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Anning Yang
- Department of Pathophysiology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Shengchao Ma
- Department of Pathophysiology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Yong Yang
- Department of Nuclear Medicine, The People's Hospital in Ningxia Hui Autonomous Region, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Kai Wu
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Yideng Jiang
- Department of Pathophysiology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| |
Collapse
|
26
|
Matshazi DM, Weale CJ, Erasmus RT, Kengne AP, Davids SFG, Raghubeer S, Hector S, Davison GM, Matsha TE. MicroRNA Profiles in Normotensive and Hypertensive South African Individuals. Front Cardiovasc Med 2021; 8:645541. [PMID: 33937359 PMCID: PMC8085261 DOI: 10.3389/fcvm.2021.645541] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 03/23/2021] [Indexed: 12/18/2022] Open
Abstract
Hypertension has a complex pathogenesis and symptoms appear in advanced disease. Dysregulation of gene expression regulatory factors like microRNAs has been reported in disease development. Identifying biomarkers which could help understand the pathogenesis and prognosis of hypertension is essential. The study's objective was to investigate microRNA expression profiles according to participant blood pressure status. Next generation sequencing was used to identify microRNAs in the whole blood of 48 body mass index-, smoking- and age-matched normotensive (n = 12), screen-detected hypertensive (n = 16) and known hypertensive (n = 20) female participants. Quantitative reverse transcription polymerase chain reaction was used to validate the next generation sequencing findings in a larger, independent sample of 84 men and 179 women. Using next generation sequencing, 30 dysregulated microRNAs were identified and miR-1299 and miR-30a-5p were the most significantly differentially expressed. Both microRNAs were upregulated in known hypertensives or screen-detected hypertensives compared to the normotensives. Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis indicated possible involvement of platelet activation, calcium signaling and aldosterone synthesis pathways. Further validation of miR-1299 and miR-30a-5p using quantitative reverse transcription polymerase chain reaction confirmed sequencing results while yielding new findings. These findings demonstrate microRNA dysregulation in hypertension and their expression may be related to genes and biological pathways essential for blood pressure homeostasis.
Collapse
Affiliation(s)
- Don M Matshazi
- South African Medical Research Council/Cape Peninsula University of Technology Cardiometabolic Health Research Unit, Department of Biomedical Sciences, Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Cape Town, South Africa
| | - Cecil J Weale
- South African Medical Research Council/Cape Peninsula University of Technology Cardiometabolic Health Research Unit, Department of Biomedical Sciences, Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Cape Town, South Africa
| | - Rajiv T Erasmus
- Division of Chemical Pathology, Faculty of Health Sciences, National Health Laboratory Service and Stellenbosch University, Cape Town, South Africa
| | - Andre P Kengne
- Non-communicable Diseases Research Unit, South African Medical Research Council, Cape Town, South Africa.,Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Saarah F G Davids
- South African Medical Research Council/Cape Peninsula University of Technology Cardiometabolic Health Research Unit, Department of Biomedical Sciences, Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Cape Town, South Africa
| | - Shanel Raghubeer
- South African Medical Research Council/Cape Peninsula University of Technology Cardiometabolic Health Research Unit, Department of Biomedical Sciences, Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Cape Town, South Africa
| | - Stanton Hector
- South African Medical Research Council/Cape Peninsula University of Technology Cardiometabolic Health Research Unit, Department of Biomedical Sciences, Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Cape Town, South Africa
| | - Glenda M Davison
- South African Medical Research Council/Cape Peninsula University of Technology Cardiometabolic Health Research Unit, Department of Biomedical Sciences, Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Cape Town, South Africa
| | - Tandi E Matsha
- South African Medical Research Council/Cape Peninsula University of Technology Cardiometabolic Health Research Unit, Department of Biomedical Sciences, Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Cape Town, South Africa
| |
Collapse
|
27
|
Cruz MS, da Silva AMG, de Souza KSC, Luchessi AD, Silbiger VN. miRNAs emerge as circulating biomarkers of post-myocardial infarction heart failure. Heart Fail Rev 2021; 25:321-329. [PMID: 31254148 DOI: 10.1007/s10741-019-09821-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Heart failure (HF) is a clinical syndrome that involves structural changes in the heart, leading to a decrease in cardiac output, mainly caused by myocardial infarction (MI), which is the most common form of cardiovascular disease worldwide. Clinical evaluation remains the most accurate diagnostic method for ischemic HF, since the known biomarkers have high cost, are difficult to use for early diagnosis, and have low specificity. This often leads to late diagnosis since only ~ 25% symptoms of HF appear after MI. Studies suggest that small non-coding RNAs (miRNAs) play an important role in the regulation of this pathophysiological process and are, therefore, important targets in the discovery of non-invasive biomarkers for HF. Thus, the aim of this review was to identify circulating miRNAs (plasma, serum, and whole blood) described for post-MI HF patients. This review covered 19 experimental studies on humans, which investigated the relationship between circulating miRNAs and the development, monitoring, or prognosis of ischemic HF. This analysis was aimed at proposing potential targets for HF and the future application of miRNAs as HF biomarkers.
Collapse
Affiliation(s)
- Marina Sampaio Cruz
- Department of Clinical and Toxicology Analysis, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | | | - Karla Simone Costa de Souza
- Department of Clinical and Toxicology Analysis, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - André Ducati Luchessi
- Department of Clinical and Toxicology Analysis, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Vivian Nogueira Silbiger
- Department of Clinical and Toxicology Analysis, Federal University of Rio Grande do Norte, Natal, RN, Brazil.
| |
Collapse
|
28
|
Niderla-Bielińska J, Ścieżyńska A, Moskalik A, Jankowska-Steifer E, Bartkowiak K, Bartkowiak M, Kiernozek E, Podgórska A, Ciszek B, Majchrzak B, Ratajska A. A Comprehensive miRNome Analysis of Macrophages Isolated from db/db Mice and Selected miRNAs Involved in Metabolic Syndrome-Associated Cardiac Remodeling. Int J Mol Sci 2021; 22:2197. [PMID: 33672153 PMCID: PMC7926522 DOI: 10.3390/ijms22042197] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/19/2021] [Accepted: 02/20/2021] [Indexed: 01/10/2023] Open
Abstract
Cardiac macrophages are known from various activities, therefore we presume that microRNAs (miRNAs) produced or released by macrophages in cardiac tissue have impact on myocardial remodeling in individuals with metabolic syndrome (MetS). We aim to assess the cardiac macrophage miRNA profile by selecting those miRNA molecules that potentially exhibit regulatory functions in MetS-related cardiac remodeling. Cardiac tissue macrophages from control and db/db mice (an animal model of MetS) were counted and sorted with flow cytometry, which yielded two populations: CD45+CD11b+CD64+Ly6Chi and CD45+CD11b+CD64+Ly6Clow. Total RNA was then isolated, and miRNA expression profiles were evaluated with Next Generation Sequencing. We successfully sequenced 1400 miRNAs in both macrophage populations: CD45+CD11b+CD64+Ly6Chi and CD45+CD11b+CD64+Ly6Clow. Among the 1400 miRNAs, about 150 showed different expression levels in control and db/db mice and between these two subpopulations. At least 15 miRNAs are possibly associated with MetS pathology in cardiac tissue due to direct or indirect regulation of the expression of miRNAs for proteins involved in angiogenesis, fibrosis, or inflammation. In this paper, for the first time we describe the miRNA transcription profile in two distinct macrophage populations in MetS-affected cardiac tissue. Although the results are preliminary, the presented data provide a foundation for further studies on intercellular cross-talk/molecular mechanism(s) involved in the regulation of MetS-related cardiac remodeling.
