1
|
Kumar Maji R, Fischer A, Rogg EM, Möller M, Gasparoni G, Simon M, Dimmeler S, Schulz MH. A cell type-specific expression atlas of small and total RNA in the heart after myocardial infarction. Sci Data 2025; 12:816. [PMID: 40389446 PMCID: PMC12089340 DOI: 10.1038/s41597-025-05061-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 04/24/2025] [Indexed: 05/21/2025] Open
Abstract
Acute myocardial infarction (AMI) is a leading cause of mortality worldwide. MicroRNAs (miRNAs), among other small non-coding RNAs, shape the transcriptome and control cellular functions. Although single-cell technologies are now available to study myocardial ischemia response, the study of small RNA regulation is limited by depth of expression, capture efficiency and lack of full coverage of transcripts. In addition, the kinetic expression of miRNAs is unknown. Using paired small and total RNA sequencing, we built an expression atlas to study the temporal dynamics of miRNAs and genes in four major heart cell types after AMI. Expression dynamics reveal enriched functions highlighting cell type-specific AMI stress responses. Many deregulated mouse genes after AMI overlap with known human cardiovascular disease genes. The dataset is highly valuable for additional research on small and long non-coding RNAs, such as regulation of RNA variants by splicing or alternative ORFs. All in all, the RNA expression atlas provides a useful resource to study different roles of RNAs in major cell types of the heart after AMI.
Collapse
Affiliation(s)
- Ranjan Kumar Maji
- Institute for Cardiovascular Regeneration, Goethe-University; German Centre for Cardiovascular Research (DZHK), Partner site Rhine-Main, 60590, Frankfurt, Germany
- Institute for Computational Genomic Medicine, Goethe University Frankfurt, 60590, Frankfurt, Germany
| | - Ariane Fischer
- Institute for Cardiovascular Regeneration, Goethe-University; German Centre for Cardiovascular Research (DZHK), Partner site Rhine-Main, 60590, Frankfurt, Germany
| | - Eva-Maria Rogg
- Institute for Cardiovascular Regeneration, Goethe-University; German Centre for Cardiovascular Research (DZHK), Partner site Rhine-Main, 60590, Frankfurt, Germany
- Immundiagnostik AG, 64625, Bensheim, Germany
| | - Melanie Möller
- Molecular Cell Biology & Microbiology, Wuppertal University, Wuppertal, Germany
| | | | - Martin Simon
- Molecular Cell Biology & Microbiology, Wuppertal University, Wuppertal, Germany
| | - Stefanie Dimmeler
- Institute for Cardiovascular Regeneration, Goethe-University; German Centre for Cardiovascular Research (DZHK), Partner site Rhine-Main, 60590, Frankfurt, Germany.
| | - Marcel H Schulz
- Institute for Cardiovascular Regeneration, Goethe-University; German Centre for Cardiovascular Research (DZHK), Partner site Rhine-Main, 60590, Frankfurt, Germany.
- Institute for Computational Genomic Medicine, Goethe University Frankfurt, 60590, Frankfurt, Germany.
| |
Collapse
|
2
|
Abubakar M, Hajjaj M, Naqvi ZEZ, Shanawaz H, Naeem A, Padakanti SSN, Bellitieri C, Ramar R, Gandhi F, Saleem A, Abdul Khader AHS, Faraz MA. Non-Coding RNA-Mediated Gene Regulation in Cardiovascular Disorders: Current Insights and Future Directions. J Cardiovasc Transl Res 2024; 17:739-767. [PMID: 38092987 DOI: 10.1007/s12265-023-10469-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 11/23/2023] [Indexed: 09/04/2024]
Abstract
Cardiovascular diseases (CVDs) pose a significant burden on global health. Developing effective diagnostic, therapeutic, and prognostic indicators for CVDs is critical. This narrative review explores the role of select non-coding RNAs (ncRNAs) and provides an in-depth exploration of the roles of miRNAs, lncRNAs, and circRNAs in different aspects of CVDs, offering insights into their mechanisms and potential clinical implications. The review also sheds light on the diverse functions of ncRNAs, including their modulation of gene expression, epigenetic modifications, and signaling pathways. It comprehensively analyzes the interplay between ncRNAs and cardiovascular health, paving the way for potential novel interventions. Finally, the review provides insights into the methodologies used to investigate ncRNA-mediated gene regulation in CVDs, as well as the implications and challenges associated with translating ncRNA research into clinical applications. Considering the broader implications, this research opens avenues for interdisciplinary collaborations, enhancing our understanding of CVDs across scientific disciplines.
Collapse
Affiliation(s)
- Muhammad Abubakar
- Department of Internal Medicine, Ameer-Ud-Din Medical College, Lahore General Hospital, Lahore, Punjab, Pakistan.
| | - Mohsin Hajjaj
- Department of Internal Medicine, Jinnah Hospital, Lahore, Punjab, Pakistan
| | - Zil E Zehra Naqvi
- Department of Internal Medicine, Jinnah Hospital, Lahore, Punjab, Pakistan
| | - Hameed Shanawaz
- Department of Internal Medicine, Windsor University School of Medicine, Cayon, Saint Kitts and Nevis
| | - Ammara Naeem
- Department of Cardiology, Heart & Vascular Institute, Dearborn, Michigan, USA
| | | | | | - Rajasekar Ramar
- Department of Internal Medicine, Rajah Muthiah Medical College, Chidambaram, Tamil Nadu, India
| | - Fenil Gandhi
- Department of Family Medicine, Lower Bucks Hospital, Bristol, PA, USA
| | - Ayesha Saleem
- Department of Internal Medicine, Jinnah Hospital, Lahore, Punjab, Pakistan
| | | | - Muhammad Ahmad Faraz
- Department of Forensic Medicine, Postgraduate Medical Institute, Lahore, Punjab, Pakistan
| |
Collapse
|
3
|
Silva ED, Pereira-Sousa D, Ribeiro-Costa F, Cerqueira R, Enguita FJ, Gomes RN, Dias-Ferreira J, Pereira C, Castanheira A, Pinto-do-Ó P, Leite-Moreira AF, Nascimento DS. Pericardial Fluid Accumulates microRNAs That Regulate Heart Fibrosis after Myocardial Infarction. Int J Mol Sci 2024; 25:8329. [PMID: 39125899 PMCID: PMC11313565 DOI: 10.3390/ijms25158329] [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: 06/06/2024] [Revised: 07/17/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
Pericardial fluid (PF) has been suggested as a reservoir of molecular targets that can be modulated for efficient repair after myocardial infarction (MI). Here, we set out to address the content of this biofluid after MI, namely in terms of microRNAs (miRs) that are important modulators of the cardiac pathological response. PF was collected during coronary artery bypass grafting (CABG) from two MI cohorts, patients with non-ST-segment elevation MI (NSTEMI) and patients with ST-segment elevation MI (STEMI), and a control group composed of patients with stable angina and without previous history of MI. The PF miR content was analyzed by small RNA sequencing, and its biological effect was assessed on human cardiac fibroblasts. PF accumulates fibrotic and inflammatory molecules in STEMI patients, namely causing the soluble suppression of tumorigenicity 2 (ST-2), which inversely correlates with the left ventricle ejection fraction. Although the PF of the three patient groups induce similar levels of fibroblast-to-myofibroblast activation in vitro, RNA sequencing revealed that PF from STEMI patients is particularly enriched not only in pro-fibrotic miRs but also anti-fibrotic miRs. Among those, miR-22-3p was herein found to inhibit TGF-β-induced human cardiac fibroblast activation in vitro. PF constitutes an attractive source for screening diagnostic/prognostic miRs and for unveiling novel therapeutic targets in cardiac fibrosis.
Collapse
Affiliation(s)
- Elsa D. Silva
- i3S—Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (E.D.S.); (F.R.-C.); (R.N.G.); (J.D.-F.); (C.P.); (A.C.); (P.P.-d.-Ó.)
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, University of Porto, 4050-313 Porto, Portugal
- INEB—Instituto Nacional de Engenharia Biomédica, University of Porto, 4200-135 Porto, Portugal
| | - Daniel Pereira-Sousa
- i3S—Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (E.D.S.); (F.R.-C.); (R.N.G.); (J.D.-F.); (C.P.); (A.C.); (P.P.-d.-Ó.)
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, University of Porto, 4050-313 Porto, Portugal
- Center for Translational Medicine (CTM), International Clinical Research Centre (ICRC), St. Anne’s Hospital, 60200 Brno, Czech Republic
- Department of Biomedical Sciences, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic
| | - Francisco Ribeiro-Costa
- i3S—Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (E.D.S.); (F.R.-C.); (R.N.G.); (J.D.-F.); (C.P.); (A.C.); (P.P.-d.-Ó.)
- INEB—Instituto Nacional de Engenharia Biomédica, University of Porto, 4200-135 Porto, Portugal
| | - Rui Cerqueira
- Cardiovascular R&D Center, Faculty of Medicine, University of Porto, 4150-180 Porto, Portugal; (R.C.)
| | - Francisco J. Enguita
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal;
| | - Rita N. Gomes
- i3S—Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (E.D.S.); (F.R.-C.); (R.N.G.); (J.D.-F.); (C.P.); (A.C.); (P.P.-d.-Ó.)
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, University of Porto, 4050-313 Porto, Portugal
- INEB—Instituto Nacional de Engenharia Biomédica, University of Porto, 4200-135 Porto, Portugal
| | - João Dias-Ferreira
- i3S—Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (E.D.S.); (F.R.-C.); (R.N.G.); (J.D.-F.); (C.P.); (A.C.); (P.P.-d.-Ó.)
- INEB—Instituto Nacional de Engenharia Biomédica, University of Porto, 4200-135 Porto, Portugal
| | - Cassilda Pereira
- i3S—Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (E.D.S.); (F.R.-C.); (R.N.G.); (J.D.-F.); (C.P.); (A.C.); (P.P.-d.-Ó.)
- INEB—Instituto Nacional de Engenharia Biomédica, University of Porto, 4200-135 Porto, Portugal
- Center for Translational Health and Medical Biotechnology Research (TBIO)/Health Research Network (RISE-Health), ESS, Polytechnic of Porto, 4200-072 Porto, Portugal
- Chemical and Biomolecular Sciences, School of Health (ESS), Polytechnic of Porto, 4200-465 Porto, Portugal
| | - Ana Castanheira
- i3S—Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (E.D.S.); (F.R.-C.); (R.N.G.); (J.D.-F.); (C.P.); (A.C.); (P.P.-d.-Ó.)
- INEB—Instituto Nacional de Engenharia Biomédica, University of Porto, 4200-135 Porto, Portugal
- INL—International Iberian Nanotechnology Laboratory, 4715-330 Braga, Portugal
| | - Perpétua Pinto-do-Ó
- i3S—Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (E.D.S.); (F.R.-C.); (R.N.G.); (J.D.-F.); (C.P.); (A.C.); (P.P.-d.-Ó.)
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, University of Porto, 4050-313 Porto, Portugal
- INEB—Instituto Nacional de Engenharia Biomédica, University of Porto, 4200-135 Porto, Portugal
| | - Adelino F. Leite-Moreira
- Cardiovascular R&D Center, Faculty of Medicine, University of Porto, 4150-180 Porto, Portugal; (R.C.)
| | - Diana S. Nascimento
- i3S—Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (E.D.S.); (F.R.-C.); (R.N.G.); (J.D.-F.); (C.P.); (A.C.); (P.P.-d.-Ó.)
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, University of Porto, 4050-313 Porto, Portugal
- INEB—Instituto Nacional de Engenharia Biomédica, University of Porto, 4200-135 Porto, Portugal
| |
Collapse
|
4
|
Canale P, Borghini A. Mitochondrial microRNAs: New Emerging Players in Vascular Senescence and Atherosclerotic Cardiovascular Disease. Int J Mol Sci 2024; 25:6620. [PMID: 38928325 PMCID: PMC11204228 DOI: 10.3390/ijms25126620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/04/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs that play an important role by controlling gene expression in the cytoplasm in almost all biological pathways. Recently, scientists discovered that miRNAs are also found within mitochondria, the energy-producing organelles of cells. These mitochondrial miRNAs, known as mitomiRs, can originate from the nuclear or mitochondrial genome, and they are pivotal in controlling mitochondrial function and metabolism. New insights indicate that mitomiRs may influence key aspects of the onset and progression of cardiovascular disease, especially concerning mitochondrial function and metabolic regulation. While the importance of mitochondria in cardiovascular health and disease is well-established, our understanding of mitomiRs' specific functions in crucial biological pathways, including energy metabolism, oxidative stress, inflammation, and cell death, is still in its early stages. Through this review, we aimed to delve into the mechanisms of mitomiR generation and their impacts on mitochondrial metabolic pathways within the context of vascular cell aging and atherosclerotic cardiovascular disease. The relatively unexplored field of mitomiR biology holds promise for future research investigations, with the potential to yield novel diagnostic tools and therapeutic interventions.
Collapse
Affiliation(s)
- Paola Canale
- Health Science Interdisciplinary Center, Sant’Anna School of Advanced Studies, 56124 Pisa, Italy;
- CNR Institute of Clinical Physiology, 56124 Pisa, Italy
| | | |
Collapse
|
5
|
Ruan Y, Meng S, Jia R, Cao X, Jin Z. MicroRNA-322-5p protects against myocardial infarction through targeting BTG2. Am J Med Sci 2024; 367:397-405. [PMID: 38437946 DOI: 10.1016/j.amjms.2024.02.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 01/21/2024] [Accepted: 02/29/2024] [Indexed: 03/06/2024]
Abstract
BACKGROUND Numerous studies have explored the therapeutic potential of microRNA (miR) in myocardial infarction (MI) treatment. This study focuses on the role of miR-322-5p in MI, particularly in its regulatory interaction with B-cell translocation gene 2 (BTG2). MATERIALS AND METHODS Expression levels of miR-322-5p and BTG2 were assessed in a rat MI model. Adenovirus altering miR-322-5p or BTG2 expression were administered to MI rats. Evaluation included cardiac function, inflammation, myocardial injury, pathological changes, apoptosis, and NF-κB pathway-related genes in MI rats post-targeted treatment. The miR-322-5p and BTG2 targeting relationship was investigated. RESULTS MI rats exhibited low miR-322-5p and high BTG2 expression in the myocardial tissues. Restoration of miR-322-5p enhanced cardiac function, alleviated inflammation and myocardial injury, mitigated pathological changes and apoptosis, and deactivated the NF-κB pathway in MI rats. BTG2 expression was negatively-regulated by miR-322-5p. Overexpressed BTG2 counteracted miR-322-5p-induced cardioprotection on MI rats. CONCLUSION This study provides evidence that miR-322-5p protects against MI by suppressing BTG2 expression.
