101
|
Aranda A. MicroRNAs and thyroid hormone action. Mol Cell Endocrinol 2021; 525:111175. [PMID: 33515639 DOI: 10.1016/j.mce.2021.111175] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/29/2020] [Accepted: 01/21/2021] [Indexed: 12/12/2022]
Abstract
MicroRNAs (miRNAs) are small noncoding RNAs that post-transcriptionally repress gene expression by binding generally to the 3'-untranslated regions of their target mRNAs. miRNAs regulate a large fraction of the genome, playing a key role in most physiological and pathological processes. The thyroid hormones (T4 and T3) are major regulators of development, metabolism and cell growth. The thyroid hormones (THs) are synthetized in the thyroid gland and enter the cells through transporter proteins. In the cells, T4 and T3 are metabolized by deiodinase enzymes and bind to nuclear receptors (TRs), which have a higher affinity by T3. TRs act as hormone dependent transcription factors by binding to thyroid hormone response elements (TREs) in the target genes and recruiting transcriptional coregulators. There is increasing evidence that a variety of miRNAs target deiodinases and the receptor, thus regulating TH signaling is different tissues. In turn, the THs have been shown to modulate the expression of specific miRNAs and their mRNA targets in different cell types and organs. In many cases, the existence of TREs in the regulatory regions of these miRNAs has been identified, and the hormone bound receptors transcriptionally regulate expression of these molecules. Changes in the levels of miRNAs have been demonstrated to mediate some of the important actions of the THs in processes such as muscle and heart function, lipid liver metabolism or skin physiology. In addition, miRNA regulation is involved in the effects of TRs on cell proliferation and cancer.
Collapse
Affiliation(s)
- Ana Aranda
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.
| |
Collapse
|
102
|
Moriscot A, Miyabara EH, Langeani B, Belli A, Egginton S, Bowen TS. Firearms-related skeletal muscle trauma: pathophysiology and novel approaches for regeneration. NPJ Regen Med 2021; 6:17. [PMID: 33772028 PMCID: PMC7997931 DOI: 10.1038/s41536-021-00127-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 02/24/2021] [Indexed: 02/07/2023] Open
Abstract
One major cause of traumatic injury is firearm-related wounds (i.e., ballistic trauma), common in both civilian and military populations, which is increasing in prevalence and has serious long-term health and socioeconomic consequences worldwide. Common primary injuries of ballistic trauma include soft-tissue damage and loss, haemorrhage, bone fracture, and pain. The majority of injuries are of musculoskeletal origin and located in the extremities, such that skeletal muscle offers a major therapeutic target to aid recovery and return to normal daily activities. However, the underlying pathophysiology of skeletal muscle ballistic trauma remains poorly understood, with limited evidence-based treatment options. As such, this review will address the topic of firearm-related skeletal muscle injury and regeneration. We first introduce trauma ballistics and the immediate injury of skeletal muscle, followed by detailed coverage of the underlying biological mechanisms involved in regulating skeletal muscle dysfunction following injury, with a specific focus on the processes of muscle regeneration, muscle wasting and vascular impairments. Finally, we evaluate novel approaches for minimising muscle damage and enhancing muscle regeneration after ballistic trauma, which may have important relevance for primary care in victims of violence.
Collapse
Affiliation(s)
- Anselmo Moriscot
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Elen H Miyabara
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Antonio Belli
- NIHR Surgical Reconstruction and Microbiology Research Centre, University of Birmingham, Birmingham, UK
| | - Stuart Egginton
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - T Scott Bowen
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK.
| |
Collapse
|
103
|
Wang W, Zheng H. Myocardial Infarction: The Protective Role of MiRNAs in Myocardium Pathology. Front Cardiovasc Med 2021; 8:631817. [PMID: 33748196 PMCID: PMC7973051 DOI: 10.3389/fcvm.2021.631817] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 02/08/2021] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases have been regarded as the leading cause of death around the world, with myocardial infarction (MI) being the most severe form. MI leads to myocardial apoptosis, cardiomyocyte fibrosis, and cardiomyocyte hypertrophy, ultimately leading to heart failure, and death. Micro RNAs (miRNAs) participate in the genesis and progression of myocardial pathology after MI by playing an important regulatory role. This review aims to summarize all available knowledge on the role of miRNAs in the myocardial pathological process after MI to uncover potential major target pathways. In addition, the main therapeutic methods and their latest progress are also reviewed. miRNAs can regulate the main signaling pathways as well as pathological processes. Thus, they have the potential to induce therapeutic effects. Hence, the combination of miRNAs with recently developed exosome nanocomplexes may represent the future direction of therapeutics.