Collapse
Affiliation(s)
- Justyna Niderla-Bielińska
- Department of Histology and Embryology, Collegium Anatomicum, Medical University of Warsaw, 02-004 Warsaw, Poland; (J.N.-B.); (A.Ś.); (E.J.-S.)
| | - Aneta Ścieżyńska
- Department of Histology and Embryology, Collegium Anatomicum, Medical University of Warsaw, 02-004 Warsaw, Poland; (J.N.-B.); (A.Ś.); (E.J.-S.)
| | - Aneta Moskalik
- Postgraduate School of Molecular Medicine, Collegium Anatomicum, Medical University of Warsaw, 02-004 Warsaw, Poland;
| | - Ewa Jankowska-Steifer
- Department of Histology and Embryology, Collegium Anatomicum, Medical University of Warsaw, 02-004 Warsaw, Poland; (J.N.-B.); (A.Ś.); (E.J.-S.)
| | - Krzysztof Bartkowiak
- Student Scientific Group, Department of Histology and Embryology, Collegium Anatomicum, Medical University of Warsaw, 02-004 Warsaw, Poland; (K.B.); (M.B.)
| | - Mateusz Bartkowiak
- Student Scientific Group, Department of Histology and Embryology, Collegium Anatomicum, Medical University of Warsaw, 02-004 Warsaw, Poland; (K.B.); (M.B.)
- Department of History of Medicine, Medical University of Warsaw, 00-575 Warsaw, Poland
| | - Ewelina Kiernozek
- Department of Immunology, Faculty of Biology, University of Warsaw, 02-096 Warsaw, Poland;
| | - Anna Podgórska
- Molecular Biology Laboratory, Department of Medical Biology, Cardinal Stefan Wyszyński Institute of Cardiology, 04-628 Warsaw, Poland;
| | - Bogdan Ciszek
- Department of Clinical Anatomy, Collegium Anatomicum, Medical University of Warsaw, 02-004 Warsaw, Poland;
| | - Barbara Majchrzak
- Department of Pathology, Collegium Anatomicum, Medical University of Warsaw, 02-004 Warsaw, Poland;
| | - Anna Ratajska
- Department of Pathology, Collegium Anatomicum, Medical University of Warsaw, 02-004 Warsaw, Poland;
| |
Collapse
|
29
|
Wu Y, Li Q, Zhang R, Dai X, Chen W, Xing D. Circulating microRNAs: Biomarkers of disease. Clin Chim Acta 2021; 516:46-54. [PMID: 33485903 DOI: 10.1016/j.cca.2021.01.008] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 01/12/2021] [Accepted: 01/14/2021] [Indexed: 02/07/2023]
Abstract
MicroRNAs are a class of endogenous noncoding single-stranded RNA molecules with approximately 20-24 nucleotides and are associated with a broad range of biological processes. Researchers found that microRNAs are abundant in tissues, and more importantly, there are also trace circulating microRNAs that exist in biological fluids. In recent years, circulating microRNAs had emerged as promising diagnostic and prognostic biomarkers for the noninvasive detection of diseases with high specificity and sensitivity. More importantly, specific microRNA expression signatures reflect not only the existence of early-stage diseases but also the dynamic development of advanced-stage diseases, disease prognosis prediction, and drug resistance. To date, an increasing number of potential miRNA biomarkers have been reported, but their practical application prospects are still unclear. Therefore, microRNAs, as potential diagnostic and prognostic biomarkers in a variety of diseases, need to be updated, as they are of great importance in the diagnosis, prognosis and prediction of therapeutic responses. In this review, we summary our current understanding of microRNAs as potential biomarkers in the major diseases (e.g., cancers and cardio-cerebrovascular diseases), which provide the basis for the design of diagnosis and treatment plan and the improvement of the cure rate.
Collapse
Affiliation(s)
- Yudong Wu
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Qingdao Cancer Institute, Qingdao University, Qingdao 266071, China
| | - Qian Li
- Qingdao Cancer Institute, Qingdao University, Qingdao 266071, China
| | - Renshuai Zhang
- Qingdao Cancer Institute, Qingdao University, Qingdao 266071, China
| | - Xiaoli Dai
- Qingdao Cancer Institute, Qingdao University, Qingdao 266071, China
| | - Wujun Chen
- Qingdao Cancer Institute, Qingdao University, Qingdao 266071, China.
| | - Dongming Xing
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Qingdao Cancer Institute, Qingdao University, Qingdao 266071, China; School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
30
|
Videira RF, da Costa Martins PA, Falcão-Pires I. Non-Coding RNAs as Blood-Based Biomarkers in Cardiovascular Disease. Int J Mol Sci 2020; 21:ijms21239285. [PMID: 33291434 PMCID: PMC7730567 DOI: 10.3390/ijms21239285] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/28/2020] [Accepted: 11/30/2020] [Indexed: 12/12/2022] Open
Abstract
In 2020, cardiovascular diseases (CVDs) remain a leading cause of mortality and morbidity, contributing to the burden of the already overloaded health system. Late or incorrect diagnosis of patients with CVDs compromises treatment efficiency and patient's outcome. Diagnosis of CVDs could be facilitated by detection of blood-based biomarkers that reliably reflect the current condition of the heart. In the last decade, non-coding RNAs (ncRNAs) present on human biofluids including serum, plasma, and blood have been reported as potential biomarkers for CVDs. This paper reviews recent studies that focus on the use of ncRNAs as biomarkers of CVDs.
Collapse
Affiliation(s)
- Raquel Figuinha Videira
- CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6229 ER Maastricht, The Netherlands; (R.F.V.); (P.A.d.C.M.)
- Department of Molecular Genetics, Faculty of Science and Engineering, Maastricht University, 6229 ER Maastricht, The Netherlands
- Cardiovascular Research and Development Center, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Paula A. da Costa Martins
- CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6229 ER Maastricht, The Netherlands; (R.F.V.); (P.A.d.C.M.)