Collapse
Affiliation(s)
- Yang Ruan
- Department of Cardiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Shuai Meng
- Department of Cardiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Ruofei Jia
- Department of Cardiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xiaojing Cao
- Department of Cardiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zening Jin
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China.
| |
Collapse
|
6
|
Marketou M, Kontaraki J, Zacharis E, Maragkoudakis S, Fragkiadakis K, Kampanieris E, Plevritaki A, Savva E, Malikides O, Chlouverakis G, Kochiadakis G. Peripheral Blood MicroRNA-21 as a Predictive Biomarker for Heart Failure With Preserved Ejection Fraction in Old Hypertensives. Am J Hypertens 2024; 37:298-305. [PMID: 37976292 DOI: 10.1093/ajh/hpad109] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/22/2023] [Accepted: 11/14/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND Heart failure with preserved ejection fraction (HFpEF) is a major health issue with high morbidity and mortality. The epidemiology and the factors that cause HFpEF have not been fully clarified, while accurate predictive biomarkers are lacking. Our aim was to determine whether levels of microRNA-21 (miR-21) in peripheral blood monocytes, which play a critical role in many pathophysiological pathways of hypertensive heart disease, can predict the occurrence of HFpEF in older hypertensives, as well as the associated mortality and morbidity. METHODS We enrolled 151 elderly patients >60 years old with essential hypertension but without HF at baseline. miRs expression levels in peripheral blood mononuclear cells had been quantified by real-time reverse transcription polymerase chain reaction. RESULTS During a median follow-up of 8.2 years, 56 patients (37%) had an event. Levels of miR-21 in peripheral mononuclear blood cells proved to be significantly associated with the occurrence of HFpEF. More specifically, the median HFpEF-free period was 110 months for those with miR-21 >2.1 and 114 months for those with miR-21 <2.1. In addition, multivariate analysis showed that miR-21 (hazard ratio 11.14), followed by hemoglobin (Hg) (hazard ratio 0.56 for Hg >13.6 g/dl, a 45% risk reduction), were independent and the most significant predictors of HFpEF events. CONCLUSIONS miR-21 levels in peripheral blood monocytes are associated with the development of future HFpEF. Our findings may alter the risk models of HFpEF and support the rationale for further research into the modulation of miRs as biomarkers and treatment targets for HFpEF.
Collapse
Affiliation(s)
- Maria Marketou
- Cardiology Department, Heraklion University General Hospital, Crete, Greece
- Cardiology Department, School of Medicine, University of Crete, Crete, Greece
| | - Joanna Kontaraki
- Cardiology Department, School of Medicine, University of Crete, Crete, Greece
| | - Evangelos Zacharis
- Cardiology Department, Heraklion University General Hospital, Crete, Greece
| | | | | | | | | | - Eirini Savva
- Cardiology Department, Heraklion University General Hospital, Crete, Greece
| | | | - Gregory Chlouverakis
- Division of Biostatistics, School of Medicine, University of Crete, Crete, Greece
| | - George Kochiadakis
- Cardiology Department, Heraklion University General Hospital, Crete, Greece
- Cardiology Department, School of Medicine, University of Crete, Crete, Greece
| |
Collapse
|
7
|
Yang Y, Liang F, Gao J, Li J, Jiang C, Xie W, Wu S, Wang Y, Yi J. Salidroside Ameliorates Ischemia/Reperfusion-Induced Human Cardiomyocyte Injury by Inhibiting the Circ_0097682/miR-671-5p/USP46 Pathway. Cardiovasc Toxicol 2023; 23:406-418. [PMID: 37740139 DOI: 10.1007/s12012-023-09808-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/06/2023] [Indexed: 09/24/2023]
Abstract
Salidroside shows an inhibitory effect on myocardial ischemia/reperfusion (I/R) injury; however, the underlying mechanism remains to be explored. The present work analyzes the mechanism that drives salidroside to ameliorate I/R-induced human cardiomyocyte injury. Human cardiomyocytes were subjected to I/R treatment to simulate a myocardial infarction cell model. Cell viability, cell proliferation, and cell apoptosis were analyzed by CCK-8 assay, EdU assay, and flow cytometry analysis, respectively. RNA expression levels of circ_0097682, miR-671-5p, and F-box and ubiquitin-specific peptidase 46 (USP46) were detected by qRT-PCR. Protein expression was measured by Western blotting assay. The levels of IL-6, IL-1β, and TNF-α in cell supernatant were detected by enzyme-linked immunosorbent assays. Salidroside treatment relieved I/R-induced inhibitory effect on AC16 cell proliferation and promoting effects on cell apoptosis, inflammation, and oxidative stress. Salidroside inhibited circ_0097682 expression in I/R-treated AC16 cells. Salidroside-mediated inhibition of I/R-induced cell injury involved the downregulation of circ_0097682 expression. In addition, circ_0097682 bound to miR-671-5p in AC16 cells, and miR-671-5p inhibitors rescued salidroside pretreatment-mediated effects in I/R-treated AC16 cells. Moreover, miR-671-5p targeted USP46 in AC16 cells, and USP46 introduction partially relieved circ_0097682 depletion or salidroside pretreatment-induced effects in I/R-treated AC16 cells. Salidroside ameliorated I/R-induced AC16 cell injury by inhibiting the circ_0097682/miR-671-5p/USP46 pathway.
Collapse
Affiliation(s)
- Yuyang Yang
- College of Traditional Chinese Medicine, North China University of Science Technology, Qinhuangdao, China
| | - Fangqian Liang
- Department of General Practice, North China University of Science and Technology Affiliated Hospital, No. 73, Jianshe South Road, Lubei District, Tangshan, 063000, Hebei, China
| | - Jingyuan Gao
- Department of General Practice, North China University of Science and Technology Affiliated Hospital, No. 73, Jianshe South Road, Lubei District, Tangshan, 063000, Hebei, China.
| | - Jian Li
- College of Traditional Chinese Medicine, North China University of Science Technology, Qinhuangdao, China
| | - Chunhua Jiang
- College of Traditional Chinese Medicine, North China University of Science Technology, Qinhuangdao, China
| | - Wei Xie
- College of Traditional Chinese Medicine, North China University of Science Technology, Qinhuangdao, China
| | - Shujuan Wu
- College of Traditional Chinese Medicine, North China University of Science Technology, Qinhuangdao, China
| | - Ya Wang
- College of Traditional Chinese Medicine, North China University of Science Technology, Qinhuangdao, China
| | - Jing Yi
- College of Traditional Chinese Medicine, North China University of Science Technology, Qinhuangdao, China
| |
Collapse
|
8
|
Li H, Wang Q, Shi L, Li T. Sensitively detecting endogenous homocysteine in human serum and cardiomyocytes with a specific fluorescent probe. Analyst 2023; 148:5935-5941. [PMID: 37850493 DOI: 10.1039/d3an01430d] [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: 10/19/2023]
Abstract
The elevated level of homocysteine (Hcy) in circulating blood is generally regarded as a risk factor for a variety of diseases including acute myocardial infarction (AMI), but there is no clear answer to whether circulating Hcy can be used for AMI diagnosis. To address it, here we have designed a tetraazacycle-based fluorescent probe for sensitive detection of endogenous Hcy in AMI patients' serum and cardiomyocytes, showing a perfect selectivity over other biothiols (e.g. Cys and GSH). It mainly relies on the formation of a stable six-membered ring structure when this probe responds to Hcy, which is accompanied by a weakening of photoinduced electron transfer (PET) that induces a sharp increase in the fluorescence emission. In this way, Hcy can be probed in biofluids with high sensitivity. We then employed this fluorescent sensor to statistically analyze the levels of Hcy in human circulating blood, indicating a big difference between AMI patients and the healthy participants. To tell whether such a difference is applicable to AMI diagnosis, we further compare the expression levels of Hcy in cardiomyocytes and other tissue cells. It reveals a lower level of endogenous Hcy in cardiomyocytes, implying no direct relationship between the elevated Hcy and cardiomyocyte damage. This observation suggests that Hcy in circulating blood cannot be utilized as a potential biomarker for AMI diagnosis, although it is proven as a risk factor for this disease.
Collapse
Affiliation(s)
- Huan Li
- Department of Chemistry, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui 230026, China.
| | - Qiwei Wang
- Department of Chemistry, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui 230026, China.
| | - Lili Shi
- Department of Chemistry, Anhui University, 111 Jiulong Road, Hefei, Anhui 230601, China.
| | - Tao Li
- Department of Chemistry, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui 230026, China.
| |
Collapse
|
9
|
Asjad E, Dobrzynski H. MicroRNAs: Midfielders of Cardiac Health, Disease and Treatment. Int J Mol Sci 2023; 24:16207. [PMID: 38003397 PMCID: PMC10671258 DOI: 10.3390/ijms242216207] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/05/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
MicroRNAs (miRNAs) are a class of small non-coding RNA molecules that play a role in post-transcriptional gene regulation. It is generally accepted that their main mechanism of action is the negative regulation of gene expression, through binding to specific regions in messenger RNA (mRNA) and repressing protein translation. By interrupting protein synthesis, miRNAs can effectively turn genes off and influence many basic processes in the body, such as developmental and apoptotic behaviours of cells and cardiac organogenesis. Their importance is highlighted by inhibiting or overexpressing certain miRNAs, which will be discussed in the context of coronary artery disease, atrial fibrillation, bradycardia, and heart failure. Dysregulated levels of miRNAs in the body can exacerbate or alleviate existing disease, and their omnipresence in the body makes them reliable as quantifiable markers of disease. This review aims to provide a summary of miRNAs as biomarkers and their interactions with targets that affect cardiac health, and intersperse it with current therapeutic knowledge. It intends to succinctly inform on these topics and guide readers toward more comprehensive works if they wish to explore further through a wide-ranging citation list.
Collapse
Affiliation(s)
- Emman Asjad
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK;
| | - Halina Dobrzynski
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK;
- Department of Anatomy, Jagiellonian University Medical College, 31-034 Krakow, Poland
| |
Collapse
|
10
|
The Role of ncRNAs in Cardiac Infarction and Regeneration. J Cardiovasc Dev Dis 2023; 10:jcdd10030123. [PMID: 36975887 PMCID: PMC10052289 DOI: 10.3390/jcdd10030123] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/06/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023] Open
Abstract
Myocardial infarction is the most prevalent cardiovascular disease worldwide, and it is defined as cardiomyocyte cell death due to a lack of oxygen supply. Such a temporary absence of oxygen supply, or ischemia, leads to extensive cardiomyocyte cell death in the affected myocardium. Notably, reactive oxygen species are generated during the reperfusion process, driving a novel wave of cell death. Consequently, the inflammatory process starts, followed by fibrotic scar formation. Limiting inflammation and resolving the fibrotic scar are essential biological processes with respect to providing a favorable environment for cardiac regeneration that is only achieved in a limited number of species. Distinct inductive signals and transcriptional regulatory factors are key components that modulate cardiac injury and regeneration. Over the last decade, the impact of non-coding RNAs has begun to be addressed in many cellular and pathological processes including myocardial infarction and regeneration. Herein, we provide a state-of-the-art review of the current functional role of diverse non-coding RNAs, particularly microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), in different biological processes involved in cardiac injury as well as in distinct experimental models of cardiac regeneration.
Collapse
|
11
|
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
|
12
|
MicroRNA Let-7a, -7e and -133a Attenuate Hypoxia-Induced Atrial Fibrosis via Targeting Collagen Expression and the JNK Pathway in HL1 Cardiomyocytes. Int J Mol Sci 2022; 23:ijms23179636. [PMID: 36077031 PMCID: PMC9455749 DOI: 10.3390/ijms23179636] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 08/16/2022] [Accepted: 08/18/2022] [Indexed: 11/29/2022] Open
Abstract
Fibrosis is a hallmark of atrial structural remodeling. The main aim of this study was to investigate the role of micro-ribonucleic acids (miRNAs) in the modulation of fibrotic molecular mechanisms in response to hypoxic conditions, which may mediate atrial fibrosis. Under a condition of hypoxia induced by a hypoxia chamber, miRNA arrays were used to identify the specific miRNAs associated with the modulation of fibrotic genes. Luciferase assay, real-time polymerase chain reaction, immunofluorescence and Western blotting were used to investigate the effects of miRNAs on the expressions of the fibrotic markers collagen I and III (COL1A, COL3A) and phosphorylation levels of the stress kinase c-Jun N-terminal kinase (JNK) pathway in a cultured HL-1 atrial cardiomyocytes cell line. COL1A and COL3A were found to be the direct regulatory targets of miR-let-7a, miR-let-7e and miR-133a in hypoxic atrial cardiac cells in vitro. The expressions of COL1A and COL3A were influenced by treatment with miRNA mimic and antagomir while hypoxia-induced collagen expression was inhibited by the delivery of miR-133a, miR-let-7a or miR-let-7e. The JNK pathway was critical in the pathogenesis of atrial fibrosis. The JNK inhibitor SP600125 increased miRNA expressions and repressed the fibrotic markers COL1A and COL3A. In conclusion, MiRNA let-7a, miR-let-7e and miR-133a play important roles in hypoxia-related atrial fibrosis by inhibiting collagen expression and post-transcriptional repression by the JNK pathway. These novel findings may lead to the development of new therapeutic strategies.