Collapse
Affiliation(s)
- Wei Wang
- Graduate School of Bengbu Medical College, Bengbu, China
| | - Hao Zheng
- Department of Cardiovascular Medicine, Zhejiang Provincial People's Hospital, Hangzhou, China
| |
Collapse
|
104
|
Role of Selected miRNAs as Diagnostic and Prognostic Biomarkers in Cardiovascular Diseases, Including Coronary Artery Disease, Myocardial Infarction and Atherosclerosis. J Cardiovasc Dev Dis 2021; 8:jcdd8020022. [PMID: 33669699 PMCID: PMC7923109 DOI: 10.3390/jcdd8020022] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 02/13/2021] [Accepted: 02/15/2021] [Indexed: 02/08/2023] Open
Abstract
Cardiovascular diseases are the leading cause of death worldwide in different cohorts. It is well known that miRNAs have a crucial role in regulating the development of cardiovascular physiology, thus impacting the pathophysiology of heart diseases. MiRNAs also have been reported to be associated with cardiac reactions, leading to myocardial infarction (MCI) and ultimately heart failure (HF). To prevent these heart diseases, proper and timely diagnosis of cardiac dysfunction is pivotal. Though there are many symptoms associated with an irregular heart condition and though there are some biomarkers available that may indicate heart disease, authentic, specific and sensitive markers are the need of the hour. In recent times, miRNAs have proven to be promising candidates in this regard. They are potent biomarkers as they can be easily detected in body fluids (blood, urine, etc.) due to their remarkable stability and presence in apoptotic bodies and exosomes. Existing studies suggest the role of miRNAs as valuable biomarkers. A single biomarker may be insufficient to diagnose coronary artery disease (CAD) or acute myocardial infarction (AMI); thus, a combination of different miRNAs may prove fruitful. Therefore, this review aims to highlight the role of circulating miRNA as diagnostic and prognostic biomarkers in cardiovascular diseases such as coronary artery disease (CAD), myocardial infarction (MI) and atherosclerosis.
Collapse
|
105
|
Chua SK, Wang BW, Yu YJ, Fang WJ, Lin CM, Shyu KG. Cyclic stretching boosts microRNA-499 to regulate Bcl-2 via microRNA-208a in atrial fibroblasts. J Cell Mol Med 2021; 25:3113-3123. [PMID: 33605072 PMCID: PMC7957261 DOI: 10.1111/jcmm.16373] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 01/25/2021] [Accepted: 02/01/2021] [Indexed: 01/20/2023] Open
Abstract
MicroRNAs that modulate transcription can regulate other microRNAs and are also up-regulated under pathological stress. MicroRNA-499 (miR-499), microRNA-208a (miR-208a) and B-cell lymphoma 2 (Bcl-2) play roles in cardiovascular diseases, such as direct reprogramming of cardiac fibroblast into cardiomyocyte and cardiomyocyte apoptosis. Whether miR208a, miR499 and Bcl-2 were critical regulators in cardiac fibroblast apoptosis under mechanical stretching conditions in human cardiac fibroblasts-adult atrial (HCF-aa) was investigated. Using negative pressure, HCF-aa grown on a flexible membrane base were cyclically stretched to 20% of their maximum elongation. In adult rats, an aortocaval shunt was used to create an in vivo model of volume overload. MiR208a was up-regulated early by stretching and returned to normal levels with longer stretching cycles, whereas the expression of miR499 and Bcl-2 was up-regulated by longer stretching times. Pre-treatment with antagomir-499 reversed the miR-208a down-regulation, whereas Bcl-2 expression could be suppressed by miR-208a overexpression. In the HCF-aa under stretching for 1 h, miR-499 overexpression decreased pri-miR-208a luciferase activity; this inhibition of pri-miR-208a luciferase activity with stretching was reversed when the miR-499-5p binding site in pri-miR-208a was mutated. The addition of antagomir-208a reversed the Bcl-2-3'UTR suppression from stretching for 1 h. Flow cytometric analysis revealed that pre-treatment with miR-499 or antagomir-208a inhibited cellular apoptosis in stretched HCF-aa. In hearts with volume overload, miR-499 overexpression inhibited myocardial miR-208a expression, whereas Bcl-2 expression could be suppressed by the addition of miR-208a. In conclusion, miR-208a mediated the regulation of miR-499 on Bcl-2 expression in stretched HCF-aa and hearts with volume overload.
Collapse
Affiliation(s)
- Su-Kiat Chua
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei, Taiwan.,Division of Cardiology, Department of Internal Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Bao-Wei Wang
- Division of Cardiology, Department of Internal Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Ying-Ju Yu
- Division of Cardiology, Department of Internal Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Wei-Jen Fang
- Division of Cardiology, Department of Internal Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Chiu-Mei Lin
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei, Taiwan.,Department of Emergency Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Kou-Gi Shyu
- Division of Cardiology, Department of Internal Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| |
Collapse
|
106
|
Li H, Xu JD, Fang XH, Zhu JN, Yang J, Pan R, Yuan SJ, Zeng N, Yang ZZ, Yang H, Wang XP, Duan JZ, Wang S, Luo JF, Wu SL, Shan ZX. Circular RNA circRNA_000203 aggravates cardiac hypertrophy via suppressing miR-26b-5p and miR-140-3p binding to Gata4. Cardiovasc Res 2021; 116:1323-1334. [PMID: 31397837 PMCID: PMC7243276 DOI: 10.1093/cvr/cvz215] [Citation(s) in RCA: 185] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 07/01/2019] [Accepted: 08/08/2019] [Indexed: 01/05/2023] Open
Abstract
Aims Circular RNAs (circRNAs) are involved in gene regulation in a variety of physiological and pathological processes. The present study aimed to investigate the effect of circRNA_000203 on cardiac hypertrophy and the potential mechanisms involved. Methods and results CircRNA_000203 was found to be up-regulated in the myocardium of Ang-II-infused mice and in the cytoplasma of Ang-II-treated neonatal mouse ventricular cardiomyocytes (NMVCs). Enforced expression of circRNA_000203 enhances cell size and expression of atrial natriuretic peptide and β-myosin heavy chain in NMVCs. In vivo, heart function was impaired and cardiac hypertrophy was aggravated in Ang-II-infused myocardium-specific circRNA_000203 transgenic mice (Tg-circ203). Mechanistically, we found that circRNA_000203 could specifically sponge miR-26b-5p, -140-3p in NMVCs. Further, dual-luciferase reporter assay showed that miR-26b-5p, -140-3p could interact with 3′-UTRs of Gata4 gene, and circRNA_000203 could block the above interactions. In addition, Gata4 expression is transcriptionally inhibited by miR-26b-5p, -140-3p mimic in NMVCs but enhanced by over-expression of circRNA_000203 in vitro and in vivo. Functionally, miR-26b-5p, -140-3p, and Gata4 siRNA, could reverse the hypertrophic growth in Ang-II-induced NMVCs, as well as eliminate the pro-hypertrophic effect of circRNA_000203 in NMVCs. Furthermore, we demonstrated that NF-κB signalling mediates the up-regulation of circRNA_000203 in NMVCs exposed to Ang-II treatment. Conclusions Our data demonstrated that circRNA_000203 exacerbates cardiac hypertrophy via suppressing miR-26b-5p and miR-140-3p leading to enhanced Gata4 levels.