- Department of Molecular Genetics, Faculty of Science and Engineering, Maastricht University, 6229 ER Maastricht, The Netherlands
- Cardiovascular Research and Development Center, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Inês Falcão-Pires
- Cardiovascular Research and Development Center, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Correspondence:
| |
Collapse
|
31
|
Aleshcheva G, Pietsch H, Escher F, Schultheiss HP. MicroRNA profiling as a novel diagnostic tool for identification of patients with inflammatory and/or virally induced cardiomyopathies. ESC Heart Fail 2020; 8:408-422. [PMID: 33215881 PMCID: PMC7835602 DOI: 10.1002/ehf2.13090] [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: 03/10/2020] [Revised: 10/07/2020] [Accepted: 10/22/2020] [Indexed: 01/03/2023] Open
Abstract
AIMS MicroRNAs (miRNAs) might be used as prospective biomarkers for the identification of unexplained heart failure caused by a viral and/or inflammatory process. The aim of this study was to identify and to evaluate prognostic miRNAs in serum of patients with inflammatory heart diseases diagnosed by endomyocardial biopsies. METHODS AND RESULTS After TaqMan® OpenArray® screening of 754 unique circulating miRNAs in serum of biopsy-proven patients [184 patients with inflammatory and/or virally induced myocardial diseases (DCMi), 25 patients with dilated cardiomyopathy (DCM), and 25 healthy donors], we identified seven miRNAs of interest (P < 0.05). These data have been verified by single qRT-PCR assays in other biopsy-proven patients (159 patients with viral and/or inflammatory myocardial diseases, 46 patients with DCM, and 60 healthy donors). The expression of let-7f, miR-197, miR-223, miR-93, and miR-379 allowed us to differentiate between patients with a virus and/or inflammation and healthy donors (P < 0.05) with the specificity over 93%. Based on the expression of miR-21 and miR-30a-5p, we could sort out patients with DCM from all other study groups (P < 0.05) with the specificity over 95%. CONCLUSIONS This miRNA profile provides for the first time a new non-invasive diagnostic perspective to identify patients with intramyocardial inflammation and/or viral persistence only from single serum sample, independently of prescribed therapy and time of symptoms onset. It allows the early finding of those patients relevant for myocardial biopsy for exact diagnosis and further proscription of causal aetiology-driven specific treatment.
Collapse
Affiliation(s)
- Ganna Aleshcheva
- Institute for Cardiac Diagnostics and Therapy (IKDT), Berlin, 12203, Germany
| | - Heiko Pietsch
- Institute for Cardiac Diagnostics and Therapy (IKDT), Berlin, 12203, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Felicitas Escher
- Institute for Cardiac Diagnostics and Therapy (IKDT), Berlin, 12203, Germany.,Department of Cardiology, Campus Virchow, Charité - University Hospital Berlin, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | | |
Collapse
|
32
|
Lv XB, Niu QH, Zhang M, Feng L, Feng J. Critical functions of microRNA-30a-5p-E2F3 in cardiomyocyte apoptosis induced by hypoxia/reoxygenation. Kaohsiung J Med Sci 2020; 37:92-100. [PMID: 33058540 DOI: 10.1002/kjm2.12309] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 08/26/2020] [Accepted: 09/13/2020] [Indexed: 01/01/2023] Open
Abstract
The high-mortality rate of cardiovascular diseases (CVDs) is associated with the myocardial ischemia and reperfusion (I/R). Recent investigations have revealed that microRNAs (miRNAs) exert vital functions in the apoptosis of cardiomyocyte cell. Nevertheless, the potential role of miR-30a-5p in the regulation of cardiomyocyte cell apoptosis needs to be illuminated. In the current study, we observed that hypoxia/reoxygenation (H/R) remarkably raised the level of miR-30a-5p but reduced the expression of E2F transcription factor 3 (E2F3) in H9c2 cardiomyocytes. In vivo, miR-30a-5p was found to be significantly upregulated in the hearts of rats following I/R. Downregulation of miR-30a-5p using anti-miR-30a-5p decreased H9c2 cardiomyocytes apoptosis caused by H/R and promoted the proliferation of H9c2 inhibited by H/R. Moreover, E2F3 was a possible target gene of miR-30a-5p and upregulation of miR-30a-5p reduced the expression level of E2F3 in H9c2 cardiomyocytes. We further identified that E2F3 silencing reversed the effect of anti-miR-30a-5p on the proliferation and apoptosis in H/R treated H9c2 cells. These studies suggested that downregulation of miR-30a-5p attenuated the impact of H/R on H9c2 cardiomyocytes through targeting E2F3.
Collapse
Affiliation(s)
- Xiao-Bing Lv
- Department of Cardiology, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qing-Hui Niu
- Liver Disease Center, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Min Zhang
- Department of Geriatrics, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Li Feng
- Department of Cardiology, the Central Hospital of Lijin County, Dongying, China
| | - Jia Feng
- Department of Pediatrics, the Central Hospital of Shengli Oil Field, Dongying, China
| |
Collapse
|
33
|
Ambrosini S, Mohammed SA, Costantino S, Paneni F. Disentangling the epigenetic landscape in cardiovascular patients: a path toward personalized medicine. Minerva Cardiol Angiol 2020; 69:331-345. [PMID: 32996305 DOI: 10.23736/s2724-5683.20.05326-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Despite significant advances in our understanding of cardiovascular disease (CVD) we are still far from having developed breakthrough strategies to combat coronary atherosclerosis and heart failure, which account for most of CV deaths worldwide. Available cardiovascular therapies have failed to show to be equally effective in all patients, suggesting that inter-individual diversity is an important factor when it comes to conceive and deliver effective personalized treatments. Genome mapping has proved useful in identifying patients who could benefit more from specific drugs depending on genetic variances; however, our genetic make-up determines only a limited part of an individual's risk profile. Recent studies have demonstrated that epigenetic changes - defined as dynamic changes of DNA and histones which do not affect DNA sequence - are key players in the pathophysiology of cardiovascular disease and may participate to delineate cardiovascular risk trajectories over the lifetime. Epigenetic modifications include changes in DNA methylation, histone modifications and non-coding RNAs and these epigenetic signals have shown to cooperate in modulating chromatin accessibility to transcription factors and gene expression. Environmental factors such as air pollution, smoking, psychosocial context, and unhealthy diet regimens have shown to significantly modify the epigenome thus leading to altered transcriptional programs and CVD phenotypes. Therefore, the integration of genetic and epigenetic information might be invaluable to build individual maps of cardiovascular risk and hence, could be employed for the design of customized diagnostic and therapeutic strategies. In the present review, we discuss the growing importance of epigenetic information and its putative implications in cardiovascular precision medicine.
Collapse
Affiliation(s)
- Samuele Ambrosini
- Center for Molecular Cardiology, University of Zürich, Zurich, Switzerland
| | - Shafeeq A Mohammed
- Center for Molecular Cardiology, University of Zürich, Zurich, Switzerland
| | - Sarah Costantino
- Center for Molecular Cardiology, University of Zürich, Zurich, Switzerland
| | - Francesco Paneni
- Center for Molecular Cardiology, University of Zürich, Zurich, Switzerland - .,Department of Cardiology, University Heart Center, University Hospital Zurich, Zurich, Switzerland.,Department of Research and Education, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
34
|
Liu T, Zhang G, Wang Y, Rao M, Zhang Y, Guo A, Wang M. Identification of Circular RNA-MicroRNA-Messenger RNA Regulatory Network in Atrial Fibrillation by Integrated Analysis. BIOMED RESEARCH INTERNATIONAL 2020; 2020:8037273. [PMID: 33062700 PMCID: PMC7545447 DOI: 10.1155/2020/8037273] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 08/13/2020] [Accepted: 08/14/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Circular RNA (circRNA) is a noncoding RNA that forms a closed-loop structure, and its abnormal expression may cause disease. We aimed to find potential network for circRNA-related competitive endogenous RNA (ceRNA) in atrial fibrillation (AF). METHODS The circRNA, miRNA, and mRNA expression profiles in the heart tissue from AF patients were retrieved from the Gene Expression Omnibus database and analyzed comprehensively. Differentially expressed circRNAs (DEcircRNAs), differentially expressed miRNAs (DEmiRNAs), and differentially expressed mRNAs (DEmRNAs) were identified, followed by the establishment of DEcircRNA-DEmiRNA-DEmRNA regulatory network. Functional annotation analysis of host gene of DEcircRNAs and DEmRNAs in ceRNA regulatory network was performed. In vitro experiment and electronic validation were used to validate the expression of DEcircRNAs, DEmiRNAs, and DEmRNAs. RESULTS A total of 1611 DEcircRNAs, 51 DEmiRNAs, and 1250 DEmRNAs were identified in AF. The DEcircRNA-DEmiRNA-DEmRNA network contained 62 circRNAs, 14 miRNAs, and 728 mRNAs. Among which, two ceRNA regulatory pairs of hsa-circRNA-100053-hsa-miR-455-5p-TRPV1 and hsa-circRNA-005843-hsa-miR-188-5p-SPON1 were identified. In addition, six miRNA-mRNA regulatory pairs including hsa-miR-34c-5p-INMT, hsa-miR-1253-DDIT4L, hsa-miR-508-5p-SMOC2, hsa-miR-943-ACTA1, hsa-miR-338-3p-WIPI1, and hsa-miR-199a-3p-RAP1GAP2 were also obtained. MTOR was a significantly enriched signaling pathway of host gene of DEcircRNAs. In addition, arrhythmogenic right ventricular cardiomyopathy, dilated cardiomyopathy, and hypertrophic cardiomyopathy were remarkably enriched signaling pathways of DEmRNAs in DEcircRNA-DEmiRNA-DEmRNA regulatory network. The expression validation of hsa-circRNA-402565, hsa-miR-34c-5p, hsa-miR-188-5p, SPON1, DDIT4L, SMOC2, and WIPI1 was consistent with the integrated analysis. CONCLUSION We speculated that hsa-circRNA-100053-hsa-miR-455-5p-TRPV1 and hsa-circRNA-005843-hsa-miR-188-5p-SPON1 interaction pairs may be involved in AF.