Collapse
|
13
|
Marwarha G, Røsand Ø, Slagsvold KH, Høydal MA. GSK3β Inhibition Is the Molecular Pivot That Underlies the Mir-210-Induced Attenuation of Intrinsic Apoptosis Cascade during Hypoxia. Int J Mol Sci 2022; 23:ijms23169375. [PMID: 36012628 PMCID: PMC9409400 DOI: 10.3390/ijms23169375] [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: 06/30/2022] [Revised: 08/09/2022] [Accepted: 08/17/2022] [Indexed: 11/16/2022] Open
Abstract
Apoptotic cell death is a deleterious consequence of hypoxia-induced cellular stress. The master hypoxamiR, microRNA-210 (miR-210), is considered the primary driver of the cellular response to hypoxia stress. We have recently demonstrated that miR-210 attenuates hypoxia-induced apoptotic cell death. In this paper, we unveil that the miR-210-induced inhibition of the serine/threonine kinase Glycogen Synthase Kinase 3 beta (GSK3β) in AC-16 cardiomyocytes subjected to hypoxia stress underlies the salutary protective response of miR-210 in mitigating the hypoxia-induced apoptotic cell death. Using transient overexpression vectors to augment miR-210 expression concomitant with the ectopic expression of the constitutive active GSK3β S9A mutant (ca-GSK3β S9A), we exhaustively performed biochemical and molecular assays to determine the status of the hypoxia-induced intrinsic apoptosis cascade. Caspase-3 activity analysis coupled with DNA fragmentation assays cogently demonstrate that the inhibition of GSK3β kinase activity underlies the miR-210-induced attenuation in the hypoxia-driven apoptotic cell death. Further elucidation and delineation of the upstream cellular events unveiled an indispensable role of the inhibition of GSK3β kinase activity in mediating the miR-210-induced mitigation of the hypoxia-driven BAX and BAK insertion into the outer mitochondria membrane (OMM) and the ensuing Cytochrome C release into the cytosol. Our study is the first to unveil that the inhibition of GSK3β kinase activity is indispensable in mediating the miR-210-orchestrated protective cellular response to hypoxia-induced apoptotic cell death.
Collapse
Affiliation(s)
- Gurdeep Marwarha
- Group of Molecular and Cellular Cardiology, Department of Circulation and Medical Imaging, Faculty of Medicine and Health, Norwegian University of Science and Technology (NTNU), 7034 Trondheim, Norway
| | - Øystein Røsand
- Group of Molecular and Cellular Cardiology, Department of Circulation and Medical Imaging, Faculty of Medicine and Health, Norwegian University of Science and Technology (NTNU), 7034 Trondheim, Norway
| | - Katrine Hordnes Slagsvold
- Group of Molecular and Cellular Cardiology, Department of Circulation and Medical Imaging, Faculty of Medicine and Health, Norwegian University of Science and Technology (NTNU), 7034 Trondheim, Norway
- Department of Cardiothoracic Surgery, St. Olavs University Hospital, 7030 Trondheim, Norway
| | - Morten Andre Høydal
- Group of Molecular and Cellular Cardiology, Department of Circulation and Medical Imaging, Faculty of Medicine and Health, Norwegian University of Science and Technology (NTNU), 7034 Trondheim, Norway
- Correspondence: ; Tel.: +47-48134843
| |
Collapse
|
14
|
Xu C, Jia Z, Cao X, Wang S, Wang J, an L. Hsa_circ_0007059 promotes apoptosis and inflammation in cardiomyocytes during ischemia by targeting microRNA-378 and microRNA-383. Cell Cycle 2022; 21:1003-1019. [PMID: 35192424 PMCID: PMC9037457 DOI: 10.1080/15384101.2022.2040122] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Circular RNAs (circRNAs) are a class of non-coding RNA molecules that are associated with not only normal physiological functions but also various diseases, including cardiac diseases such as myocardial infarction (MI). The present study explored the potential role of circRNA_0007059 (circ_0007059) during MI pathogenesis using in vitro studies. Microarray and quantitative PCR analyses demonstrated elevated circ_0007059 expression and downregulated miR-378 and miR-383 expression in H2O2-treated mice cardiomyocytes and infarcted hearts of MI mouse model as compared those in relevant controls. Moreover, circ_0007059 knockdown improved cardiomyocyte viability after H2O2 treatment as revealed by the CCK-8 and colony formation assays. Flow cytometry and caspase activity assays demonstrated that circ_0007059 suppressed H2O2-induced cardiomyocyte apoptosis. Enzyme-linked immunosorbent assays and Western blotting revealed that inflammatory cytokine (interleukin-1β, interleukin-18 and C-C motif chemokine ligand 5) expression was induced by H2O2 treatment and that circ_0007059 repressed H2O2-induced inflammation. Bioinformatics analyses and dual-luciferase reporter assays showed that circ_0000759 acts as a miR-378 and miR-383 sponge. Furthermore, the upregulation or suppression of miR-378 and miR-383 expression in H2O2-treated cardiomyocytes had similar effects on the apoptosis and inflammation of cardiomyocytes as that of circ_0007059 knockdown or overexpression, respectively. Additionally, lentiviral shRNA-circ_0007059 administration to mice with MI considerably reduced the size of infarcted regions and promoted cardiac activity. Collectively, our findings suggest that circ_0007059 expression is upregulated in mice cardiomyocytes in response to oxidative stress and cardiac tissues of MI mouse model, suggesting its involvement in the pathogenesis of MI by targeting miR-378 and miR-383.
Collapse
Affiliation(s)
- Chaorui Xu
- Department of Geriatric Cardiology, Heilongjiang Provincial Hospital, Harbin, Heilongjiang, China
| | - Zhuowen Jia
- Department of Geriatric Cardiology, Heilongjiang Provincial Hospital, Harbin, Heilongjiang, China
| | - Xuefei Cao
- Department of Geriatric Cardiology, Heilongjiang Provincial Hospital, Harbin, Heilongjiang, China
| | - Sha Wang
- Department of Geriatric Cardiology, Heilongjiang Provincial Hospital, Harbin, Heilongjiang, China
| | - Jipeng Wang
- Department of Geriatric Cardiology, Heilongjiang Provincial Hospital, Harbin, Heilongjiang, China
| | - Liping an
- Department of Geriatric Cardiology, Heilongjiang Provincial Hospital, Harbin, Heilongjiang, China,CONTACT Liping An Department of Geriatric Cardiology, Heilongjiang Provincial Hospital, No. 82 Zhongshan Road, Xiangfang District, Harbin, Heilongjiang150036, China
| |
Collapse
|
15
|
Sun Y, Fang L, Han Y, Feng A, Liu S, Zhang K, Xu JJ. Reversible Ratiometric Electrochemiluminescence Biosensor Based on DNAzyme Regulated Resonance Energy Transfer for Myocardial miRNA Detection. Anal Chem 2022; 94:7035-7040. [PMID: 35467832 DOI: 10.1021/acs.analchem.2c00195] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Myocardial miRNAs in peripheral blood are closely related to the pathogenic process of myocardial infarction. Rapid identification and accurate quantification of myocardial miRNAs are of great significance to clinical interventions for treating cardiovascular lesions. Therefore, a ratiometric electrochemiluminescence (ECL) biosensor integrating DNAzyme with a resonance energy transfer (RET) system was designed to detect myocardial miRNA. The dual-signal system was composed of rA marked substrate strand functionalized CdTe quantum dots (QDs) as reductive-oxidative (R-O) emitters and Cy5-labeled strand-functionalized Ru(bpy)32+-filled silica nanoparticles (RuSi NPs) as oxidative-reductive (O-R) emitters. In the presence of target miRNA, DNAzyme was activated to cut substrate strands on the CdTe QDs and release triggers for opening hairpin probes. Then, the Cy5 molecule-labeled hairpin DNA on the RuSi NPs was opened to introduce Cy5 molecules and RuSi NPs into the system. The R-O ECL was quenched by ECL-RET between CdTe QDs and Cy5 molecules and the O-R ECL was introduced by the RuSi NPs. In this way, based on the simultaneous changing of the ECL signal, the dual-potential dynamic signal ratiometric ECL sensing platform was developed. By measuring the ratio of O-R ECL signal to R-O ECL signal, the concentration of miRNA-499 was accurately quantified in the range of 10 fM to 10 nM, and the detection limit was as low as 2.44 fM (S/N = 3). This DNAzyme guided dual-potential ratiometric ECL method provides a sensitive and reliable method for myocardial miRNA detection, and it has great potential in clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Yudie Sun
- School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma Xiang Road, Ma 'anshan, Anhui 243032, People's Republic of China
| | - La Fang
- School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma Xiang Road, Ma 'anshan, Anhui 243032, People's Republic of China
| | - Yunxiang Han
- School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma Xiang Road, Ma 'anshan, Anhui 243032, People's Republic of China
| | - Aobo Feng
- School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma Xiang Road, Ma 'anshan, Anhui 243032, People's Republic of China
| | - Shengjun Liu
- School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma Xiang Road, Ma 'anshan, Anhui 243032, People's Republic of China
| | - Kui Zhang
- School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma Xiang Road, Ma 'anshan, Anhui 243032, People's Republic of China
| | - Jing-Juan Xu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, People's Republic of China
| |
Collapse
|
16
|
Marinescu MC, Lazar AL, Marta MM, Cozma A, Catana CS. Non-Coding RNAs: Prevention, Diagnosis, and Treatment in Myocardial Ischemia-Reperfusion Injury. Int J Mol Sci 2022; 23:ijms23052728. [PMID: 35269870 PMCID: PMC8911068 DOI: 10.3390/ijms23052728] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 02/24/2022] [Accepted: 02/26/2022] [Indexed: 12/17/2022] Open
Abstract
Recent knowledge concerning the role of non-coding RNAs (ncRNAs) in myocardial ischemia/reperfusion (I/R) injury provides new insight into their possible roles as specific biomarkers for early diagnosis, prognosis, and treatment. MicroRNAs (miRNAs) have fewer than 200 nucleotides, while long ncRNAs (lncRNAs) have more than 200 nucleotides. The three types of ncRNAs (miRNAs, lncRNAs, and circRNAs) act as signaling molecules strongly involved in cardiovascular disorders (CVD). I/R injury of the heart is the main CVD correlated with acute myocardial infarction (AMI), cardiac surgery, and transplantation. The expression levels of many ncRNAs and miRNAs are highly modified in the plasma of MI patients, and thus they have the potential to diagnose and treat MI. Cardiomyocyte and endothelial cell death is the major trigger for myocardial ischemia–reperfusion syndrome (MIRS). The cardioprotective effect of inflammasome activation in MIRS and the therapeutics targeting the reparative response could prevent progressive post-infarction heart failure. Moreover, the pharmacological and genetic modulation of these ncRNAs has the therapeutic potential to improve clinical outcomes in AMI patients.
Collapse
Affiliation(s)
- Mihnea-Cosmin Marinescu
- County Clinical Emergency Hospital of Brasov Romania, 500326 Brașov, Romania;
- Department of Vascular Surgery, Second Surgical Clinic, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Andrada-Luciana Lazar
- Department of Dermatology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
| | - Monica Mihaela Marta
- Department of Medical Education, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
| | - Angela Cozma
- Department of Internal Medicine, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- Correspondence:
| | - Cristina-Sorina Catana
- Department of Medical Biochemistry, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
| |
Collapse
|
17
|
Han J, Zhang Z, Zhang Z, Yang S. Artemisinin relieves myocardial ischemia-reperfusion injury via modulating miR-29b-3p and hemicentin 1. Front Pharmacol 2022; 13:918966. [PMID: 36034861 PMCID: PMC9403756 DOI: 10.3389/fphar.2022.918966] [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: 04/28/2022] [Accepted: 07/15/2022] [Indexed: 11/13/2022] Open
Abstract
Objective: To explore the impact of artemisinin (ARS) on myocardial ischemia-reperfusion (I/R) injury and the underlying mechanism. Methods: Myocardial I/R rat model and cell model were used in this study. The cell viability, morphological changes, apoptosis, and oxidative stress were evaluated in cardiomyocytes H9c2 cells in vitro by using cell counting kit-8, microscope, flow cytometry, and commercial kits. High throughput sequencing is used to identify molecular targets of ARS on myocardial I/R injury, and then the gene-gene interaction network was constructed. MiR-29b-3p, hemicentin 1 (HMCN1), and apoptosis-related genes were tested by qRT-PCR and Western blotting. In the myocardial I/R rat model, echocardiography, (Triphenyl tetrazolium chloride) TTC staining, Hematoxylin-eosin (H&E) staining, Masson Trichrome staining, and TUNEL staining are applied to evaluate the protective effect of ARS on the myocardial injury. Results: In vitro, we demonstrated that ARS alleviated H2O2-induced myocardial I/R injury, manifested by increased H9c2 viability, decreased pathological changes, apoptosis, and oxidative stress biomarker ROS, LDH, and CK-MB. Then, sequencing analysis revealed that miR-29b-3p/HMCN1 was the target of ARS for myocardial I/R injury. Notably, rescue experiments indicated that ARS inhibited myocardial I/R injury through targeted regulation miR-29b-3p/HMCN1. In vivo, we confirmed that ARS reduced myocardial injury, fibrosis, and apoptosis via modulation of miR-29b-3p/HMCN1. Conclusion: This study demonstrated the functional role of the ARS/miR-29b-3p/HMCN1 axis in alleviating myocardial I/R injury, which provided a new direction for myocardial I/R injury therapy.