Collapse
Affiliation(s)
- Hui Li
- School of Pharmacy, Southern Medical University, Guangzhou 510515, China.,Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Cardiovascular Institute, Guangzhou 510080, China.,Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Jin-Dong Xu
- Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China.,Department of Anesthesiology, Guangdong Cardiovascular Institute, Guangzhou 510080, China
| | - Xian-Hong Fang
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Cardiovascular Institute, Guangzhou 510080, China.,Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Jie-Ning Zhu
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Cardiovascular Institute, Guangzhou 510080, China
| | - Jing Yang
- School of Medicine, South China University of Technology, Guangzhou 510632, China
| | - Rong Pan
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510632, China
| | - Shu-Jing Yuan
- School of Medicine, South China University of Technology, Guangzhou 510632, China
| | - Ni Zeng
- School of Medicine, South China University of Technology, Guangzhou 510632, China
| | - Zhen-Zhen Yang
- School of Medicine, South China University of Technology, Guangzhou 510632, China
| | - Hui Yang
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Cardiovascular Institute, Guangzhou 510080, China.,Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Xi-Pei Wang
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Cardiovascular Institute, Guangzhou 510080, China.,Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Jin-Zhu Duan
- Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Sheng Wang
- Department of Anesthesiology, Guangdong Cardiovascular Institute, Guangzhou 510080, China
| | - Jian-Fang Luo
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Cardiovascular Institute, Guangzhou 510080, China
| | - Shu-Lin Wu
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Cardiovascular Institute, Guangzhou 510080, China.,Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Zhi-Xin Shan
- School of Pharmacy, Southern Medical University, Guangzhou 510515, China.,Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Cardiovascular Institute, Guangzhou 510080, China.,Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| |
Collapse
|
107
|
Saadat S, Noureddini M, Mahjoubin-Tehran M, Nazemi S, Shojaie L, Aschner M, Maleki B, Abbasi-Kolli M, Rajabi Moghadam H, Alani B, Mirzaei H. Pivotal Role of TGF-β/Smad Signaling in Cardiac Fibrosis: Non-coding RNAs as Effectual Players. Front Cardiovasc Med 2021; 7:588347. [PMID: 33569393 PMCID: PMC7868343 DOI: 10.3389/fcvm.2020.588347] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/15/2020] [Indexed: 12/21/2022] Open
Abstract
Unintended cardiac fibroblast proliferation in many pathophysiological heart conditions, known as cardiac fibrosis, results in pooling of extracellular matrix (ECM) proteins in the heart muscle. Transforming growth factor β (TGF-β) as a pivotal cytokine/growth factor stimulates fibroblasts and hastens ECM production in injured tissues. The TGF-β receptor is a heterodimeric receptor complex on the plasma membrane, made up from TGF-β type I, as well as type II receptors, giving rise to Smad2 and Smad3 transcription factors phosphorylation upon canonical signaling. Phosphorylated Smad2, Smad3, and cytoplasmic Smad4 intercommunicate to transfer the signal to the nucleus, culminating in provoked gene transcription. Additionally, TGF-β receptor complex activation starts up non-canonical signaling that lead to the mitogen-stimulated protein kinase cascade activation, inducing p38, JNK1/2 (c-Jun NH2-terminal kinase 1/2), and ERK1/2 (extracellular signal–regulated kinase 1/2) signaling. TGF-β not only activates fibroblasts and stimulates them to differentiate into myofibroblasts, which produce ECM proteins, but also promotes fibroblast proliferation. Non-coding RNAs (ncRNAs) are important regulators of numerous pathways along with cellular procedures. MicroRNAs and circular long ncRNAs, combined with long ncRNAs, are capable of affecting TGF-β/Smad signaling, leading to cardiac fibrosis. More comprehensive knowledge based on these processes may bring about new diagnostic and therapeutic approaches for different cardiac disorders.