Collapse
Affiliation(s)
- Tao Liu
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Guoru Zhang
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Yaling Wang
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Mingyue Rao
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Yang Zhang
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Anjun Guo
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Mei Wang
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| |
Collapse
|
35
|
Galeano-Otero I, Del Toro R, Guisado A, Díaz I, Mayoral-González I, Guerrero-Márquez F, Gutiérrez-Carretero E, Casquero-Domínguez S, Díaz-de la Llera L, Barón-Esquivias G, Jiménez-Navarro M, Smani T, Ordóñez-Fernández A. Circulating miR-320a as a Predictive Biomarker for Left Ventricular Remodelling in STEMI Patients Undergoing Primary Percutaneous Coronary Intervention. J Clin Med 2020; 9:E1051. [PMID: 32276307 PMCID: PMC7230612 DOI: 10.3390/jcm9041051] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 04/03/2020] [Accepted: 04/06/2020] [Indexed: 12/11/2022] Open
Abstract
Restoration of epicardial coronary blood flow, achieved by early reperfusion with primary percutaneous coronary intervention (PPCI), is the guideline recommended to treat patients with ST-segment-elevation myocardial infarction (STEMI). However, despite successful blood restoration, increasing numbers of patients develop left ventricular adverse remodelling (LVAR) and heart failure. Therefore, reliable prognostic biomarkers for LVAR in STEMI are urgently needed. Our aim was to investigate the role of circulating microRNAs (miRNAs) and their association with LVAR in STEMI patients following the PPCI procedure. We analysed the expression of circulating miRNAs in blood samples of 56 patients collected at admission and after revascularization (at 3, 6, 12 and 24 h). The associations between miRNAs and left ventricular end diastolic volumes at 6 months were estimated to detect LVAR. miRNAs were also analysed in samples isolated from peripheral blood mononuclear cells (PBMCs) and human myocardium of failing hearts. Kinetic analysis of miRNAs showed a fast time-dependent increase in miR-133a, miR-133b, miR-193b, miR-499, and miR-320a in STEMI patients compared to controls. Moreover, the expression of miR-29a, miR-29b, miR-324, miR-208, miR-423, miR-522, and miR-545 was differentially expressed even before PPCI in STEMI. Furthermore, the increase in circulating miR-320a and the decrease in its expression in PBMCs were significantly associated with LVAR and correlated with the expression of miR-320a in human failing myocardium from ischaemic origin. In conclusion, we determined the time course expression of new circulating miRNAs in patients with STEMI treated with PPCI and we showed that miR-320a was positively associated with LVAR.
Collapse
Affiliation(s)
- Isabel Galeano-Otero
- Departamento de Fisiología Médica y Biofísica, Universidad de Sevilla, 41009 Sevilla, Spain (R.D.T.)
- Grupo de Fisiopatología Cardiovascular, Instituto de Biomedicina de Sevilla-IBiS, Universidad de Sevilla/HUVR/Junta de Andalucía/CSIC, Sevilla 41013, CIBERCV, 28029 Madrid, Spain; (I.D.); (I.M.-G.); (E.G.-C.); (G.B.-E.)
| | - Raquel Del Toro
- Departamento de Fisiología Médica y Biofísica, Universidad de Sevilla, 41009 Sevilla, Spain (R.D.T.)
- Grupo de Fisiopatología Cardiovascular, Instituto de Biomedicina de Sevilla-IBiS, Universidad de Sevilla/HUVR/Junta de Andalucía/CSIC, Sevilla 41013, CIBERCV, 28029 Madrid, Spain; (I.D.); (I.M.-G.); (E.G.-C.); (G.B.-E.)
| | - Agustín Guisado
- Servicio de Cardiología, Hospital Universitario Virgen del Rocío, 41013 Sevilla, Spain; (A.G.); (F.G.-M.); (S.C.-D.)
| | - Ignacio Díaz
- Grupo de Fisiopatología Cardiovascular, Instituto de Biomedicina de Sevilla-IBiS, Universidad de Sevilla/HUVR/Junta de Andalucía/CSIC, Sevilla 41013, CIBERCV, 28029 Madrid, Spain; (I.D.); (I.M.-G.); (E.G.-C.); (G.B.-E.)
| | - Isabel Mayoral-González
- Grupo de Fisiopatología Cardiovascular, Instituto de Biomedicina de Sevilla-IBiS, Universidad de Sevilla/HUVR/Junta de Andalucía/CSIC, Sevilla 41013, CIBERCV, 28029 Madrid, Spain; (I.D.); (I.M.-G.); (E.G.-C.); (G.B.-E.)
| | - Francisco Guerrero-Márquez
- Servicio de Cardiología, Hospital Universitario Virgen del Rocío, 41013 Sevilla, Spain; (A.G.); (F.G.-M.); (S.C.-D.)
| | - Encarnación Gutiérrez-Carretero
- Grupo de Fisiopatología Cardiovascular, Instituto de Biomedicina de Sevilla-IBiS, Universidad de Sevilla/HUVR/Junta de Andalucía/CSIC, Sevilla 41013, CIBERCV, 28029 Madrid, Spain; (I.D.); (I.M.-G.); (E.G.-C.); (G.B.-E.)