Collapse
Affiliation(s)
- Junyu Han
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Ziguan Zhang
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Zhonghe Zhang
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Shuyu Yang
- Xiamen Diabetes Institute, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
18
|
Liu W, Lin W, Yu L. Long non-coding RNA muscleblind like splicing regulator 1 antisense RNA 1 (LncRNA MBNL1-AS1) promotes the progression of acute myocardial infarction by regulating the microRNA-132-3p/SRY-related high-mobility-group box 4 (SOX4) axis. Bioengineered 2022; 13:1424-1435. [PMID: 34978261 PMCID: PMC8805923 DOI: 10.1080/21655979.2021.2018974] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 12/10/2021] [Indexed: 12/18/2022] Open
Abstract
Long non-coding RNA muscleblind like splicing regulator 1 antisense RNA 1 (LncRNA MBNL1-AS1) exerts vital role in various physiological processes. However, its functions in acute myocardial infarction (AMI) are not elucidated. AMI model was constructed using Wistar rats and it was found that LncRNA MBNL1-AS1 was upregulated in AMI model according to the quantitative real-time polymerase chain reaction (qRT-PCR) results. The left ventricular systolic pressure (LVSP), left ventricular end diastolic pressure (LVEDP) and maximum rate of rise/fall of left ventricle pressure (±dp/dt max) were detected through hemodynamics test, which showed that knockdown of MBNL1-AS1 improved cardiac function in AMI model. Next, the myocardial infarction area was estimated by triphenyltetrazole chloride (TTC) staining, and the levels of cardiac troponin I (cTn-I) and creatine kinase-MB (CK-MB) were detected by enzyme-linked immunosorbent assay (ELISA) kit. The results revealed that silencing MBLN1-AS1 alleviated myocardial injury in AMI model. Additionally, MBNL1-AS1 knockdown inhibited apoptosis of myocardial cells and reduced the expression of apoptotic proteins. According to DIANA database and luciferase reporter assay, miR-132-3p was the direct target of MBNL1-AS1 and was negatively regulated by MBNL1-AS1. Furthermore, Targetscan database predicted that SRY-related high-mobility-group box 4 (SOX4) was the direct target of miR-132-3p and was regulated by MBNL1-AS1 through miR-132-3p. Moreover, overexpression of SOX4 partially eliminated effects of MBNL1-AS1 on myocardial cells. In conclusion, this investigation for the first time revealed that LncRNA MBNL1-AS1 was the potential target for treating AMI and expounded the underlying mechanisms of it.
Collapse
Affiliation(s)
- Weifeng Liu
- Department of Cardiology, Yantai Yuhuangding Hospital, Qingdao Medical College, Qingdao University, Yantai, China
| | - Wenyuan Lin
- Department of Cardiology, Yantai Yuhuangding Hospital, Qingdao Medical College, Qingdao University, Yantai, China
| | - Liangliang Yu
- Department of Cardiology, Yantai Yuhuangding Hospital, Qingdao Medical College, Qingdao University, Yantai, China
| |
Collapse
|
19
|
Ji Z, Wang C, Tong Q. Role of miRNA-324-5p-Modified Adipose-Derived Stem Cells in Post-Myocardial Infarction Repair. Int J Stem Cells 2021; 14:298-309. [PMID: 34158416 PMCID: PMC8429947 DOI: 10.15283/ijsc21025] [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: 02/03/2021] [Revised: 04/26/2021] [Accepted: 04/30/2021] [Indexed: 11/09/2022] Open
Abstract
Background and Objectives To seek out the role of mircoRNA (miR)-324-5p-modified adipose-derived stem cells (ADSCs) in post-myocardial infarction (MI) myocardial repair. Methods and Results Rat ADSCs were cultivated and then identified by morphologic observation, osteogenesis and adipogenesis induction assays and flow cytometry. Afterwards, ADSCs were modified by miR-324-5p lentiviral vector, with ADSC proliferation and migration measured. Then, rat MI model was established, which was treated by ADSCs or miR-324-5p-modified ADSCs. Subsequently, the function of miR-324-5p-modified ADSCs in myocardial repair of MI rats was assessed through functional assays. Next, the binding relation of miR-324-5p and Toll-interacting protein (TOLLIP) was validated. Eventually, functional rescue assay of TOLLIP was performed to verify the role of TOLLIP in MI. First, rat ADSCs were harvested. Overexpressed miR-324-5p improved ADSC viability. ADSC transplantation moderately enhanced cardiac function of MI rats, reduced enzyme levels and decreased infarct size and apoptosis; while miR-324-5p-modified ADSCs could better promote post-MI repair. Mechanically, miR-324-5p targeted TOLLIP in myocardial tissues. Moreover, TOLLIP overexpression debilitated the promotive role of miR-324-5p-modified ADSCs in post-MI repair in rats. Conclusions miR-324-5p-modified ADSCs evidently strengthened post-MI myocardial repair by targeting TOLLIP in myocardial tissues.
Collapse
Affiliation(s)
- Zhou Ji
- Department of Cardiovascular Medicine, The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Chan Wang
- Jinzhou Hospital of Traditional Chinese Medicine, Jinzhou, China
| | - Qing Tong
- Office of Academic Research, The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
20
|
A Brief Review on the Biology and Effects of Cellular and Circulating microRNAs on Cardiac Remodeling after Infarction. Int J Mol Sci 2021; 22:ijms22094995. [PMID: 34066757 PMCID: PMC8125864 DOI: 10.3390/ijms22094995] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/03/2021] [Accepted: 05/05/2021] [Indexed: 01/21/2023] Open
Abstract
Despite advances in diagnostic, prognostic, and treatment modalities, myocardial infarction (MI) remains a leading cause of morbidity and mortality. Impaired cellular signaling after an MI causes maladaptive changes resulting in cardiac remodeling. MicroRNAs (miRNAs/miR) along with other molecular components have been investigated for their involvement in cellular signaling in the pathogenesis of various cardiac conditions like MI. miRNAs are small non-coding RNAs that negatively regulate gene expression. They bind to complementary mRNAs and regulate the rate of protein synthesis by altering the stability of their targeted mRNAs. A single miRNA can modulate several cellular signaling pathways by targeting hundreds of mRNAs. This review focuses on the biogenesis and beneficial effects of cellular and circulating (exosomal) miRNAs on cardiac remodeling after an MI. Particularly, miR-1, -133, 135, and -29 that play an essential role in cardiac remodeling after an MI are described in detail. The limitations that will need to be addressed in the future for the further development of miRNA-based therapeutics for cardiovascular conditions will also be discussed.
Collapse
|
21
|
Tual-Chalot S, Stellos K. MicroRNA-based therapy of postmyocardial infarction heart failure. Hellenic J Cardiol 2021; 62:149-151. [PMID: 33852921 DOI: 10.1016/j.hjc.2021.03.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 03/23/2021] [Indexed: 11/28/2022] Open
Affiliation(s)
- Simon Tual-Chalot
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Konstantinos Stellos
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK; Department of Cardiology, Freeman Hospital, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK.
| |
Collapse
|
22
|
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: 17] [Impact Index Per Article: 4.3] [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
|
23
|
Dai B, Wang F, Nie X, Du H, Zhao Y, Yin Z, Li H, Fan J, Wen Z, Wang DW, Chen C. The Cell Type-Specific Functions of miR-21 in Cardiovascular Diseases. Front Genet 2020; 11:563166. [PMID: 33329700 PMCID: PMC7714932 DOI: 10.3389/fgene.2020.563166] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 09/18/2020] [Indexed: 12/16/2022] Open
Abstract
Cardiovascular diseases are one of the prime reasons for disability and death worldwide. Diseases and conditions, such as hypoxia, pressure overload, infection, and hyperglycemia, might initiate cardiac remodeling and dysfunction by inducing hypertrophy or apoptosis in cardiomyocytes and by promoting proliferation in cardiac fibroblasts. In the vascular system, injuries decrease the endothelial nitric oxide levels and affect the phenotype of vascular smooth muscle cells. Understanding the underlying mechanisms will be helpful for the development of a precise therapeutic approach. Various microRNAs are involved in mediating multiple pathological and physiological processes in the heart. A cardiac enriched microRNA, miR-21, which is essential for cardiac homeostasis, has been demonstrated to act as a cell–cell messenger with diverse functions. This review describes the cell type–specific functions of miR-21 in different cardiovascular diseases and its prospects in clinical therapy.
Collapse
Affiliation(s)
- Beibei Dai
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Feng Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Xiang Nie
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Hengzhi Du
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Yanru Zhao
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Zhongwei Yin
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Huaping Li
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Jiahui Fan
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Zheng Wen
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Chen Chen
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| |
Collapse
|
24
|
Cardioprotective Effects of Dietary Flaxseed Post-Infarction Are Associated with Changes in MicroRNA Expression. Biomolecules 2020; 10:biom10091297. [PMID: 32911872 PMCID: PMC7564197 DOI: 10.3390/biom10091297] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/01/2020] [Accepted: 09/05/2020] [Indexed: 12/13/2022] Open
Abstract
MicroRNAs (miRNAs/miRs) such as miR-1, miR-133a, miR-133b, miR-135a, and miR-29b play a key role in many cardiac pathological remodeling processes, including apoptosis, fibrosis, and arrhythmias, after a myocardial infarction (MI). Dietary flaxseed has demonstrated a protective effect against an MI. The present study was carried out to test the hypothesis that dietary flaxseed supplementation before and after an MI regulates the expression of above-mentioned miRNAs to produce its cardioprotective effect. Animals were randomized after inducing MI by coronary artery ligation into: (a) sham MI with normal chow, (b) MI with normal chow, and (c–e) MI supplemented with either 10% milled flaxseed, or 4.4% flax oil enriched in alpha-linolenic acid (ALA), or 0.44% flax lignan secoisolariciresinol diglucoside. The feeding protocol consisted of 2 weeks before and 8 weeks after the surgery. Dietary flax oil supplementation selectively upregulated the cardiac expression of miR-133a, miR-135a, and miR-29b. The levels of collagen I expression were reduced in the flax oil group. We conclude that miR-133a, miR-135a, and miR-29b are sensitive to dietary flax oil, likely due to its rich ALA content. The cardioprotective effect of flaxseed in an MI could be due to modulation of these miRNAs.
Collapse
|
25
|
Williams AL, Khadka VS, Anagaran MCT, Lee K, Avelar A, Deng Y, Shohet RV. miR-125 family regulates XIRP1 and FIH in response to myocardial infarction. Physiol Genomics 2020; 52:358-368. [PMID: 32716698 DOI: 10.1152/physiolgenomics.00041.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
MicroRNAs (miRNAs) are powerful regulators of protein expression. Many play important roles in cardiac development and disease. While several miRNAs and targets have been well characterized, the abundance of miRNAs and the numerous potential targets for each suggest that the vast majority of these interactions have yet to be described. The goal of this study was to characterize miRNA expression in the mouse heart after coronary artery ligation (LIG) and identify novel mRNA targets altered during the initial response to ischemic stress. We performed small RNA sequencing (RNA-Seq) of ischemic heart tissue 1 day and 3 days after ligation and identified 182 differentially expressed miRNAs. We then selected relevant mRNA targets from all potential targets by correlating miRNA and mRNA expression from a corresponding RNA-Seq data set. From this analysis we chose to focus, as proof of principle, on two miRNAs from the miR-125 family, miR-125a and miR-351, and two of their potential mRNA targets, Xin actin-binding repeat-containing protein 1 (XIRP1) and factor inhibiting hypoxia-inducible factor (FIH). We found miR-125a to be less abundant and XIRP1 more abundant after ligation. In contrast, the related murine miRNA miR-351 was substantially upregulated in response to ischemic injury, and FIH expression correspondingly decreased. Luciferase reporter assays confirmed direct interactions between these miRNAs and targets. In summary, we utilized a correlative analysis strategy combining miRNA and mRNA expression data to identify functional miRNA-mRNA relationships in the heart after ligation. These findings provide insight into the response to ischemic injury and suggest future therapeutic targets.
Collapse
Affiliation(s)
- Allison Lesher Williams
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| | - Vedbar S Khadka
- Bioinformatics Core, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| | - Ma C T Anagaran
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| | - Katie Lee
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| | - Abigail Avelar
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| | - Youping Deng
- Bioinformatics Core, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| | - Ralph V Shohet
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| |
Collapse
|
26
|
Castellan RF, Vitiello M, Vidmar M, Johnstone S, Iacobazzi D, Mellis D, Cathcart B, Thomson A, Ruhrberg C, Caputo M, Newby DE, Gray GA, Baker AH, Caporali A, Meloni M. miR-96 and miR-183 differentially regulate neonatal and adult postinfarct neovascularization. JCI Insight 2020; 5:134888. [PMID: 32544097 PMCID: PMC7453899 DOI: 10.1172/jci.insight.134888] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 06/10/2020] [Indexed: 12/18/2022] Open
Abstract
Following myocardial infarction (MI), the adult heart has minimal regenerative potential. Conversely, the neonatal heart can undergo extensive regeneration, and neovascularization capacity was hypothesized to contribute to this difference. Here, we demonstrate the higher angiogenic potential of neonatal compared with adult mouse cardiac endothelial cells (MCECs) in vitro and use this difference to identify candidate microRNAs (miRs) regulating cardiac angiogenesis after MI. miR expression profiling revealed miR-96 and miR-183 upregulation in adult compared with neonatal MCECs. Their overexpression decreased the angiogenic potential of neonatal MCECs in vitro and prevented scar resolution and neovascularization in neonatal mice after MI. Inversely, their inhibition improved the angiogenic potential of adult MCECs, and miR-96/miR-183–KO mice had increased peri-infarct neovascularization. In silico analyses identified anillin (ANLN) as a direct target of miR-96 and miR-183. In agreement, Anln expression declined following their overexpression and increased after their inhibition in vitro. Moreover, ANLN expression inversely correlated with miR-96 expression and age in cardiac ECs of cardiovascular patients. In vivo, ANLN+ vessels were enriched in the peri-infarct area of miR-96/miR-183–KO mice. These findings identify miR-96 and miR-183 as regulators of neovascularization following MI and miR-regulated genes, such as anillin, as potential therapeutic targets for cardiovascular disease. MiR-96 and miR-183 act as molecular switches to regulate endothelial cells angiogenic potential and differentially regulate neovascularization following myocardial infarction in neonatal and adult mice.