Collapse
Affiliation(s)
- Somayeh Saadat
- Physiology Research Centre, Kashan University of Medical Sciences, Kashan, Iran
| | - Mahdi Noureddini
- Physiology Research Centre, Kashan University of Medical Sciences, Kashan, Iran
| | - Maryam Mahjoubin-Tehran
- Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sina Nazemi
- Vascular and Thorax Surgery Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Layla Shojaie
- Department of Medicine, Research Center for Liver Diseases, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Behnaz Maleki
- Physiology Research Centre, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Abbasi-Kolli
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hasan Rajabi Moghadam
- Department of Cardiology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Behrang Alani
- Department of Applied Cell Sciences, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
108
|
Zhou W, Cai H, Li J, Xu H, Wang X, Men H, Zheng Y, Cai L. Potential roles of mediator Complex Subunit 13 in Cardiac Diseases. Int J Biol Sci 2021; 17:328-338. [PMID: 33390853 PMCID: PMC7757031 DOI: 10.7150/ijbs.52290] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 11/25/2020] [Indexed: 12/16/2022] Open
Abstract
Mediator complex subunit 13 (MED13, previously known as THRAP1 and TRAP240) is a subunit of the cyclin-dependent kinase 8 (CDK8) kinase module in the eukaryotic mediator complex. MED13 has been known to play critical roles in cell cycle, development, and growth. The purpose of this review is to comprehensively discuss its newly identified potential roles in myocardial energy metabolism and non-metabolic cardiovascular diseases. Evidence indicates that cardiac MED13 mainly participates in the regulation of nuclear receptor signaling, which drives the transcription of genes involved in modulating cardiac and systemic energy homeostasis. MED13 is also associated with several pathological conditions, such as metabolic syndrome and thyroid disease-associated heart failure. Therefore, MED13 constitutes a potential therapeutic target for the regulation of metabolic disorders and other cardiovascular diseases.
Collapse
Affiliation(s)
- Wenqian Zhou
- The Center of Cardiovascular Diseases, the First Hospital of Jilin University, Changchun 130021, China
- Pediatric Research Institute, the Department of Pediatrics of University of Louisville, Louisville, KY 40202, USA
| | - He Cai
- The Center of Cardiovascular Diseases, the First Hospital of Jilin University, Changchun 130021, China
| | - Jia Li
- Pediatric Research Institute, the Department of Pediatrics of University of Louisville, Louisville, KY 40202, USA
- Department of Nephrology, the First Hospital of Jilin University, Changchun 130021, China
| | - He Xu
- Department of Respiratory Medicine, the First Hospital of Jilin University (Eastern Division), Changchun 130031, China
| | - Xiang Wang
- The Center of Cardiovascular Diseases, the First Hospital of Jilin University, Changchun 130021, China
- Pediatric Research Institute, the Department of Pediatrics of University of Louisville, Louisville, KY 40202, USA
| | - Hongbo Men
- The Center of Cardiovascular Diseases, the First Hospital of Jilin University, Changchun 130021, China
- Pediatric Research Institute, the Department of Pediatrics of University of Louisville, Louisville, KY 40202, USA
| | - Yang Zheng
- The Center of Cardiovascular Diseases, the First Hospital of Jilin University, Changchun 130021, China
| | - Lu Cai
- Pediatric Research Institute, the Department of Pediatrics of University of Louisville, Louisville, KY 40202, USA
- Department of Pharmacology and Toxicology, the University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|
109
|
Gong L, Jiang H, Qiu G, Sun K. miR-208a Promotes Apoptosis in H9c2 Cardiomyocytes by Targeting GATA4. CONGENIT HEART DIS 2021. [DOI: 10.32604/chd.2021.015831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/05/2023]
|
110
|
Affiliation(s)
- Ajit Magadum
- Cardiovascular Research Center (A.M., A.A.K., E.C., Y.S., L.Z.), Icahn School of Medicine at Mount Sinai, New York.,Department of Genetics and Genomic Sciences (A.M., A.A.K., E.C., L.Z.), Icahn School of Medicine at Mount Sinai, New York.,Black Family Stem Cell Institute (A.M., A.A.K., E.C., L.Z.), Icahn School of Medicine at Mount Sinai, New York
| | - Ann Anu Kurian
- Cardiovascular Research Center (A.M., A.A.K., E.C., Y.S., L.Z.), Icahn School of Medicine at Mount Sinai, New York.,Department of Genetics and Genomic Sciences (A.M., A.A.K., E.C., L.Z.), Icahn School of Medicine at Mount Sinai, New York.,Black Family Stem Cell Institute (A.M., A.A.K., E.C., L.Z.), Icahn School of Medicine at Mount Sinai, New York
| | - Elena Chepurko
- Cardiovascular Research Center (A.M., A.A.K., E.C., Y.S., L.Z.), Icahn School of Medicine at Mount Sinai, New York.,Department of Genetics and Genomic Sciences (A.M., A.A.K., E.C., L.Z.), Icahn School of Medicine at Mount Sinai, New York.,Black Family Stem Cell Institute (A.M., A.A.K., E.C., L.Z.), Icahn School of Medicine at Mount Sinai, New York
| | - Yassine Sassi
- Cardiovascular Research Center (A.M., A.A.K., E.C., Y.S., L.Z.), Icahn School of Medicine at Mount Sinai, New York
| | - Roger J Hajjar
- Phospholamban Foundation, Amsterdam, The Netherlands (R.J.H.)