- Servicio de Cardiología, Hospital Universitario Virgen del Rocío, 41013 Sevilla, Spain; (A.G.); (F.G.-M.); (S.C.-D.)
| | - Sara Casquero-Domínguez
- Servicio de Cardiología, Hospital Universitario Virgen del Rocío, 41013 Sevilla, Spain; (A.G.); (F.G.-M.); (S.C.-D.)
| | - Luis Díaz-de la Llera
- Servicio de Cardiología, Hospital Universitario Virgen del Rocío, 41013 Sevilla, Spain; (A.G.); (F.G.-M.); (S.C.-D.)
| | - Gonzalo Barón-Esquivias
- Grupo de Fisiopatología Cardiovascular, Instituto de Biomedicina de Sevilla-IBiS, Universidad de Sevilla/HUVR/Junta de Andalucía/CSIC, Sevilla 41013, CIBERCV, 28029 Madrid, Spain; (I.D.); (I.M.-G.); (E.G.-C.); (G.B.-E.)
- Servicio de Cardiología, Hospital Universitario Virgen del Rocío, 41013 Sevilla, Spain; (A.G.); (F.G.-M.); (S.C.-D.)
| | - Manuel Jiménez-Navarro
- Hospital Universitario Virgen de la Victoria, Málaga 29010, CIBERCV, 28029 Madrid, Spain;
| | - Tarik Smani
- Departamento de Fisiología Médica y Biofísica, Universidad de Sevilla, 41009 Sevilla, Spain (R.D.T.)
- Grupo de Fisiopatología Cardiovascular, Instituto de Biomedicina de Sevilla-IBiS, Universidad de Sevilla/HUVR/Junta de Andalucía/CSIC, Sevilla 41013, CIBERCV, 28029 Madrid, Spain; (I.D.); (I.M.-G.); (E.G.-C.); (G.B.-E.)
| | - Antonio Ordóñez-Fernández
- Grupo de Fisiopatología Cardiovascular, Instituto de Biomedicina de Sevilla-IBiS, Universidad de Sevilla/HUVR/Junta de Andalucía/CSIC, Sevilla 41013, CIBERCV, 28029 Madrid, Spain; (I.D.); (I.M.-G.); (E.G.-C.); (G.B.-E.)
- Servicio de Cardiología, Hospital Universitario Virgen del Rocío, 41013 Sevilla, Spain; (A.G.); (F.G.-M.); (S.C.-D.)
| |
Collapse
|
36
|
Kiliszek M, Maciak K, Maciejak A, Krzyżanowski K, Wierzbowski R, Gora M, Burzynska B, Segiet A, Skrobowski A. Serum microRNA in patients undergoing atrial fibrillation ablation. Sci Rep 2020; 10:4424. [PMID: 32157193 PMCID: PMC7064599 DOI: 10.1038/s41598-020-61322-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 01/29/2020] [Indexed: 12/13/2022] Open
Abstract
MicroRNAs mediate posttranscriptional gene regulation. The aim of the study was to find a microRNA predictor of successful atrial fibrillation (AF) ablation. A total of 109 patients undergoing first-time AF ablation were included. Nineteen patients were selected to undergo serum microRNA sequencing (study group). The sequencing data were used to select several microRNAs that correlated with 12-month recurrences after AF ablation. Those microRNAs were validated by digital droplet PCR in samples from remaining 90 patients. All patients underwent pulmonary vein isolation (RF ablation, contact force catheter, electroanatomical system). The endpoint of the study was the 12-month AF recurrence rate; the overall recurrence rate was 42.5%. In total, levels of 34 miRNAs were significantly different in sera from patients with AF recurrence compared to patients without AF recurrence. Six microRNAs (miR-183-5p, miR-182-5p, miR-32-5p, miR-107, miR-574-3p, and miR-144-3p) were validated in the whole group. Data from the validation group did not confirm the observations from the study group, as no significant differences were found between miRNAs serum levels in patients with and without recurrences 12 months after AF ablation.
Collapse
Affiliation(s)
- Marek Kiliszek
- Department of Cardiology and Internal Diseases, Military Institute of Medicine, Warsaw, Poland.
| | - Karolina Maciak
- Institute of Biochemistry and Biophysics, PAS, Warsaw, Poland
| | - Agata Maciejak
- Department of Clinical Chemistry and Laboratory Diagnostics, Medical University of Warsaw, Warsaw, Poland
| | - Krystian Krzyżanowski
- Department of Cardiology and Internal Diseases, Military Institute of Medicine, Warsaw, Poland
| | - Robert Wierzbowski
- Department of Cardiology and Internal Diseases, Military Institute of Medicine, Warsaw, Poland
| | - Monika Gora
- Institute of Biochemistry and Biophysics, PAS, Warsaw, Poland
| | - Beata Burzynska
- Institute of Biochemistry and Biophysics, PAS, Warsaw, Poland
| | - Agnieszka Segiet
- Chair and Department of Experimental and Clinical Physiology, Medical University of Warsaw, Warsaw, Poland
| | - Andrzej Skrobowski
- Department of Cardiology and Internal Diseases, Military Institute of Medicine, Warsaw, Poland
| |
Collapse
|
37
|
Zhang C, Gan X, Liang R, Jian J. Exosomes Derived From Epigallocatechin Gallate-Treated Cardiomyocytes Attenuated Acute Myocardial Infarction by Modulating MicroRNA-30a. Front Pharmacol 2020; 11:126. [PMID: 32161548 PMCID: PMC7054242 DOI: 10.3389/fphar.2020.00126] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Accepted: 01/29/2020] [Indexed: 12/15/2022] Open
Abstract
Background Ischemia-derived exosomes can restrict excessive autophagy by transferring microRNA-30a (miR30a) to cells. Reports have confirmed that epigallocatechin gallate (EGCG) alleviates acute myocardial infarction (AMI) by regulating autophagy; however, research evaluating the communication with cardiomyocytes and exosomes is lacking. This study aimed to explore whether exosomes derived from EGCG-treated cardiomyocytes mitigated AMI by adjusting miR30a to inactivate apoptosis and autophagy. Methods Exosomes were extracted from cardiomyocytes, cultured either in control or AMI condition, with or without EGCG pretreatment. The exosome characteristics were analyzed by nanoparticle tracking analyses and transmission electron microscopy. The change in miR30a in cells and exosomes was demonstrated by qRT-PCR. H9c2 or stable miR30a knockdown (miR30aKD) cell lines were incubated with exosomes derived from EGCG-treated cardiomyocytes in vitro or in vivo. The effect of EGCG and exosomes on I/R-induced cardiomyocyte apoptosis and autophagy was assessed. Results EGCG improved the activity of cardiomyocytes, and increased average diameter, concentration, miR30a mRNA level, and specific protein expression in AMI-derived exosomes produced by cardiomyocytes. Moreover, the coincubation of AMI cells with EGCG or exosomes derived from EGCG-treated cardiomyocytes attenuated cardiomyocyte apoptosis and autophagy. Conclusions The findings showed that EGCG upregulates miR30a, which was efficiently transferred via exosomes between cardiomyocytes, thereby contributing to the suppression of apoptosis and autophagy. By focusing on the cardiomyocyte microenvironment, we identified a new target of EGCG alleviating AMI by regulating apoptosis and autophagy.