Collapse
Affiliation(s)
- Raphael Fp Castellan
- British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom.,UCL Institute of Ophthalmology, London, United Kingdom
| | - Milena Vitiello
- British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Martina Vidmar
- British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Steven Johnstone
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Dominga Iacobazzi
- Bristol Heart Institute, School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| | - David Mellis
- British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Benjamin Cathcart
- British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Adrian Thomson
- British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | | | - Massimo Caputo
- Bristol Heart Institute, School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| | - David E Newby
- British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Gillian A Gray
- British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Andrew H Baker
- British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Andrea Caporali
- British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Marco Meloni
- British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
27
|
Ma C, Peng P, Zhou Y, Liu T, Wang L, Lu C. MicroRNA‑93 promotes angiogenesis and attenuates remodeling via inactivation of the Hippo/Yap pathway by targeting Lats2 after myocardial infarctionω. Mol Med Rep 2020; 22:483-493. [PMID: 32319642 PMCID: PMC7248469 DOI: 10.3892/mmr.2020.11085] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 04/01/2020] [Indexed: 01/10/2023] Open
Abstract
Inactivation of the Hippo pathway protects the myocardium from cardiac ischemic injury. MicroRNAs (miRs) have been reported to play pivotal roles in the progression of myocardial infarction (MI). The present study examined whether miR-93 could promote angiogenesis and attenuate remodeling after MI via inactivation of the Hippo/Yes-associated protein (Yap) pathway, by targeting large tumor suppressor kinase 2 (Lats2). It was identified that transfection of human umbilical vein endothelial cells with miR-93 mimic significantly decreased Lats2 expression and Yap phosphorylation, increased cell viability and migration, and attenuated cell apoptosis following hypoxia/reoxygenation injury. Moreover, increased expression of miR-93 resulted in an improvement of cardiac function, promotion of angiogenesis and attenuation of remodeling after MI. Additionally, miR-93 overexpression significantly decreased intracellular adhesion molecule 1 and vascular cell adhesion protein 1 expression levels, as well as attenuated the infiltration of neutrophils and macrophages into the myocardium after MI. Furthermore, it was found that miR-93 overexpression significantly suppressed Lats2 expression and decreased the levels of phosphorylated Yap in the myocardium after MI. Collectively, the present results suggested that miR-93 may exert a protective effect against MI via inactivation of the Hippo/Yap pathway by targeting Lats2.
Collapse
Affiliation(s)
- Chengjie Ma
- Clinical and Research Center of Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, P.R. China
| | - Peipei Peng
- Department of Anesthesiology, First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Yan Zhou
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Tianya Liu
- Jiangsu Province Key Laboratory of Anesthesiology, School of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Lijuan Wang
- Jiangsu Province Key Laboratory of Anesthesiology, School of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Chen Lu
- Jiangsu Province Key Laboratory of Anesthesiology, School of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| |
Collapse
|
28
|
Seok H. A New Member of Myocardial Ischemia-Reperfusion (MI/R) Associated miRNAs, miR-484: Its Potential Cardiac Protection Role. Korean Circ J 2020; 50:264-266. [PMID: 32100482 PMCID: PMC7043955 DOI: 10.4070/kcj.2019.0413] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 12/27/2019] [Indexed: 11/11/2022] Open
Affiliation(s)
- Heeyoung Seok
- Division of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea.
| |
Collapse
|
29
|
Mi L, Sun Y, Shi L, Li T. Hemin-Bridged MOF Interface with Double Amplification of G-Quadruplex Payload and DNAzyme Catalysis: Ultrasensitive Lasting Chemiluminescence MicroRNA Imaging. ACS APPLIED MATERIALS & INTERFACES 2020; 12:7879-7887. [PMID: 31983198 DOI: 10.1021/acsami.9b18053] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Here, we report a double-amplified sensing platform for ultrasensitive chemiluminescence (CL) miRNA detection in real patients' blood in which a hemin-bridged metal-organic framework (MOF) is employed as a functional interface to boost the payload and catalysis of G-quadruplex (G4) DNAzymes. Hemin is here used as the organic ligand for the MOF synthesis, which endows the MOF with an intrinsic peroxidase-like catalytic activity. Most importantly, the MOF surface provides a large amount of binding sites for polymeric G4 DNAzymes that are produced by miRNA-triggered rolling circle amplification reactions, and meanwhile, the interfaced G4 DNAzymes on MOFs (G4/MOFzymes) display an about 100-fold higher catalytic activity than those in solution. By using the G4/MOFzyme catalysts in the luminol/H2O2 CL system, the amplification detection of two acute myocardial infarction (AMI)-related miRNAs (low to 1 fM seen with naked eyes) is achieved in human serum with a smartphone as a portable imaging detector, which provides a facile methodology for point-of-care (POC) diagnosis of AMI. Compared with previous smartphone-based counterparts not requiring sophisticated equipment, this new facile methodology shows both 6 orders of magnitude higher sensitivity and an ∼50-fold longer duration for CL miRNA imaging. These unique features allow our developed G4/MOFzymes to be further employed as a novel luminescent ink for printing commonly used patterns.
Collapse
Affiliation(s)
- Lan Mi
- Department of Chemistry , University of Science and Technology of China , Hefei , Anhui 230026 , China
| | - Yudie Sun
- Department of Chemistry , University of Science and Technology of China , Hefei , Anhui 230026 , China
| | - Lin Shi
- Department of Chemistry , University of Science and Technology of China , Hefei , Anhui 230026 , China
| | - Tao Li
- Department of Chemistry , University of Science and Technology of China , Hefei , Anhui 230026 , China
| |
Collapse
|
30
|
Kura B, Kalocayova B, Devaux Y, Bartekova M. Potential Clinical Implications of miR-1 and miR-21 in Heart Disease and Cardioprotection. Int J Mol Sci 2020; 21:ijms21030700. [PMID: 31973111 PMCID: PMC7037063 DOI: 10.3390/ijms21030700] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 01/10/2020] [Accepted: 01/17/2020] [Indexed: 02/07/2023] Open
Abstract
The interest in non-coding RNAs, which started more than a decade ago, has still not weakened. A wealth of experimental and clinical studies has suggested the potential of non-coding RNAs, especially the short-sized microRNAs (miRs), to be used as the new generation of therapeutic targets and biomarkers of cardiovascular disease, an ever-growing public health issue in the modern world. Among the hundreds of miRs characterized so far, microRNA-1 (miR-1) and microRNA-21 (miR-21) have received some attention and have been associated with cardiac injury and cardioprotection. In this review article, we summarize the current knowledge of the function of these two miRs in the heart, their association with cardiac injury, and their potential cardioprotective roles and biomarker value. While this field has already been extensively studied, much remains to be done before research findings can be translated into clinical application for patient’s benefit.
Collapse
Affiliation(s)
- Branislav Kura
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 84104 Bratislava, Slovakia; (B.K.); (B.K.)
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, 81372 Bratislava, Slovakia
| | - Barbora Kalocayova
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 84104 Bratislava, Slovakia; (B.K.); (B.K.)
| | - Yvan Devaux
- Cardiovascular Research Unit, Department of Population Health, Luxembourg Institute of Health, L-1445 Strassen, Luxembourg;
| | - Monika Bartekova
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 84104 Bratislava, Slovakia; (B.K.); (B.K.)
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, 81372 Bratislava, Slovakia
- Correspondence: ; Tel.: +421-2-3229-5427
| |
Collapse
|
31
|
Wu T, Cui C, Huang Y, Liu Y, Fan C, Han X, Yang Y, Xu Z, Liu B, Fan G, Liu W. Coadministration of an Adhesive Conductive Hydrogel Patch and an Injectable Hydrogel to Treat Myocardial Infarction. ACS APPLIED MATERIALS & INTERFACES 2020; 12:2039-2048. [PMID: 31859471 DOI: 10.1021/acsami.9b17907] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Over the past decade, tissue-engineering strategies, mainly involving injectable hydrogels and epicardial biomaterial patches, have been pursued to treat myocardial infarction. However, only limited therapeutic efficacy is achieved with a single means. Here, a combined therapy approach is proposed, that is, the coadministration of a conductive hydrogel patch and injectable hydrogel to the infarcted myocardium. The self-adhesive conductive hydrogel patch is fabricated based on Fe3+-induced ionic coordination between dopamine-gelatin (GelDA) conjugates and dopamine-functionalized polypyrrole (DA-PPy), which form a homogeneous network. The injectable and cleavable hydrogel is formed in situ via a Schiff base reaction between oxidized sodium hyaluronic acid (HA-CHO) and hydrazided hyaluronic acid (HHA). Compared with a single-mode system, injecting the HA-CHO/HHA hydrogel intramyocardially followed by painting a conductive GelDA/DA-PPy hydrogel patch on the heart surface results in a more pronounced improvement of the cardiac function in terms of echocardiographical, histological, and angiogenic outcomes.
Collapse
Affiliation(s)
- Tengling Wu
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials , Tianjin University , Tianjin 300350 , China
| | - Chunyan Cui
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials , Tianjin University , Tianjin 300350 , China
| | - Yuting Huang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Medical Experiment Center , Tianjin University of Traditional Chinese Medicine , Tianjin 300193 , China
| | - Yang Liu
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials , Tianjin University , Tianjin 300350 , China
| | - Chuanchuan Fan
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials , Tianjin University , Tianjin 300350 , China
| | - Xiaoxu Han
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials , Tianjin University , Tianjin 300350 , China
| | - Yang Yang
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials , Tianjin University , Tianjin 300350 , China
| | - Ziyang Xu
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials , Tianjin University , Tianjin 300350 , China
| | - Bo Liu
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials , Tianjin University , Tianjin 300350 , China
| | - Guanwei Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Medical Experiment Center , Tianjin University of Traditional Chinese Medicine , Tianjin 300193 , China
| | - Wenguang Liu
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials , Tianjin University , Tianjin 300350 , China
| |
Collapse
|
32
|
Mitochondrial MiRNA in Cardiovascular Function and Disease. Cells 2019; 8:cells8121475. [PMID: 31766319 PMCID: PMC6952824 DOI: 10.3390/cells8121475] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 11/13/2019] [Accepted: 11/18/2019] [Indexed: 12/19/2022] Open
Abstract
MicroRNAs (miRNAs) are small noncoding RNAs functioning as crucial post-transcriptional regulators of gene expression involved in cardiovascular development and health. Recently, mitochondrial miRNAs (mitomiRs) have been shown to modulate the translational activity of the mitochondrial genome and regulating mitochondrial protein expression and function. Although mitochondria have been verified to be essential for the development and as a therapeutic target for cardiovascular diseases, we are just beginning to understand the roles of mitomiRs in the regulation of crucial biological processes, including energy metabolism, oxidative stress, inflammation, and apoptosis. In this review, we summarize recent findings regarding how mitomiRs impact on mitochondrial gene expression and mitochondrial function, which may help us better understand the contribution of mitomiRs to both the regulation of cardiovascular function under physiological conditions and the pathogenesis of cardiovascular diseases.
Collapse
|
33
|
MicroRNA-21 Mediates the Protective Effect of Cardiomyocyte-Derived Conditioned Medium on Ameliorating Myocardial Infarction in Rats. Cells 2019; 8:cells8080935. [PMID: 31430983 PMCID: PMC6721717 DOI: 10.3390/cells8080935] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 08/13/2019] [Accepted: 08/16/2019] [Indexed: 01/07/2023] Open
Abstract
Conditioned medium derived from ischemic myocardium improves rodent cardiac function after myocardial infarction. Exosomal miRNA-mediated intercellular communication is considered to mediate the protective effect of conditioned medium against ischemic injury. Oxygen–glucose-deprivation (OGD)-treated cardiac cells and a rat model with acute myocardial infarction (AMI) were applied. The expression profiles of myocardial-disease-associated miRNAs in cardiomyocytes, cardiac fibroblasts, ventricular myocardium, and conditioned medium derived from cardiomyocytes under ischemic stresses were analyzed. Primary cultured cell model and a rat model with myocardial infarction were applied to examine the role of miRNA in regulating cardiomyocyte apoptosis, fibroblast activation, immune cell infiltration, and myocardial infarction. Results showed that expression levels of miR-21 in cardiomyocytes, cardiac fibroblasts, and conditioned medium (CM) derived from cardiomyocytes were up-regulated with OGD treatment. With the depletion of miR-21, the protective effect of CM on cardiomyocytes against oxidative stress, enhanced fibroblast activation, and promotion of angiogenesis in endothelial cells were reduced. Administration of CM reduced the infarcted size and immune cell infiltration in myocardium of rats with AMI, while depletion of miR-21 reduced the effect of CM. In conclusion, miR-21 plays a role in intercellular communication among ischemic cardiac cells. The expression of miR-21 is important for the protective effect of conditioned medium against myocardial infarction.