| | - Lior Zangi
- Cardiovascular Research Center (A.M., A.A.K., E.C., Y.S., L.Z.), Icahn School of Medicine at Mount Sinai, New York.,Department of Genetics and Genomic Sciences (A.M., A.A.K., E.C., L.Z.), Icahn School of Medicine at Mount Sinai, New York.,Black Family Stem Cell Institute (A.M., A.A.K., E.C., L.Z.), Icahn School of Medicine at Mount Sinai, New York
| |
Collapse
|
111
|
Tong H, Snow SJ, Chen H, Schladweiler MC, Carswell G, Chorley B, Kodavanti UP. Fish oil and olive oil-enriched diets alleviate acute ozone-induced cardiovascular effects in rats. Toxicol Appl Pharmacol 2020; 409:115296. [PMID: 33091443 DOI: 10.1016/j.taap.2020.115296] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 10/13/2020] [Accepted: 10/16/2020] [Indexed: 12/13/2022]
Abstract
Fish oil (FO) and olive oil (OO) supplementations attenuate the cardiovascular responses to inhaled concentrated ambient particles in human volunteers. This study was designed to examine the cardiovascular effects of ozone (O3) exposure and the efficacy of FO and OO-enriched diets in attenuating the cardiovascular effects from O3 exposure in rats. Rats were fed either a normal diet (ND), a diet enriched with 6% FO or OO starting at 4 weeks of age. Eight weeks following the start of these diet, animals were exposed to filtered air (FA) or 0.8 ppm O3, 4 h/day for 2 consecutive days. Immediately after exposure, cardiac function was measured as the indices of left-ventricular developed pressure (LVDP) and contractility (dP/dtmax and dP/dtmin) before ischemia. In addition, selective microRNAs (miRNAs) of inflammation, endothelial function, and cardiac function were assessed in cardiac tissues to examine the molecular alterations of diets and O3 exposure. Pre-ischemic LVDP and dP/dtmax were lower after O3 exposure in rats fed ND but not FO and OO. Cardiac miRNAs expressions were altered by both diet and O3 exposure. Specifically, O3-induced up-regulation of miR-150-5p and miR-208a-5p were attenuated by FO and/or OO. miR-21 was up-regulated by both FO and OO after O3 exposure. This study demonstrated that O3-induced cardiovascular responses appear to be blunted by FO and OO diets. O3-induced alterations in miRNAs linked to inflammation, cardiac function, and endothelial dysfunction support these pathways are involved, and dietary supplementation with FO or OO may alleviate these adverse cardiovascular effects in rats.
Collapse
Affiliation(s)
- Haiyan Tong
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, Office of Research and Development, U.S. Environmental Protection Agency, Chapel Hill, NC 27514, United States.
| | - Samantha J Snow
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, United States
| | - Hao Chen
- Oak Ridge Institute of Science and Education, Oak Ridge, TN 37830, United States.
| | - Mette C Schladweiler
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, United States.
| | - Gleta Carswell
- Biomolecular and Computational Toxicology Division, Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, United States.
| | - Brian Chorley
- Biomolecular and Computational Toxicology Division, Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, United States.
| | - Urmila P Kodavanti
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, United States.
| |
Collapse
|
112
|
Xie WQ, Men C, He M, Li YS, Lv S. The Effect of MicroRNA-Mediated Exercise on Delaying Sarcopenia in Elderly Individuals. Dose Response 2020; 18:1559325820974543. [PMID: 33293908 PMCID: PMC7705785 DOI: 10.1177/1559325820974543] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/30/2020] [Accepted: 10/27/2020] [Indexed: 11/24/2022] Open
Abstract
Sarcopenia is often regarded as an early sign of weakness and is the core element
of muscle weakness in elderly individuals. Sarcopenia is closely related to the
reduction of exercise, and elderly individuals often suffer from decreased
muscle mass and function due to a lack of exercise. At present, studies have
confirmed that resistance and aerobic exercise are related to muscle mass,
strength and fiber type and to the activation and proliferation of muscle stem
cells (MuSCs). Increasing evidence shows that microRNAs (miRNAs) play an
important role in exercise-related changes in the quantity, composition and
function of skeletal muscle. At the cellular level, miRNAs have been shown to
regulate the proliferation and differentiation of muscle cells. In addition,
miRNAs are related to the composition and transformation of muscle fibers and
involved in the transition of MuSCs from the resting state to the activated
state. Therefore, exercise may delay sarcopenia in elderly individuals by
regulating miRNAs in skeletal muscle. In future miRNA-focused treatment
strategies, these studies will provide valuable information for the formulation
of exercise methods and will provide useful and targeted exercise programs for
elderly individuals with sarcopenia.
Collapse
Affiliation(s)
- Wen-Qing Xie
- Deparment of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Chen Men
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Miao He
- Deparment of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yu-Sheng Li
- Deparment of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shan Lv
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
113
|
Bhattacharyya S, Munshi NV. Development of the Cardiac Conduction System. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a037408. [PMID: 31988140 DOI: 10.1101/cshperspect.a037408] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The cardiac conduction system initiates and propagates each heartbeat. Specialized conducting cells are a well-conserved phenomenon across vertebrate evolution, although mammalian and avian species harbor specific components unique to organisms with four-chamber hearts. Early histological studies in mammals provided evidence for a dominant pacemaker within the right atrium and clarified the existence of the specialized muscular axis responsible for atrioventricular conduction. Building on these seminal observations, contemporary genetic techniques in a multitude of model organisms has characterized the developmental ontogeny, gene regulatory networks, and functional importance of individual anatomical compartments within the cardiac conduction system. This review describes in detail the transcriptional and regulatory networks that act during cardiac conduction system development and homeostasis with a particular emphasis on networks implicated in human electrical variation by large genome-wide association studies. We conclude with a discussion of the clinical implications of these studies and describe some future directions.