Collapse
Affiliation(s)
- Chan Zhang
- Department of Pharmacology, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaowen Gan
- Department of Pharmacology, Guilin Medical University, Guilin, China
| | - Ronggan Liang
- Department of Pharmacology, Guilin Medical University, Guilin, China
| | - Jie Jian
- Department of Pharmacology, Guilin Medical University, Guilin, China
| |
Collapse
|
38
|
Kwak YH, Kwak DK, Kim NY, Kim YJ, Lim JS, Yoo JH. Significant changes in synovial fluid microRNAs after high tibial osteotomy in medial compartmental knee osteoarthritis: Identification of potential prognostic biomarkers. PLoS One 2020; 15:e0227596. [PMID: 31923920 PMCID: PMC6954069 DOI: 10.1371/journal.pone.0227596] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 12/24/2019] [Indexed: 01/03/2023] Open
Abstract
High tibial osteotomy (HTO) is a well-established treatment for medial compartmental knee osteoarthritis. Several microRNAs (miRNAs) are involved in osteoarthritis progression and are useful as osteoarthritis-related biomarkers. In this prospective study, we investigated differentially expressed microRNAs in the synovial fluid (SF) before and after HTO in patients with medial compartmental knee osteoarthritis to identify microRNAs that can be used as prognostic biomarkers. We used miRNA-PCR arrays to screen for miRNAs in SF samples obtained preoperatively and 6 months postoperatively from 6 patients with medial compartmental knee osteoarthritis who were treated with medial open wedge HTO. Differentially expressed miRNAs identified in the profiling stage were validated by real-time quantitative PCR in 22 other patients who had also been treated with HTO. All patients radiographically corresponded to Kellgren-Lawrence grade II or III with medial compartmental osteoarthritis. These patients were clinically assessed using a visual analogue scale and Western Ontario McMaster Universities scores. Mechanical axis changes were measured on standing anteroposterior radiographs of the lower limbs assessed preoperatively and at 6 months postoperatively. Among 84 miRNAs known to be involved in the inflammatory process, 14 were expressed in all SF specimens and 3 (miR-30a-5p, miR-29a-3p, and miR-30c-5p) were differentially expressed in the profiling stage. These 3 miRNAs, as well as 4 other miRNAs (miR-378a-5p, miR-140-3p, miR-23a-3p, miR-27b-3p), are related to osteoarthritis progression. These results were validated in the SF from 22 patients. Clinical and radiological outcomes improved after HTO in all patients, and only 2 miRNAs (miR-30c-5p and miR-23a-3p) were significantly differentially expressed between preoperative and postoperative 6-month SF samples (p = 0.006 and 0.007, respectively). Of these two miRNAs, miR-30c-5p correlated with postoperative pain relief. This study provides potential prognostic miRNAs after HTO and further investigations should be considered to determine clinical implications of these miRNAs.
Collapse
Affiliation(s)
- Yoon Hae Kwak
- Department of Orthopaedic Surgery, Severance Children’s Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Dae-Kyung Kwak
- Department of Orthopaedic Surgery, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, Republic of Korea
| | - Nan Young Kim
- Hallym Institute of Translational Genomics & Bioinformatics, Hallym University Medical Center, Anyang, Republic of Korea
| | - Yun Joong Kim
- Hallym Institute of Translational Genomics & Bioinformatics, Hallym University Medical Center, Anyang, Republic of Korea
- Ilsong Institute of Life Science, Hallym University, Anyang, Republic of Korea
- Department of Neurology, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, Republic of Korea
| | - Jeong Seop Lim
- Department of Orthopaedic Surgery, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, Republic of Korea
| | - Je-Hyun Yoo
- Department of Orthopaedic Surgery, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, Republic of Korea
| |
Collapse
|
39
|
Weale CJ, Matshazi DM, Davids SFG, Raghubeer S, Erasmus RT, Kengne AP, Davison GM, Matsha TE. Circulating miR-30a-5p and miR-182-5p in Prediabetes and Screen-Detected Diabetes Mellitus. Diabetes Metab Syndr Obes 2020; 13:5037-5047. [PMID: 33376373 PMCID: PMC7762450 DOI: 10.2147/dmso.s286081] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 11/26/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND microRNAs (miRNAs) have been touted as potential diagnostic and prognostic biomarkers for various diseases. The aim of the present study was to evaluate the diagnostic value of miR-30a-5p and miR-182-5p for prediabetes and screen-detected type 2 diabetes mellitus (T2DM). METHODS The study included 1270 participants (207 prediabetes, 94 screen-detected diabetes and 969 normotolerant) from the Vascular and Metabolic Health (VMH) study. Whole blood levels of miR-30a-5p and miR-182-5p were quantitated by RT-qPCR. Multivariable logistic regressions were used to relate miRNAs with prediabetes or T2DM and receiver operating characteristic (ROC) curves were used to evaluate the ability of each miRNA to diagnose these conditions. RESULTS Both miRNAs were significantly highly expressed in individuals with prediabetes or T2DM (both ≥3.2-fold, and p<0.001). We also observed significant under-expression in T2DM relative to prediabetes for miR-182-5p (0.49-fold, p=0.001). Age, sex and BMI-adjusted partial correlation coefficient analysis revealed a significant correlation between the two miRNAs across glucose tolerance statuses (r≥0.932, p<0.001). In normotolerant individuals, both miRNAs showed a negative correlation with waist circumference and positive correlation with HDL-cholesterol whilst in T2DM they correlated positively with hip circumference, 2-hour insulin, HDL- and LDL-cholesterol. Multivariable logistic regressions revealed both miRNAs to be consistently and continuously associated with prediabetes or T2DM (OR≥1.18, 95% 95% CI: 1.10-1.28, p<0.001), while only miR-182-5p associated with a reduced prevalence of T2DM relative to prediabetes (OR: 0.89, 95% CI: 0.83-0.96, p=0.003). In ROC analyses, miR-182-5p almost outperformed HbA1c in diagnosing prediabetes; area under the curve 0.74 vs 0.69. CONCLUSION Our findings demonstrate that miR-30a-5p and miR-182-5p are associated with dysglycaemia and could potentially predict prediabetes, particularly miR-182-5p.