Collapse
|
34
|
Zhang Y, Hou YM, Gao F, Xiao JW, Li CC, Tang Y. lncRNA GAS5 regulates myocardial infarction by targeting the miR-525-5p/CALM2 axis. J Cell Biochem 2019; 120:18678-18688. [PMID: 31429119 DOI: 10.1002/jcb.29156] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 05/16/2019] [Accepted: 05/22/2019] [Indexed: 12/12/2022]
Abstract
Long noncoding RNAs (lncRNAs) play critical roles in the pathogenesis of cardiovascular diseases, especially in myocardial infarction (MI). However, the underlying molecular mechanism of how lncRNA involves and affect MI still remains unclear. This study aimed to investigate the expression of lncRNA growth arrest-specific transcript 5 (GAS5) and its effects on myocardial cells' proliferation, cell cycle, and apoptosis. The possible mechanisms involved in GAS5, calmodulin 2 (CALM2), and microRNA (miR)-525-5p were also explored. The messenger RNA (mRNA) level of CALM2, GAS5, and miR-525-5p in postmyocardial infarction (MI) and normal cells were examined by quantitative real-time polymerase chain reaction (RT-qPCR). Western blot analysis assay was conducted to detect the protein levels of CALM2. The changes of cell cycle/apoptosis and cell viability of post-MI myocardial cells (PMMC) were determined by flow cytometry analysis and MTT (3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide) assay after knockdown of GAS5 or CALM2, respectively. Dual luciferase reporter assay and RNA-binding protein immunoprecipitation (RIP) assay were performed to verify the targeting relationship between miR-525-5p and GAS5, CALM2 in myocardial. Hypoxic preconditioning was performed in normal cells, which constructed a simulated MI environment, and the effect of GAS5 on cardiomyocyte apoptosis was detected. Our data showed that the expression of GAS5 and CALM2 in PMMC was significantly upregulated, while the expression of miR-525-5p was downregulated. Overexpression of GAS5 and CALM2 profoundly promoted the apoptosis of myocardial cell. However, the proliferation of myocardial cell was inhibited by the upregulation of GAS5 and CALM2. Moreover, GAS5 was proved to be the target of miR-525-5p and GAS5 downregulated the expression of miR-525-5p and CALM2. In addition, lncRNA GAS5 promotes MI, while CALM2 induced MI can be reversed by miR-525-5p. These data suggested that lncRNA GAS5 promoted the development and progression of MI via targeting of the miR-525-5p/CALM2 axis and it has the potential to be explored as a therapeutic target for the treatment of MI in the future.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Geratology, Affiliated Fengxian Hospital of Southern Medical University, Shenzhen, China.,Department of Cardiology, Affiliated Fuzhou First Hospital of Fujian Medical University, Fuzhou, Fujian Province, China
| | - Yue-Mei Hou
- Department of Geratology, Affiliated Fengxian Hospital of Southern Medical University, Shenzhen, China
| | - Feng Gao
- Department of Geratology, Affiliated Fengxian Hospital of Southern Medical University, Shenzhen, China.,Department of Cardiology, Affiliated Xiamen Zhongshan Hospital of Xiamen University, Xiamen, Fujian Province, China
| | - Jin-Wen Xiao
- Department of Cardiology, Affiliated Fuzhou First Hospital of Fujian Medical University, Fuzhou, Fujian Province, China
| | - Chuan-Chuan Li
- Department of Cardiology, Affiliated Fuzhou First Hospital of Fujian Medical University, Fuzhou, Fujian Province, China
| | - Yuan Tang
- Department of Cardiology, Affiliated Fuzhou First Hospital of Fujian Medical University, Fuzhou, Fujian Province, China
| |
Collapse
|
35
|
Qian L, Pan S, Shi L, Zhou Y, Sun L, Wan Z, Ding Y, Qian J. Downregulation of microRNA-218 is cardioprotective against cardiac fibrosis and cardiac function impairment in myocardial infarction by binding to MITF. Aging (Albany NY) 2019; 11:5368-5388. [PMID: 31408435 PMCID: PMC6710048 DOI: 10.18632/aging.102112] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 07/16/2019] [Indexed: 04/14/2023]
Abstract
OBJECTIVE This study is intended to figure out the function of microRNA-218 (miR-218) together with microphthalmia-associated transcription factor (MITF) on the cardiac fibrosis and cardiac function impairment in rat models of myocardial infarction (MI). RESULTS The rats with MI exhibited cardiac function impairment, cardiac fibrosis, oxidative stress, cardiomyocyte apoptosis, as well as inflammatory injury. Additionally, upregulated miR-218 and downregulated MITF were detected in cardiac tissues of MI rats. MI rats injected with miR-218 inhibitors or overexpressed MITF exhibited elevated MITF expression, improved cardiac function, and diminished pathological damages, infarct size, cardiomyocyte apoptosis, cardiac fibrosis, oxidative stress as well as inflammatory injury in cardiac tissues. Furthermore, downregulated miR-218 and MITF aggravated the conditions than downregulation of miR-218 alone in MI rats. METHODS MI models were performed in rats, and then the rats were injected with miR-218 inhibitors and/or MITF overexpression plasmid to elucidate the role of miR-218 and/or MITF on the cardiac function, pathological damage, cardiac fibrosis, angiogenesis, oxidative stress and inflammatory injury of cardiac tissues in MI rats by performing a series of assays. CONCLUSION Collectively, we found that the suppression of miR-218 alleviates cardiac fibrosis and cardiac function impairment, and stimulates angiogenesis in MI rats through inhibiting MITF.
Collapse
Affiliation(s)
- Linfeng Qian
- Department of Cardiothoracic Surgery, First Affiliated Hospital of Zhejiang University, Hangzhou 310003, PR. China
| | - Shaobo Pan
- Operating Room, First Affiliated Hospital of Zhejiang University, Hangzhou 310003, PR. China
| | - Liping Shi
- Department of Cardiothoracic Surgery, First Affiliated Hospital of Zhejiang University, Hangzhou 310003, PR. China
| | - Yongyi Zhou
- Department of Cardiothoracic Surgery, First Affiliated Hospital of Zhejiang University, Hangzhou 310003, PR. China
| | - Lai Sun
- Department of Cardiothoracic Surgery, First Affiliated Hospital of Zhejiang University, Hangzhou 310003, PR. China
| | - Zhedong Wan
- Department of Cardiothoracic Surgery, First Affiliated Hospital of Zhejiang University, Hangzhou 310003, PR. China
| | - Yufang Ding
- Operating Room, First Affiliated Hospital of Zhejiang University, Hangzhou 310003, PR. China
| | - Jia Qian
- Operating Room, First Affiliated Hospital of Zhejiang University, Hangzhou 310003, PR. China
| |
Collapse
|
36
|
Deutsch MA, Doppler SA, Li X, Lahm H, Santamaria G, Cuda G, Eichhorn S, Ratschiller T, Dzilic E, Dreßen M, Eckart A, Stark K, Massberg S, Bartels A, Rischpler C, Gilsbach R, Hein L, Fleischmann BK, Wu SM, Lange R, Krane M. Reactivation of the Nkx2.5 cardiac enhancer after myocardial infarction does not presage myogenesis. Cardiovasc Res 2019; 114:1098-1114. [PMID: 29579159 DOI: 10.1093/cvr/cvy069] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 03/15/2018] [Indexed: 12/13/2022] Open
Abstract
Aims The contribution of resident stem or progenitor cells to cardiomyocyte renewal after injury in adult mammalian hearts remains a matter of considerable debate. We evaluated a cell population in the adult mouse heart induced by myocardial infarction (MI) and characterized by an activated Nkx2.5 enhancer element that is specific for multipotent cardiac progenitor cells (CPCs) during embryonic development. We hypothesized that these MI-induced cells (MICs) harbour cardiomyogenic properties similar to their embryonic counterparts. Methods and results MICs reside in the heart and mainly localize to the infarction area and border zone. Interestingly, gene expression profiling of purified MICs 1 week after infarction revealed increased expression of stem cell markers and embryonic cardiac transcription factors (TFs) in these cells as compared to the non-mycoyte cell fraction of adult hearts. A subsequent global transcriptome comparison with embryonic CPCs and fibroblasts and in vitro culture of MICs unveiled that (myo-)fibroblastic features predominated and that cardiac TFs were only expressed at background levels. Conclusions Adult injury-induced reactivation of a cardiac-specific Nkx2.5 enhancer element known to specifically mark myocardial progenitor cells during embryonic development does not reflect hypothesized embryonic cardiomyogenic properties. Our data suggest a decreasing plasticity of cardiac progenitor (-like) cell populations with increasing age. A re-expression of embryonic, stem or progenitor cell features in the adult heart must be interpreted very carefully with respect to the definition of cardiac resident progenitor cells. Albeit, the abundance of scar formation after cardiac injury suggests a potential to target predestinated activated profibrotic cells to push them towards cardiomyogenic differentiation to improve regeneration.
Collapse
Affiliation(s)
- Marcus-André Deutsch
- Department of Cardiovascular Surgery, German Heart Center Munich at the Technische Universität München, Lazarettstraße 36, 80636 Munich, Germany.,Department of Cardiovascular Surgery, German Heart Center, Insure (Institute for Translational Cardiac Surgery), Technische Universität München, Lothstraße 11, 80636 Munich, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Stefanie A Doppler
- Department of Cardiovascular Surgery, German Heart Center Munich at the Technische Universität München, Lazarettstraße 36, 80636 Munich, Germany.,Department of Cardiovascular Surgery, German Heart Center, Insure (Institute for Translational Cardiac Surgery), Technische Universität München, Lothstraße 11, 80636 Munich, Germany
| | - Xinghai Li
- Department of Cardiovascular Surgery, German Heart Center Munich at the Technische Universität München, Lazarettstraße 36, 80636 Munich, Germany.,Department of Cardiovascular Surgery, German Heart Center, Insure (Institute for Translational Cardiac Surgery), Technische Universität München, Lothstraße 11, 80636 Munich, Germany
| | - Harald Lahm
- Department of Cardiovascular Surgery, German Heart Center Munich at the Technische Universität München, Lazarettstraße 36, 80636 Munich, Germany.,Department of Cardiovascular Surgery, German Heart Center, Insure (Institute for Translational Cardiac Surgery), Technische Universität München, Lothstraße 11, 80636 Munich, Germany
| | - Gianluca Santamaria
- Stem Cell Laboratory, Department of Experimental and Clinical Medicine, Research Center of Advanced Biochemistry and Molecular Biology.,CIS (Centro Interdisciplinare Servizi), University 'Magna Graecia' of Catanzaro, Viale Europa, 88100 Catanzaro, Italy
| | - Giovanni Cuda
- Stem Cell Laboratory, Department of Experimental and Clinical Medicine, Research Center of Advanced Biochemistry and Molecular Biology
| | - Stefan Eichhorn
- Department of Cardiovascular Surgery, German Heart Center Munich at the Technische Universität München, Lazarettstraße 36, 80636 Munich, Germany.,Department of Cardiovascular Surgery, German Heart Center, Insure (Institute for Translational Cardiac Surgery), Technische Universität München, Lothstraße 11, 80636 Munich, Germany
| | - Thomas Ratschiller
- Department of Cardiothoracic and Vascular Surgery, Kepler University Hospital, 4021 Linz, Austria
| | - Elda Dzilic
- Department of Cardiovascular Surgery, German Heart Center Munich at the Technische Universität München, Lazarettstraße 36, 80636 Munich, Germany.,Department of Cardiovascular Surgery, German Heart Center, Insure (Institute for Translational Cardiac Surgery), Technische Universität München, Lothstraße 11, 80636 Munich, Germany
| | - Martina Dreßen
- Department of Cardiovascular Surgery, German Heart Center Munich at the Technische Universität München, Lazarettstraße 36, 80636 Munich, Germany.,Department of Cardiovascular Surgery, German Heart Center, Insure (Institute for Translational Cardiac Surgery), Technische Universität München, Lothstraße 11, 80636 Munich, Germany
| | - Annekathrin Eckart
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, 81377 Munich, Germany
| | - Konstantin Stark
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.,Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, 81377 Munich, Germany
| | - Steffen Massberg
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.,Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, 81377 Munich, Germany
| | - Anna Bartels
- Nuklearmedizinische Klinik des Klinikums Rechts der Isar, Technische Universität München, 81675 Munich, Germany
| | - Christoph Rischpler
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.,Nuklearmedizinische Klinik des Klinikums Rechts der Isar, Technische Universität München, 81675 Munich, Germany
| | - Ralf Gilsbach
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Albertstraße 25, 79104 Freiburg, Germany
| | - Lutz Hein
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Albertstraße 25, 79104 Freiburg, Germany.,BIOSS Centre for Biological Signaling Studies, University of Freiburg, Schänzlestraße 18, 79104 Freiburg, Germany
| | - Bernd K Fleischmann
- Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, 53105 Bonn, Germany
| | - Sean M Wu
- Division of Cardiovascular Medicine, Department of Medicine, Stanford Cardiovascular Institute, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, USA
| | - Rüdiger Lange
- Department of Cardiovascular Surgery, German Heart Center Munich at the Technische Universität München, Lazarettstraße 36, 80636 Munich, Germany.,Department of Cardiovascular Surgery, German Heart Center, Insure (Institute for Translational Cardiac Surgery), Technische Universität München, Lothstraße 11, 80636 Munich, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Markus Krane
- Department of Cardiovascular Surgery, German Heart Center Munich at the Technische Universität München, Lazarettstraße 36, 80636 Munich, Germany.,Department of Cardiovascular Surgery, German Heart Center, Insure (Institute for Translational Cardiac Surgery), Technische Universität München, Lothstraße 11, 80636 Munich, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
37
|
Lu Y, Thavarajah T, Gu W, Cai J, Xu Q. Impact of miRNA in Atherosclerosis. Arterioscler Thromb Vasc Biol 2019; 38:e159-e170. [PMID: 30354259 DOI: 10.1161/atvbaha.118.310227] [Citation(s) in RCA: 155] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Yao Lu
- From the Center of Clinical Pharmacology (Y.L.)
| | - Tanuja Thavarajah
- School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre, United Kingdom (T.T., W.G., Q.X.)
| | - Wenduo Gu
- School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre, United Kingdom (T.T., W.G., Q.X.)
| | - Jingjing Cai
- Department of Cardiology (J.C., Q.X.), Third Xiangya Hospital, Central South University, Changsha, China
| | - Qingbo Xu
- Department of Cardiology (J.C., Q.X.), Third Xiangya Hospital, Central South University, Changsha, China.,School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre, United Kingdom (T.T., W.G., Q.X.)