Collapse
Affiliation(s)
| | - Nikhil V Munshi
- Department of Internal Medicine, Division of Cardiology.,McDermott Center for Human Growth and Development.,Department of Molecular Biology, UT Southwestern Medical Center, Dallas, Texas 75390, USA.,Hamon Center for Regenerative Science and Medicine, Dallas, Texas 75390, USA
| |
Collapse
|
114
|
Fu Y, Wang Y, Bi K, Yang L, Sun Y, Li B, Liu Z, Zhang F, Li Y, Feng C, Bi Z. MicroRNA-208a-3p promotes osteosarcoma progression via targeting PTEN. Exp Ther Med 2020; 20:255. [PMID: 33178353 PMCID: PMC7651880 DOI: 10.3892/etm.2020.9385] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 06/11/2020] [Indexed: 12/17/2022] Open
Abstract
Osteosarcoma (OS) is a malignant bone tumor with a poor prognosis. Accumulated evidence has suggested that microRNAs (miRNAs/miRs) may function as either oncogenes or tumor suppressors, which are associated with tumorigenesis and the progression of different types of cancer. In the present study, the role of miR-208a-3p in OS was investigated. The expression levels of miR-208a-3p in OS tissues and cell lines were determined via reverse transcription-quantitative PCR (RT-qPCR). MTT and colony formation assays were performed to verify the proliferation rate of OS cells. In addition, the effects of miR-208a-3p on the migration and invasion of OS cells were revealed using wound-healing and Transwell assays, respectively. Furthermore, the association between miR-208a-3p and phosphatase and tensin homolog (PTEN) 3'-untranslated region was determined via luciferase reporter assays, western blot and RT-qPCR analysis. The results indicated that miR-208a-3p was upregulated in OS tissues and cell lines compared with adjacent normal tissues and human osteoblastic cells, respectively. miR-208a-3p overexpression promoted and miR-208a-3p knockdown inhibited OS cells proliferation and metastatic potential. Additionally, PTEN was validated as a direct target of miR-208a-3p and its expression was negatively associate with that of miR-208a-3p in OS cells. Taken together, these results may suggest that miR-208a-3p promoted OS cells proliferation and metastatic potential via targeting PTEN. Therefore, miR-208a-3p may be considered as a diagnostic biomarker for OS.
Collapse
Affiliation(s)
- Yutuo Fu
- Department of Orthopedics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, P.R. China.,Department of Orthopedics, Heilongjiang Provincial Hospital, Harbin, Heilongjiang 150000, P.R. China
| | - Yan Wang
- Department of Orthopedics, Heilongjiang Provincial Hospital, Harbin, Heilongjiang 150000, P.R. China
| | - Ke Bi
- Department of Orthopedics, Heilongjiang Provincial Hospital, Harbin, Heilongjiang 150000, P.R. China
| | - Lei Yang
- Department of Orthopedics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, P.R. China
| | - Yi Sun
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, P.R. China
| | - Boyuan Li
- Department of Orthopedics, Heilongjiang Provincial Hospital, Harbin, Heilongjiang 150000, P.R. China
| | - Zhenzhong Liu
- Department of Orthopedics, Heilongjiang Provincial Hospital, Harbin, Heilongjiang 150000, P.R. China
| | - Fulin Zhang
- Department of Orthopedics, Heilongjiang Provincial Hospital, Harbin, Heilongjiang 150000, P.R. China
| | - Yuan Li
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang 150000, P.R. China
| | - Chao Feng
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang 150000, P.R. China
| | - Zhenggang Bi
- Department of Orthopedics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, P.R. China
| |
Collapse
|
115
|
Sabet Sarvestani F, Azarpira N. microRNAs Alterations of Myocardium and Brain Ischemia-Reperfusion Injury: Insight to Improve Infarction. Immunol Invest 2020; 51:51-72. [DOI: 10.1080/08820139.2020.1808672] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
116
|
Ravelli F, Masè M. MicroRNAs: New contributors to mechano-electric coupling and atrial fibrillation. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2020; 159:146-156. [PMID: 33011190 DOI: 10.1016/j.pbiomolbio.2020.09.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 09/17/2020] [Accepted: 09/27/2020] [Indexed: 12/29/2022]
Abstract
Atrial fibrillation (AF) is a multifactorial disease, which often occurs in the presence of underlying cardiac abnormalities and is supported by electrophysiological and structural alterations, generally referred to as atrial remodeling. Abnormal substrates are commonly encountered in various conditions that predispose to AF, such as hypertension, heart failure, obesity, and sleep apnea, in which atrial stretch plays a key mechanistic role. Emerging evidence suggests a role for microRNAs (small non-coding RNAs) in the pathogenesis of AF, where they can act as post-transcriptional regulators of the genes involved in atrial remodeling. This review summarizes the experimental and clinical evidence that supports the role of microRNAs in the modulation of atrial electrical and structural remodeling with a focus on overload-induced atrial alterations, and discusses the potential contribution of microRNAs to mechano-electrical coupling and AF.