Collapse
Affiliation(s)
- Cecil Jack Weale
- SAMRC/CPUT/Cardiometabolic Health Research Unit, Department of Biomedical Sciences, Faculty of Health and Wellness Science, Cape Peninsula University of Technology, Cape Town, South Africa
| | - Don M Matshazi
- SAMRC/CPUT/Cardiometabolic Health Research Unit, Department of Biomedical Sciences, Faculty of Health and Wellness Science, Cape Peninsula University of Technology, Cape Town, South Africa
| | - Saarah F G Davids
- SAMRC/CPUT/Cardiometabolic Health Research Unit, Department of Biomedical Sciences, Faculty of Health and Wellness Science, Cape Peninsula University of Technology, Cape Town, South Africa
| | - Shanel Raghubeer
- SAMRC/CPUT/Cardiometabolic Health Research Unit, Department of Biomedical Sciences, Faculty of Health and Wellness Science, Cape Peninsula University of Technology, Cape Town, South Africa
| | - Rajiv T Erasmus
- Division of Chemical Pathology, Faculty of Health Sciences, National Health Laboratory Service (NHLS) and University of Stellenbosch, Cape Town, South Africa
| | - Andre Pascal Kengne
- Non-Communicable Diseases Research Unit, South African Medical Research Council, Cape Town, South Africa
- Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Glenda Mary Davison
- SAMRC/CPUT/Cardiometabolic Health Research Unit, Department of Biomedical Sciences, Faculty of Health and Wellness Science, Cape Peninsula University of Technology, Cape Town, South Africa
| | - Tandi E Matsha
- SAMRC/CPUT/Cardiometabolic Health Research Unit, Department of Biomedical Sciences, Faculty of Health and Wellness Science, Cape Peninsula University of Technology, Cape Town, South Africa
- Correspondence:Tandi E Matsha Department of Biomedical Sciences, Faculty of Health and Wellness Science, Cape Peninsula University of Technology, Bellville, Cape Town7530, South AfricaTel +27 21 959 6366Fax +27 21 959 6760 Email
| |
Collapse
|
40
|
Zhang H, Wang Y, Yang G, Yu H, Zhou Z, Tang M. MicroRNA-30a regulates chondrogenic differentiation of human bone marrow-derived mesenchymal stem cells through targeting Sox9. Exp Ther Med 2019; 18:4689-4697. [PMID: 31807153 PMCID: PMC6878886 DOI: 10.3892/etm.2019.8148] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 07/05/2019] [Indexed: 12/20/2022] Open
Abstract
Cartilage injury is difficult to repair since the cartilage tissue lacks self-restoration ability. Improved formation of chondrocytes differentiated from the mesenchymal stem cells (MSC) by genetic regulation is a potentially promising therapeutic option. SOX9 is a critical transcription factor for mesenchymal condensation prior to chondrogenesis. Previous studies demonstrated that several microRNAs (miRNAs or miRs) play a critical role in the chondrogenic differentiation of MSCs. However, the interactional relations between miR-30a and SOX9 during chondrogenic differentiation of MSCs need to be further elucidated. In the present study, human bone marrow-derived mesenchymal stem cells have been isolated and induced into chondrogenic differentiation to imitate the cartilage formation in vitro. Additionally, the expression levels of several miRNAs that were reported to interact with the SOX9 3'untranslated region (UTR) were examined by using reverse transcription-quantitative PCR. The interactional relations between candidate miRNAs and SOX9 were verified with the transfection of a miRNA mimic or inhibitor and a luciferase reporter gene assay. The results indicate that miR-30a and miR-195 were consistently increased during MSC chondrogenic differentiation. Additionally, the binding of miR-30a to the SOX9 3UTR was verified. Then, the authors upregulated the expression of miR-30a and found that MSC chondrogenic differentiation was inhibited. Taken together, the results of the present study demonstrate that miR-30a has a negative regulatory effect on MSC chondrogenic differentiation by targeting SOX9. Advances in epigenetic regulating methods will likely be the future of systemic treatment of cartilage injury.
Collapse
Affiliation(s)
- Hongqi Zhang
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Yunjia Wang
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Guanteng Yang
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Honggui Yu
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Zhenhai Zhou
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Mingxing Tang
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
41
|
Soler-Botija C, Gálvez-Montón C, Bayés-Genís A. Epigenetic Biomarkers in Cardiovascular Diseases. Front Genet 2019; 10:950. [PMID: 31649728 PMCID: PMC6795132 DOI: 10.3389/fgene.2019.00950] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 09/05/2019] [Indexed: 12/16/2022] Open
Abstract
Cardiovascular diseases are the number one cause of death worldwide and greatly impact quality of life and medical costs. Enormous effort has been made in research to obtain new tools for efficient and quick diagnosis and predicting the prognosis of these diseases. Discoveries of epigenetic mechanisms have related several pathologies, including cardiovascular diseases, to epigenetic dysregulation. This has implications on disease progression and is the basis for new preventive strategies. Advances in methodology and big data analysis have identified novel mechanisms and targets involved in numerous diseases, allowing more individualized epigenetic maps for personalized diagnosis and treatment. This paves the way for what is called pharmacoepigenetics, which predicts the drug response and develops a tailored therapy based on differences in the epigenetic basis of each patient. Similarly, epigenetic biomarkers have emerged as a promising instrument for the consistent diagnosis and prognosis of cardiovascular diseases. Their good accessibility and feasible methods of detection make them suitable for use in clinical practice. However, multicenter studies with a large sample population are required to determine with certainty which epigenetic biomarkers are reliable for clinical routine. Therefore, this review focuses on current discoveries regarding epigenetic biomarkers and its controversy aiming to improve the diagnosis, prognosis, and therapy in cardiovascular patients.
Collapse
Affiliation(s)
- Carolina Soler-Botija
- Heart Failure and Cardiac Regeneration (ICREC) Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Badalona, Spain
- CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Carolina Gálvez-Montón
- Heart Failure and Cardiac Regeneration (ICREC) Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Badalona, Spain
- CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Antoni Bayés-Genís
- Heart Failure and Cardiac Regeneration (ICREC) Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Badalona, Spain
- CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
- Cardiology Service, HUGTiP, Badalona, Spain
- Department of Medicine, Barcelona Autonomous University (UAB), Badalona, Spain
| |
Collapse
|
42
|
Liu Y, Qian XM, He QC, Weng JK. MiR-421 inhibition protects H9c2 cells against hypoxia/reoxygenation-induced oxidative stress and apoptosis by targeting Sirt3. Perfusion 2019; 35:255-262. [PMID: 31469043 DOI: 10.1177/0267659119870725] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND MicroRNAs (miRNAs) are involved in myocardial ischemia-reperfusion injury. miRNA-421 (miR-421) plays a significant role in the initiation of apoptosis and myocardial infarction. However, the molecular regulation of miR-421 in myocardial ischemia-reperfusion injury requires further elucidation. METHODS An in vitro hypoxia/reoxygenation model was established, and the expression levels of miR-421 and Sirtuin-3 (Sirt3) in H9c2 cells were quantified using quantitative real-time polymerase chain reaction. Flow cytometry was employed to measure the effects of miR-421 on myocardial apoptosis induced by hypoxia/reoxygenation. The activity of lactate dehydrogenase and superoxide dismutase and levels of malondialdehyde were measured. The binding sites of miR-421 on Sirt3 were predicted using TargetScan software. A luciferase reporter assay was used to validate the direct targeting of Sirt3 with miR-421. Protein expression levels of Sirt3 and its downstream proteins were evaluated using Western blot analysis. RESULTS Exposure of H9c2 cells to hypoxia/reoxygenation led to increased apoptosis, levels of malondialdehyde and lactate dehydrogenase, and decreased levels of superoxide dismutase. miR-421 knockdown resulted in decreased apoptosis, levels of lactate dehydrogenase and malondialdehyde, and increased superoxide dismutase levels in H9c2 cells. Hypoxia/reoxygenation significantly decreased the relative expression levels of Sirt3. Down-regulation of Sirt3 resulted from overexpression of miR-421, which directly targeted Sirt3. Knockdown of miR-421 up-regulated Sirt3 expression, inhibited activation of the Jun N-terminal kinase/activator protein 1 pathway and caspase 9/3-dependent cell death. CONCLUSION The miR-421-Sirt3-Jun N-terminal kinase/activator protein 1 axis is a novel molecular mechanism that accommodates hypoxia/reoxygenation-induced oxidative stress and apoptosis and provides a new direction for the study and treatment of hypoxia/reoxygenation.