| |
Collapse
|
38
|
Antunes JC, Benarroch L, Moraes FC, Juenet M, Gross MS, Aubart M, Boileau C, Caligiuri G, Nicoletti A, Ollivier V, Chaubet F, Letourneur D, Chauvierre C. Core-Shell Polymer-Based Nanoparticles Deliver miR-155-5p to Endothelial Cells. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 17:210-222. [PMID: 31265949 PMCID: PMC6610682 DOI: 10.1016/j.omtn.2019.05.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 05/20/2019] [Accepted: 05/20/2019] [Indexed: 12/12/2022]
Abstract
Heart failure occurs in over 30% of the worldwide population and most commonly originates from cardiovascular diseases such as myocardial infarction. microRNAs (miRNAs) target and silence specific mRNAs, thereby regulating gene expression. Because the endogenous miR-155-5p has been ascribed to vasculoprotection, loading it onto positively charged, core-shell poly(isobutylcyanoacrylate) (PIBCA)-polysaccharide nanoparticles (NPs) was attempted. NPs showed a decrease (p < 0.0001) in surface electrical charge (ζ potential), with negligible changes in size or shape when loaded with the anionic miR-155-5p. Presence of miR-155-5p in loaded NPs was further quantified. Cytocompatibility up to 100 μg/mL of NPs for 2 days with human coronary artery endothelial cells (hCAECs) was documented. NPs were able to enter hCAECs and were localized in the endoplasmic reticulum (ER). Expression of miR-155-5p was increased within the cells by 75-fold after 4 hours of incubation (p < 0.05) and was still noticeable at day 2. Differences between loaded NP-cultured cells and free miRNA, at days 1 (p < 0.05) and 2 (p < 0.001) suggest the ability of prolonged load release in physiological conditions. Expression of miR-155-5p downstream target BACH1 was decreased in the cells by 4-fold after 1 day of incubation (p < 0.05). This study is a first proof of concept that miR-155-5p can be loaded onto NPs and remain intact and biologically active in endothelial cells (ECs). These nanosystems could potentially increase an endogenous cytoprotective response and decrease damage within infarcted hearts.
Collapse
Affiliation(s)
- Joana C Antunes
- Université de Paris, LVTS, INSERM U1148, Université Paris 13, 75018 Paris, France
| | - Louise Benarroch
- Université de Paris, LVTS, INSERM U1148, Université Paris 13, 75018 Paris, France
| | - Fernanda C Moraes
- Université de Paris, LVTS, INSERM U1148, Université Paris 13, 75018 Paris, France
| | - Maya Juenet
- Université de Paris, LVTS, INSERM U1148, Université Paris 13, 75018 Paris, France
| | - Marie-Sylvie Gross
- Université de Paris, LVTS, INSERM U1148, Université Paris 13, 75018 Paris, France
| | - Mélodie Aubart
- Université de Paris, LVTS, INSERM U1148, Université Paris 13, 75018 Paris, France
| | - Catherine Boileau
- Université de Paris, LVTS, INSERM U1148, Université Paris 13, 75018 Paris, France
| | - Giuseppina Caligiuri
- Université de Paris, LVTS, INSERM U1148, Université Paris 13, 75018 Paris, France
| | - Antonino Nicoletti
- Université de Paris, LVTS, INSERM U1148, Université Paris 13, 75018 Paris, France
| | - Véronique Ollivier
- Université de Paris, LVTS, INSERM U1148, Université Paris 13, 75018 Paris, France
| | - Frédéric Chaubet
- Université de Paris, LVTS, INSERM U1148, Université Paris 13, 75018 Paris, France
| | - Didier Letourneur
- Université de Paris, LVTS, INSERM U1148, Université Paris 13, 75018 Paris, France
| | - Cédric Chauvierre
- Université de Paris, LVTS, INSERM U1148, Université Paris 13, 75018 Paris, France.
| |
Collapse
|
39
|
Cai Y, Li Y. Upregulation of miR-29b-3p protects cardiomyocytes from hypoxia-induced apoptosis by targeting TRAF5. Cell Mol Biol Lett 2019; 24:27. [PMID: 31011336 PMCID: PMC6460541 DOI: 10.1186/s11658-019-0151-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 03/29/2019] [Indexed: 12/18/2022] Open
Abstract
Background MicroRNAs (miRNAs) are pivotal regulators in regulating hypoxia-induced cardiomyocyte injury. This study was designed to evaluate the effects of miR-29b-3p on hypoxic cardiomyocytes. Methods Human AC16 cells were cultured under normoxic or hypoxic conditions. Hypoxic injury was confirmed based on alterations in cell viability using CCK-8 assay and apoptosis using flow cytometry and Hoechst staining. Bioinformatics analyses and the dual-luciferase reporter assay were performed to predict and validate the target gene of miR-29b-3p. Results We found that hypoxia suppressed cell viability and promoted apoptosis. TNF receptor-associated factor 5 (TRAF5) was a potential target gene of miR-29b-3p. Our in vitro experiments revealed that miR-29b-3p overexpression or TRAF6 knockdown significantly protected cardiomyocytes against hypoxia-induced injury. Moreover, knockdown of TRAF5 knockdown potentiated the protective effects of miR-29b-3p against hypoxia-induced cell injury. Conclusion These findings suggest that upregulation of miR-29b-3p could protect cardiomyocytes against hypoxia-induced injury through downregulation of TRAF5. Targeting TRAF5 with miR-29b-3p might be a potential therapeutic method for AMI.
Collapse
Affiliation(s)
- Yuhua Cai
- Department of Cardiovasology, Jingzhou First Municipal Hospital, Jingzhou, Hubei Province China
| | - Yunpeng Li
- 2Department of Cardiovasology, Dongfeng Hospital, Hubei University of Medicine, No. 16 Daling Road, Shiyan, 442008 Hubei Province China
| |
Collapse
|
40
|
|
41
|
Sun Y, Shi L, Wang Q, Mi L, Li T. Spherical Nucleic Acid Enzyme (SNAzyme) Boosted Chemiluminescence miRNA Imaging Using a Smartphone. Anal Chem 2019; 91:3652-3658. [DOI: 10.1021/acs.analchem.8b05696] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Yudie Sun
- Department of Chemistry, University of Science & Technology of China, Hefei, Anhui 230026, China
| | - Lin Shi
- Department of Chemistry, University of Science & Technology of China, Hefei, Anhui 230026, China
| | - Qiwei Wang
- Department of Chemistry, University of Science & Technology of China, Hefei, Anhui 230026, China
| | - Lan Mi
- Department of Chemistry, University of Science & Technology of China, Hefei, Anhui 230026, China
| | - Tao Li
- Department of Chemistry, University of Science & Technology of China, Hefei, Anhui 230026, China
| |
Collapse
|
42
|
Delineating the Dynamic Transcriptome Response of mRNA and microRNA during Zebrafish Heart Regeneration. Biomolecules 2018; 9:biom9010011. [PMID: 30597924 PMCID: PMC6359357 DOI: 10.3390/biom9010011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 12/19/2018] [Accepted: 12/21/2018] [Indexed: 12/12/2022] Open
Abstract
Heart diseases are the leading cause of death for the vast majority of people around the world, which is often due to the limited capability of human cardiac regeneration. In contrast, zebrafish have the capacity to fully regenerate their hearts after cardiac injury. Understanding and activating these mechanisms would improve health in patients suffering from long-term consequences of ischemia. Therefore, we monitored the dynamic transcriptome response of both mRNA and microRNA in zebrafish at 1–160 days post cryoinjury (dpi). Using a control model of sham-operated and healthy fish, we extracted the regeneration specific response and further delineated the spatio-temporal organization of regeneration processes such as cell cycle and heart function. In addition, we identified novel (miR-148/152, miR-218b and miR-19) and previously known microRNAs among the top regulators of heart regeneration by using theoretically predicted target sites and correlation of expression profiles from both mRNA and microRNA. In a cross-species effort, we validated our findings in the dynamic process of rat myoblasts differentiating into cardiomyocytes-like cells (H9c2 cell line). Concluding, we elucidated different phases of transcriptomic responses during zebrafish heart regeneration. Furthermore, microRNAs showed to be important regulators in cardiomyocyte proliferation over time.
Collapse
|
43
|
Costantino S, Libby P, Kishore R, Tardif JC, El-Osta A, Paneni F. Epigenetics and precision medicine in cardiovascular patients: from basic concepts to the clinical arena. Eur Heart J 2018; 39:4150-4158. [PMID: 29069341 PMCID: PMC6293269 DOI: 10.1093/eurheartj/ehx568] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 09/04/2017] [Accepted: 09/22/2017] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular diseases (CVDs) remain the leading cause of mortality worldwide and also inflict major burdens on morbidity, quality of life, and societal costs. Considering that CVD preventive medications improve vascular outcomes in less than half of patients (often relative risk reductions range from 12% to 20% compared with placebo), precision medicine offers an attractive approach to refine the targeting of CVD medications to responsive individuals in a population and thus allocate resources more wisely and effectively. New tools furnished by advances in basic science and translational medicine could help achieve this goal. This approach could reach beyond the practitioners 'eyeball' assessment or venerable markers derived from the physical examination and standard laboratory evaluation. Advances in genetics have identified novel pathways and targets that operate in numerous diseases, paving the way for 'precision medicine'. Yet the inherited genome determines only part of an individual's risk profile. Indeed, standard genomic approaches do not take into account the world of regulation of gene expression by modifications of the 'epi'genome. Epigenetic modifications defined as 'heritable changes to the genome that do not involve changes in DNA sequence' have emerged as a new layer of biological regulation in CVD and could advance individualized risk assessment as well as devising and deploying tailored therapies. This review, therefore, aims to acquaint the cardiovascular community with the rapidly advancing and evolving field of epigenetics and its implications in cardiovascular precision medicine.
Collapse
Affiliation(s)
- Sarah Costantino
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, Schlieren, Zurich, Switzerland
| | - Peter Libby
- Brigham and Women’s Hospital, Division of Cardiovascular Medicine, Boston, MA, USA
| | - Raj Kishore
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, MERB-953, 3500 N Broad Street, Philadelphia, PA, USA
- Department of Pharmacology, Temple University, Philadelphia, PA, USA
| | - Jean-Claude Tardif
- Montreal Health Innovations Coordinating Center (MHICC), Montreal, Canada
- Montreal Heart Institute, Université de Montréal, Montreal, Canada
| | - Assam El-Osta
- Central Clinical School, Faculty of Medicine, Monash University, Victoria, Australia
- Department of Pathology, The University of Melbourne, Parkville, Victoria, Australia
- Hong Kong Institute of Diabetes and Obesity, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR
| | - Francesco Paneni
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, Schlieren, Zurich, Switzerland
- University Heart Center, Cardiology, University Hospital Zürich, Zürich, Switzerland
| |
Collapse
|
44
|
Guedes EC, da Silva IB, Lima VM, Miranda JB, Albuquerque RP, Ferreira JCB, Barreto‐Chaves MLM, Diniz GP. High fat diet reduces the expression of miRNA‐29b in heart and increases susceptibility of myocardium to ischemia/reperfusion injury. J Cell Physiol 2018; 234:9399-9407. [DOI: 10.1002/jcp.27624] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 09/25/2018] [Indexed: 12/22/2022]
Affiliation(s)
- Elaine Castilho Guedes
- Department of Anatomy Laboratory of Cell Biology and Functional Anatomy, Institute of Biomedical Sciences, University of Sao Paulo Sao Paulo Brazil
| | - Ivson Bezerra da Silva
- Department of Anatomy Laboratory of Cell Biology and Functional Anatomy, Institute of Biomedical Sciences, University of Sao Paulo Sao Paulo Brazil
- Department of Morphology Health Sciences Center, Federal University of Paraiba Joao Pessoa Brazil
| | - Vanessa Morais Lima
- Department of Anatomy Laboratory of Cell Biology and Functional Anatomy, Institute of Biomedical Sciences, University of Sao Paulo Sao Paulo Brazil
| | - Juliane B. Miranda
- Department of Anatomy Laboratory of Cell Biology and Functional Anatomy, Institute of Biomedical Sciences, University of Sao Paulo Sao Paulo Brazil
| | - Rudá P. Albuquerque
- Department of Anatomy Laboratory of Integrative Systems Biology, Institute of Biomedical Sciences, University of Sao Paulo Sao Paulo Brazil
| | - Julio C. B. Ferreira
- Department of Anatomy Laboratory of Integrative Systems Biology, Institute of Biomedical Sciences, University of Sao Paulo Sao Paulo Brazil
| | - Maria Luiza M. Barreto‐Chaves
- Department of Anatomy Laboratory of Cell Biology and Functional Anatomy, Institute of Biomedical Sciences, University of Sao Paulo Sao Paulo Brazil
| | - Gabriela Placoná Diniz
- Department of Anatomy Laboratory of Cell Biology and Functional Anatomy, Institute of Biomedical Sciences, University of Sao Paulo Sao Paulo Brazil
| |
Collapse
|
45
|
Lakhani HV, Khanal T, Gabi A, Yousef G, Alam MB, Sharma D, Aljoudi H, Puri N, Thompson E, Shapiro JI, Sodhi K. Developing a panel of biomarkers and miRNA in patients with myocardial infarction for early intervention strategies of heart failure in West Virginian population. PLoS One 2018; 13:e0205329. [PMID: 30356307 PMCID: PMC6200226 DOI: 10.1371/journal.pone.0205329] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 09/24/2018] [Indexed: 12/30/2022] Open
Abstract
Background Myocardial infarction is the most common cause of heart failure. MI has been intricately linked to ventricular remodeling, subsequently leading to the reduction in the cardiac ejection fraction causing HF. The cumulative line of evidence suggests an important role of several biomarkers in modulating the cardiac vasculature, further contributing towards the progression of post-MI complications. Studies have demonstrated, yet not fully established, that an important biomarker, IL-10, has a causal relationship with MI and associated cardiac dysfunction. Hypothesis This study aims to establish the role of IL-10 as a prognostic marker for the cardiovascular outcomes and to develop a panel of biomarkers and circulating miRNAs that could potentially result in the early detection of HF resulting from MI, allowing for early intervention strategies. Methods and results Blood was withdrawn and echocardiography assessment was performed on a total of 43 patients that were enrolled, within 24 hours of the incidence of MI. Patients were divided in three main groups, based on the ejection fraction measurement from echocardiography: control (n = 14), MI with normal EF (MI+NEF, n = 13) and MI with low EF (MI+LEF, n = 16). Our results showed that TGFβ-1, TNF-α, IL-6 and MMP-9 were upregulated significantly in MI+NEF group and more so in MI+LEF group, as compared to control group (p<0.01). The circulating levels of miR-34a, miR-208b and miR-126 were positively correlated and showed elevated levels in the MI+NEF group, even higher in MI+LEF group, while levels of miR-24 and miR-29a were reduced in MI+NEF, and much lower in MI+LEF, as compared to the control group (p<0.01). Our results also demonstrated a direct correlation of IL-10 with the ejection fraction in patients with MI: IL-10 was elevated in MI+NEF group, however, the levels were significantly low in MI+LEF group suggesting an important role of IL-10 in predicting heart failure. Importantly, our study confirmed the correlation of IL-10 with EF by our follow-up echocardiography assessment that was performed 2 months after the incidence of MI. Conclusion Our results support the clinical application of these serum biomarkers to develop a panel for appropriate prognosis and management of adverse cardiac remodeling and development of heart failure post-myocardial infarction.