Collapse
Affiliation(s)
- Flavia Ravelli
- Laboratory of Biophysics and Biosignals, University of Trento, Trento, Italy.
| | - Michela Masè
- Institute of Mountain Emergency Medicine, Eurac Research, Bolzano, Italy; Healthcare Research and Innovation Program, IRCS-HTA, Bruno Kessler Foundation, Trento, Italy
| |
Collapse
|
117
|
MicroRNAs: roles in cardiovascular development and disease. Cardiovasc Pathol 2020; 50:107296. [PMID: 33022373 DOI: 10.1016/j.carpath.2020.107296] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/24/2020] [Accepted: 09/28/2020] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular diseases (CVDs) comprise a group of disorders ranging from peripheral artery, coronary artery, cardiac valve, cardiac muscle, and congenital heart diseases to arrhythmias and ultimately, heart failure. For all the advances in therapeutics, CVDs are still the leading cause of mortality the world over, hence the significance of a thorough understanding of CVDs at the molecular level. Disparities in the expressions of genes and microRNAs (miRNAs) play a crucial role in the determination of the fate of cellular pathways, which ultimately affect an organism's physiology. Indeed, miRNAs serve as the regulators of gene expressions in that they perform key functions both in several important cellular pathways and in the regulation of the onset of various diseases such as CVDs. Many miRNAs are expressed in embryonic, postnatal, and adult hearts; their aberrant expression or genetic deletion is associated with abnormal cardiac cell differentiation, disruption in heart development, and cardiac dysfunction. A substantial body of evidence implicates miRNAs in CVD development and suggests them as diagnostic biomarkers and intriguing therapeutic tools. The present review provides an overview of the history, biogenesis, and processing of miRNAs, as well as their function in the development, remodeling, and diseases of the heart.
Collapse
|
118
|
Kastner N, Zlabinger K, Spannbauer A, Traxler D, Mester-Tonczar J, Hašimbegović E, Gyöngyösi M. New Insights and Current Approaches in Cardiac Hypertrophy Cell Culture, Tissue Engineering Models, and Novel Pathways Involving Non-Coding RNA. Front Pharmacol 2020; 11:1314. [PMID: 32973530 PMCID: PMC7472597 DOI: 10.3389/fphar.2020.01314] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 08/07/2020] [Indexed: 12/23/2022] Open
Abstract
Cardiac hypertrophy is an ongoing clinical challenge, as risk factors such as obesity, smoking and increasing age become more widespread, which lead to an increasing prevalence of developing hypertrophy. Pathological hypertrophy is a maladaptive response to stress conditions, such as pressure overload, and involve a number of changes in cellular mechanisms, gene expression and pathway regulations. Although several important pathways involved in the remodeling and hypertrophy process have been identified, further research is needed to achieve a better understanding and explore new and better treatment options. More recently discovered pathways showed the involvement of several non-coding RNAs, including micro RNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), which either promote or inhibit the remodeling process and pose a possible target for novel therapy approaches. In vitro modeling serves as a vital tool for this further pathway analysis and treatment testing and has vastly improved over the recent years, providing a less costly and labor-intensive alternative to in vivo animal models.
Collapse
Affiliation(s)
- Nina Kastner
- Department of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Katrin Zlabinger
- Department of Cardiology, Medical University of Vienna, Vienna, Austria
| | | | - Denise Traxler
- Department of Cardiology, Medical University of Vienna, Vienna, Austria
| | | | - Ena Hašimbegović
- Department of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Mariann Gyöngyösi
- Department of Cardiology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
119
|
Stølen TO, Høydal MA, Ahmed MS, Jørgensen K, Garten K, Hortigon-Vinagre MP, Zamora V, Scrimgeour NR, Berre AMO, Nes BM, Skogvoll E, Johnsen AB, Moreira JBN, McMullen JR, Attramadal H, Smith GL, Ellingsen Ø, Wisløff U. Exercise training reveals micro-RNAs associated with improved cardiac function and electrophysiology in rats with heart failure after myocardial infarction. J Mol Cell Cardiol 2020; 148:106-119. [PMID: 32918915 DOI: 10.1016/j.yjmcc.2020.08.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 08/20/2020] [Accepted: 08/25/2020] [Indexed: 02/09/2023]
Abstract
AIMS Endurance training improves aerobic fitness and cardiac function in individuals with heart failure. However, the underlying mechanisms are not well characterized. Exercise training could therefore act as a tool to discover novel targets for heart failure treatment. We aimed to associate changes in Ca2+ handling and electrophysiology with micro-RNA (miRNA) profile in exercise trained heart failure rats to establish which miRNAs induce heart failure-like effects in Ca2+ handling and electrophysiology. METHODS AND RESULTS Post-myocardial infarction (MI) heart failure was induced in Sprague Dawley rats. Rats with MI were randomized to sedentary control (sed), moderate (mod)- or high-intensity (high) endurance training for 8 weeks. Exercise training improved cardiac function, Ca2+ handling and electrophysiology including reduced susceptibility to arrhythmia in an exercise intensity-dependent manner where high intensity gave a larger effect. Fifty-five miRNAs were significantly regulated (up or down) in MI-sed, of which 18 and 3 were changed towards Sham-sed in MI-high and MI-mod, respectively. Thereafter we experimentally altered expression of these "exercise-miRNAs" individually in human induced pluripotent stem cell-derived cardiomyocytes (hIPSC-CM) in the same direction as they were changed in MI. Of the "exercise-miRNAs", miR-214-3p prolonged AP duration, whereas miR-140 and miR-208a shortened AP duration. miR-497-5p prolonged Ca2+ release whereas miR-214-3p and miR-31a-5p prolonged Ca2+ decay. CONCLUSION Using exercise training as a tool, we discovered that miR-214-3p, miR-497-5p, miR-31a-5p contribute to heart-failure like behaviour in Ca2+ handling and electrophysiology and could be potential treatment targets.