Collapse
Affiliation(s)
- Yu Liu
- Department of Cardiac Surgery, Sir Run Run Shaw Hospital Affiliated to Medical College of Zhejiang University, Hangzhou, P.R. China
| | - Xi-Ming Qian
- Department of Cardiac Surgery, Sir Run Run Shaw Hospital Affiliated to Medical College of Zhejiang University, Hangzhou, P.R. China
| | - Qi-Cai He
- Department of Cardiac Surgery, Sir Run Run Shaw Hospital Affiliated to Medical College of Zhejiang University, Hangzhou, P.R. China
| | - Jia-Kan Weng
- Department of Cardiac Surgery, Sir Run Run Shaw Hospital Affiliated to Medical College of Zhejiang University, Hangzhou, P.R. China
| |
Collapse
|
43
|
Sun Y, Yang B, Lin M, Yu H, Chen H, Zhang Z. Identification of serum miR-30a-5p as a diagnostic and prognostic biomarker in colorectal cancer. Cancer Biomark 2019; 24:299-305. [PMID: 30829615 DOI: 10.3233/cbm-182129] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Circulating microRNAs (miRNAs) have become increasingly appreciated in the diagnosis and prognosis of colorectal cancer (CRC). OBJECTIVE The aim of this study was to assess the potential diagnostic and prognostic significance of serum miR-30a-5p as a potential biomarker. METHODS The expression levels of serum miR-30a-5p were measured in 138 cases with CRC, 50 cases with benign lesions (colorectal adenoma and polyps) and 60 healthy volunteers by quantitative real time polymerase chain reaction (qRT-PCR). RESULTS The results showed that serum miR-30a-5p levels were frequently downregulated in patients with CRC and benign lesions in comparison with normal controls. Moreover, serum miR-30a-5p levels in early-stage CRC patients were significantly increased after surgery. Receiver-operating characteristic (ROC) curve analysis demonstrated serum miR-30a-5p could well distinguish CRC patients, early-stage CRC patients from healthy controls with a relative high value of area under the curve (AUC). Furthermore, low serum miR-30a-5p expression was more frequently occurred in CRC patients with aggressive clinical variables. Additionally, CRC patients exhibiting high serum miR-30a-5p expression had significantly prolonged overall survival than those exhibiting low expression. Finally, both univariate and multivariate analyses revealed that serum miR-30a-5p expression was an independent prognostic factor for overall survival in CRC patients. CONCLUSIONS Collectively, these findings suggested serum miR-30a-5p might act as a novel biomarker for the diagnosis and prognosis of CRC.
Collapse
Affiliation(s)
- Yi Sun
- Department of Gastroenterology, Jiangsu Taizhou People's Hospital, Taizhou, Jiangsu, China
| | - Bin Yang
- Department of Gastroenterology, Jiangsu Taizhou People's Hospital, Taizhou, Jiangsu, China
| | - Maosong Lin
- Department of Gastroenterology, Jiangsu Taizhou People's Hospital, Taizhou, Jiangsu, China
| | - Hong Yu
- Department of Pathology, Jiangsu Taizhou People's Hospital, Taizhou, Jiangsu, China
| | - Hui Chen
- Department of Gastroenterology, Jiangsu Taizhou People's Hospital, Taizhou, Jiangsu, China
| | - Zhenyu Zhang
- Department of Gastroenterology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
44
|
Colpaert RMW, Calore M. MicroRNAs in Cardiac Diseases. Cells 2019; 8:E737. [PMID: 31323768 PMCID: PMC6678080 DOI: 10.3390/cells8070737] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 07/08/2019] [Accepted: 07/16/2019] [Indexed: 12/13/2022] Open
Abstract
Since their discovery 20 years ago, microRNAs have been related to posttranscriptional regulation of gene expression in major cardiac physiological and pathological processes. We know now that cardiac muscle phenotypes are tightly regulated by multiple noncoding RNA species to maintain cardiac homeostasis. Upon stress or various pathological conditions, this class of non-coding RNAs has been found to modulate different cardiac pathological conditions, such as contractility, arrhythmia, myocardial infarction, hypertrophy, and inherited cardiomyopathies. This review summarizes and updates microRNAs playing a role in the different processes underlying the pathogenic phenotypes of cardiac muscle and highlights their potential role as disease biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Robin M W Colpaert
- IMAiA-Institute for Molecular Biology and RNA Technology, Faculty of Science and Engineering, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Martina Calore
- IMAiA-Institute for Molecular Biology and RNA Technology, Faculty of Science and Engineering, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6229 ER Maastricht, The Netherlands.
| |
Collapse
|
45
|
Huang C, Wang Q, Ma S, Sun Y, Vadamootoo AS, Jin C. A four serum-miRNA panel serves as a potential diagnostic biomarker of osteosarcoma. Int J Clin Oncol 2019; 24:976-982. [PMID: 31111286 DOI: 10.1007/s10147-019-01433-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Accepted: 03/22/2019] [Indexed: 01/10/2023]
Abstract
BACKGROUND Osteosarcoma (OS) is the most common malignant bone tumor in young adults and adolescents with approximately 3 million new cases annually. Due to the lack of sensitive and specific diagnostic biomarkers, although OS patients are curable after surgical resection, many patients suffer from metastasis or recurrence. This study aimed to investigate whether circulating microRNAs (miRNAs) could serve as biomarkers for the diagnosis of OS. MATERIALS AND METHODS Healthy individuals and OS patients enrolled in this study came from Nanjing First Hospital. First, candidate miRNAs were selected by integrated analysis of two GEO datasets and a publicly available miRNA dataset. The expression of these miRNAs in tissues and serum samples were subsequently examined through qRT-PCR. The diagnostic utility of these differential miRNAs was examined by using receiver operating characteristic (ROC) curve analysis. Finally, the potential signaling pathways associated with candidate miRNAs were searched through online tools. RESULTS Four miRNAs (miR-487a, miR-493-5p, miR-501-3p and miR-502-5p) were selected to further investigate their diagnostic potential for OS. We discovered miR-487a, miR-493-5p, miR-501-3p and miR-502-5p were upregulated in OS tissues and serums. Besides, miR-487a, miR-493-5p, miR-501-3p and miR-502-5p in peripheral blood of OS patients were tumor-derived. The area under the ROC curve (AUC) was 0.83 (95% CI 0.71-0.97) for miR-487a, 0.79 (95% CI 0.66-0.93) for miR-493-5p, 0.82 (95% CI 0.68-0.95) for miR-501-3p, 0.83 (95% CI 0.72-0.95) for miR-502-5p, and 0.89 (95% CI 0.78-1.0) for miRNAs combination. CONCLUSION Circulating miR-487a, miR-493-5p, miR-501-3p and miR-502-5p were novel potential diagnostic biomarkers of OS.
Collapse
Affiliation(s)
- Congwei Huang
- Department of Orthopedic, Nanjing First Hospital, Nanjing Medical University, No.68, Changle Road, Nanjing, Jiangsu, China
| | - Qinjue Wang
- Department of Orthopedic, Nanjing First Hospital, Nanjing Medical University, No.68, Changle Road, Nanjing, Jiangsu, China
| | - Shengshan Ma
- Department of Orthopedic, Lianyungang Hospital Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yang Sun
- Department of Orthopedic, Nanjing First Hospital, Nanjing Medical University, No.68, Changle Road, Nanjing, Jiangsu, China
| | - Ashley Shane Vadamootoo
- Department of Orthopedic, Nanjing First Hospital, Nanjing Medical University, No.68, Changle Road, Nanjing, Jiangsu, China
| | - Chengzhe Jin
- Department of Orthopedic, Nanjing First Hospital, Nanjing Medical University, No.68, Changle Road, Nanjing, Jiangsu, China.
| |
Collapse
|