Collapse
Affiliation(s)
- Hari Vishal Lakhani
- Department of Internal Medicine, Marshall University Joan C Edwards School of Medicine, Huntington, WV, United States of America
| | - Tilak Khanal
- Department of Internal Medicine, Marshall University Joan C Edwards School of Medicine, Huntington, WV, United States of America
| | - Alaa Gabi
- Division of Cardiology, Department of Internal Medicine, Marshall University Joan C Edwards School of Medicine, Huntington, WV, United States of America
| | - George Yousef
- Division of Cardiology, Department of Internal Medicine, Marshall University Joan C Edwards School of Medicine, Huntington, WV, United States of America
| | - Mian Bilal Alam
- Division of Cardiology, Department of Internal Medicine, Marshall University Joan C Edwards School of Medicine, Huntington, WV, United States of America
| | - Dana Sharma
- Department of Internal Medicine, Marshall University Joan C Edwards School of Medicine, Huntington, WV, United States of America
| | - Haytham Aljoudi
- Division of Cardiology, Department of Internal Medicine, Marshall University Joan C Edwards School of Medicine, Huntington, WV, United States of America
| | - Nitin Puri
- Departments of Biomedical Sciences, Marshall University Joan C Edwards School of Medicine, Huntington, WV, United States of America
| | - Ellen Thompson
- Division of Cardiology, Department of Internal Medicine, Marshall University Joan C Edwards School of Medicine, Huntington, WV, United States of America
| | - Joseph I. Shapiro
- Department of Internal Medicine, Marshall University Joan C Edwards School of Medicine, Huntington, WV, United States of America
| | - Komal Sodhi
- Departments of Surgery and Biomedical Sciences, Marshall University Joan C Edwards School of Medicine, Huntington, WV, United States of America
- * E-mail:
| |
Collapse
|
46
|
Sun Y, Li T. Composition-Tunable Hollow Au/Ag SERS Nanoprobes Coupled with Target-Catalyzed Hairpin Assembly for Triple-Amplification Detection of miRNA. Anal Chem 2018; 90:11614-11621. [PMID: 30175580 DOI: 10.1021/acs.analchem.8b03067] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Detecting disease-related biomarkers is of great significance for disease diagnosis and therapy. In this work, we develop an ultrasensitive surface-enhanced Raman scattering (SERS) biosensor for the detection of an acute myocardial infarction-related miRNA (miR-133a) using composition-adjustable hollow Ag/Au nanosphere-based SERS probes coupled with the target-catalyzed hairpin assembly (CHA) strategy. Bimetallic probes displaying high stability and a strong surface plasmon resonance effect were synthesized with a controllable ratio of silver and gold by a galvanic replacement method and then captured by a duplex linker produced in the CHA process to accomplish signal amplification. In this way, the target miR-133a can be detected in a wide linear range with a detection limit of 0.306 fM and high selectivity over other miRNAs expressed in human hearts. Practical applications in human blood samples reveal the strong anti-interference ability and ideal sensitivity of our developed sensing platform in physiological environments, benefiting its potential biomedical applications.
Collapse
Affiliation(s)
- Yudie Sun
- Department of Chemistry , University of Science & Technology of China , Hefei , Anhui 230026 , China
| | - Tao Li
- Department of Chemistry , University of Science & Technology of China , Hefei , Anhui 230026 , China
| |
Collapse
|
47
|
Hassan S, Sidransky E, Tayebi N. The role of epigenetics in lysosomal storage disorders: Uncharted territory. Mol Genet Metab 2017; 122:10-18. [PMID: 28918065 DOI: 10.1016/j.ymgme.2017.07.012] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 07/30/2017] [Accepted: 07/31/2017] [Indexed: 12/18/2022]
Abstract
The study of the contribution of epigenetic mechanisms, including DNA methylation, histone modifications, and microRNAs, to human disease has enhanced our understanding of different cellular processes and diseased states, as well as the effect of environmental factors on phenotypic outcomes. Epigenetic studies may be particularly relevant in evaluating the clinical heterogeneity observed in monogenic disorders. The lysosomal storage disorders are Mendelian disorders characterized by a wide spectrum of associated phenotypes, ranging from neonatal presentations to symptoms that develop in late adulthood. Some lack a tight genotype/phenotype correlation. While epigenetics may explain some of the discordant phenotypes encountered in patients with the same lysosomal storage disorder, especially among patients sharing the same genotype, to date, few studies have focused on these mechanisms. We review three common epigenetic mechanisms, DNA methylation, histone modifications, and microRNAs, and highlight their applications to phenotypic variation and therapeutics. Three specific lysosomal storage diseases, Gaucher disease, Fabry disease, and Niemann-Pick type C disease are presented as prototypical disorders with vast clinical heterogeneity that may be impacted by epigenetics. Our goal is to motivate researchers to consider epigenetics as a mechanism to explain the complexities of biological functions and pathologies of these rare disorders.
Collapse
Affiliation(s)
- Shahzeb Hassan
- Medical Genetics Branch, NHGRI, NIH, Bethesda, MD, United States
| | - Ellen Sidransky
- Medical Genetics Branch, NHGRI, NIH, Bethesda, MD, United States.
| | - Nahid Tayebi
- Medical Genetics Branch, NHGRI, NIH, Bethesda, MD, United States
| |
Collapse
|
48
|
Mozos I, Malainer C, Horbańczuk J, Gug C, Stoian D, Luca CT, Atanasov AG. Inflammatory Markers for Arterial Stiffness in Cardiovascular Diseases. Front Immunol 2017; 8:1058. [PMID: 28912780 PMCID: PMC5583158 DOI: 10.3389/fimmu.2017.01058] [Citation(s) in RCA: 246] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 08/15/2017] [Indexed: 01/13/2023] Open
Abstract
Arterial stiffness predicts an increased risk of cardiovascular events. Inflammation plays a major role in large arteries stiffening, related to atherosclerosis, arteriosclerosis, endothelial dysfunction, smooth muscle cell migration, vascular calcification, increased activity of metalloproteinases, extracellular matrix degradation, oxidative stress, elastolysis, and degradation of collagen. The present paper reviews main mechanisms explaining the crosstalk between inflammation and arterial stiffness and the most common inflammatory markers associated with increased arterial stiffness, considering the most recent clinical and experimental studies. Diverse studies revealed significant correlations between the severity of arterial stiffness and inflammatory markers, such as white blood cell count, neutrophil/lymphocyte ratio, adhesion molecules, fibrinogen, C-reactive protein, cytokines, microRNAs, and cyclooxygenase-2, in patients with a broad variety of diseases, such as metabolic syndrome, diabetes, coronary heart disease, peripheral arterial disease, malignant and rheumatic disorders, polycystic kidney disease, renal transplant, familial Mediterranean fever, and oral infections, and in women with preeclampsia or after menopause. There is strong evidence that inflammation plays an important and, at least, partly reversible role in the development of arterial stiffness, and inflammatory markers may be useful additional tools in the assessment of the cardiovascular risk in clinical practice. Combined assessment of arterial stiffness and inflammatory markers may improve non-invasive assessment of cardiovascular risk, enabling selection of high-risk patients for prophylactic treatment or more regular medical examination. Development of future destiffening therapies may target pro-inflammatory mechanisms.
Collapse
Affiliation(s)
- Ioana Mozos
- Department of Functional Sciences, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania.,Center for Translational Research and Systems Medicine, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| | | | - Jarosław Horbańczuk
- The Institute of Genetics and Animal Breeding, Polish Academy of Sciences, Jastrzębiec, Poland
| | - Cristina Gug
- Department of Microscopic Morphology, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| | - Dana Stoian
- 2nd Department of Internal Medicine, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| | - Constantin Tudor Luca
- Department of Cardiology, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| | - Atanas G Atanasov
- The Institute of Genetics and Animal Breeding, Polish Academy of Sciences, Jastrzębiec, Poland.,Department of Pharmacognosy, Faculty of Life Sciences, University of Vienna, Vienna, Austria.,Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
49
|
Suppression of microRNA-142-5p attenuates hypoxia-induced apoptosis through targeting SIRT7. Biomed Pharmacother 2017; 94:394-401. [PMID: 28772218 DOI: 10.1016/j.biopha.2017.07.083] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 07/10/2017] [Accepted: 07/19/2017] [Indexed: 12/21/2022] Open
Abstract
Increasing study has suggested that microRNAs (miRNAs) are pivotal regulators in regulating hypoxia-induced injury. miR-142-5p has been suggested as a critical regulator for cellular survival. However, the role of miR-142-5p in regulating hypoxia-induced injury remains unknown. In this study, we aimed to investigate the mechanistic roles of miR-142-5p in regulating cell survival during hypoxia treatment using H9C2 cardiomyoblasts and primary cardiomyocytes. We showed that miR-142-5p expression level was significantly repressed by hypoxia treatment. Overexpression of miR-142-5p during hypoxia induced extensive cell injury and apoptosis whereas suppression of miR-142-5p significantly promoted cell viability and attenuated cell apoptosis with hypoxia treatment. Sirtuin7 (SIRT7) was identified as a direct target gene of miR-142-5p by bioinformatics analysis and dual-luciferase reporter assays. Overexpression of miR-142-5p significantly decreased SIRT7 expression, while suppression of miR-142-5p increased SIRT7 expression. Furthermore, overexpression of SIRT7 protected H9C2 cardiomyoblasts and primary cardiomyocytes against hypoxia-induced injury and apoptosis. The silencing of SIRT7 markedly abrogated the protective effect induced by miR-142-5p suppression. Taken together, these results suggest that downregulation of miR-142-5p alleviates hypoxia-induced injury through upregulation of SIRT7. Our study suggests miR-142-5p/SIRT7 as potential therapeutic targets for ischemic heart disease.
Collapse
|
50
|
Yan Y, Song X, Li Z, Zhang J, Ren J, Wu J, Li Y, Guan Y, Wang J. Elevated levels of granzyme B correlated with miR-874-3p downregulation in patients with acute myocardial infarction. Biomark Med 2017; 11:761-767. [PMID: 28699362 DOI: 10.2217/bmm-2017-0144] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
AIM Granzyme B could induce apoptosis of target cell in acute myocardial infarction (AMI) and was identified as the target of miR-874-3p by searching the miRNA database. We aim to determine the levels of granzyme B and miR-874-3p as well as assay their correlations or predict powers in AMI. PATIENTS & METHODS We measured levels of plasma granzyme B and miR-874-3p in 80 AMI patients or 40 healthy controls and assayed their correlations or predicted powers for AMI. RESULTS Elevated levels of granzyme B (16.71 ± 7.23 ng/l vs 9.27 ± 3.90 ng/l) correlated with miR-874-3p downregulation (0.20- ± 0.17-fold vs 1.00- ± 0.79-fold) in AMI patients. CONCLUSION Plasma miR-874-3p might target granzyme B and it might be an additional biomarker for AMI.
Collapse
Affiliation(s)
- Youyou Yan
- Department of Cardiology, Second Hospital of Jilin University, No. 218 Ziqiang Street, Changchun 130041, China
| | - Xinjing Song
- Department of Cardiology, Second Hospital of Jilin University, No. 218 Ziqiang Street, Changchun 130041, China
| | - Zhibo Li
- Department of Cardiology, Second Hospital of Jilin University, No. 218 Ziqiang Street, Changchun 130041, China
| | - Jinpu Zhang
- Department of Cardiology, Second Hospital of Jilin University, No. 218 Ziqiang Street, Changchun 130041, China
| | - Jiajun Ren
- Department of Cardiology, Second Hospital of Jilin University, No. 218 Ziqiang Street, Changchun 130041, China
| | - Junduo Wu
- Department of Cardiology, Second Hospital of Jilin University, No. 218 Ziqiang Street, Changchun 130041, China
| | - Yali Li
- Department of Cardiology, Second Hospital of Jilin University, No. 218 Ziqiang Street, Changchun 130041, China
| | - Yinuo Guan
- Department of Cardiology, Second Hospital of Jilin University, No. 218 Ziqiang Street, Changchun 130041, China
| | - Junnan Wang
- Department of Cardiology, Second Hospital of Jilin University, No. 218 Ziqiang Street, Changchun 130041, China
| |
Collapse
|