Collapse
Affiliation(s)
- Tomas O Stølen
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health, Norwegian University of Science and Technology, Trondheim, Norway; Department of Cardiology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway; Department of Cardiothoracic Surgery, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway.
| | - Morten A Høydal
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health, Norwegian University of Science and Technology, Trondheim, Norway; Department of Cardiology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway; Department of Cardiothoracic Surgery, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Muhammad Shakil Ahmed
- Institute for Surgical Research, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Kari Jørgensen
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health, Norwegian University of Science and Technology, Trondheim, Norway
| | - Karin Garten
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health, Norwegian University of Science and Technology, Trondheim, Norway
| | - Maria P Hortigon-Vinagre
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Science, University of Glasgow 126 University Place, Glasgow G12 8TA, United Kingdom
| | - Victor Zamora
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Science, University of Glasgow 126 University Place, Glasgow G12 8TA, United Kingdom
| | - Nathan R Scrimgeour
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health, Norwegian University of Science and Technology, Trondheim, Norway
| | - Anne Marie Ormbostad Berre
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health, Norwegian University of Science and Technology, Trondheim, Norway
| | - Bjarne M Nes
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health, Norwegian University of Science and Technology, Trondheim, Norway; Department of Cardiology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Eirik Skogvoll
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health, Norwegian University of Science and Technology, Trondheim, Norway; Department of Anesthesia and Intensive Care Medicine, St. Olav University Hospital, Trondheim, Norway
| | - Anne Berit Johnsen
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health, Norwegian University of Science and Technology, Trondheim, Norway
| | - Jose B N Moreira
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health, Norwegian University of Science and Technology, Trondheim, Norway
| | - Julie R McMullen
- Cardiac Hypertrophy Laboratory, Baker Heart & Diabetes Institute, 75 Commercial Road, Melbourne, VIC, Australia
| | - Håvard Attramadal
- Institute for Surgical Research, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Godfrey L Smith
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health, Norwegian University of Science and Technology, Trondheim, Norway; Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Science, University of Glasgow 126 University Place, Glasgow G12 8TA, United Kingdom
| | - Øyvind Ellingsen
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health, Norwegian University of Science and Technology, Trondheim, Norway; Department of Cardiology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Ulrik Wisløff
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health, Norwegian University of Science and Technology, Trondheim, Norway; School of Human Movement & Nutrition Sciences, University of Queensland, Australia
| |
Collapse
|
120
|
Myostatin as a Biomarker of Muscle Wasting and other Pathologies-State of the Art and Knowledge Gaps. Nutrients 2020; 12:nu12082401. [PMID: 32796600 PMCID: PMC7469036 DOI: 10.3390/nu12082401] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/06/2020] [Accepted: 08/08/2020] [Indexed: 12/14/2022] Open
Abstract
Sarcopenia is a geriatric syndrome with a significant impact on older patients’ quality of life, morbidity and mortality. Despite the new available criteria, its early diagnosis remains difficult, highlighting the necessity of looking for a valid muscle wasting biomarker. Myostatin, a muscle mass negative regulator, is one of the potential candidates. The aim of this work is to point out various factors affecting the potential of myostatin as a biomarker of muscle wasting. Based on the literature review, we can say that recent studies produced conflicting results and revealed a number of potential confounding factors influencing their use in sarcopenia diagnosing. These factors include physiological variables (such as age, sex and physical activity) as well as a variety of disorders (including heart failure, metabolic syndrome, kidney failure and inflammatory diseases) and differences in laboratory measurement methodology. Our conclusion is that although myostatin alone might not prove to be a feasible biomarker, it could become an important part of a recently proposed panel of muscle wasting biomarkers. However, a thorough understanding of the interrelationship of these markers, as well as establishing a valid measurement methodology for myostatin and revising current research data in the light of new criteria of sarcopenia, is needed.
Collapse
|
121
|
Li DM, Li BX, Yang LJ, Gao P, Ma ZY, Li ZJ. Diagnostic value of circulating microRNA-208a in differentiation of preserved from reduced ejection fraction heart failure. Heart Lung 2020; 50:71-74. [PMID: 32711895 DOI: 10.1016/j.hrtlng.2020.07.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 07/15/2020] [Accepted: 07/15/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND There is no satisfactory answer on the specific biomarker that might be used in differentiating heart failure with reduced EF (HFrEF), allowing for inadequacy of N-terminal prohormone brain natriuretic peptide (NT-proBNP). OBJECTIVES We aim to evaluate the value of microRNA-208a in diagnosing HFrEF patients. METHODS We included 120 HF patients and 60 healthy volunteers. Diagnostic values of NT-proBNP and miR-208a for HF patients versus controls and HFrEF versus HFpEF were described by area under curve (AUC), sensitivity and specificity. RESULTS HFrEF patients had significantly higher miR-208a level (p<0.001). As for diagnosing HFrEF patients, additional use of miR-208a and NT-proBNP yielded a significantly higher AUC than NT-proBNP alone (0.83, 95% CI 0.76-0.90 vs. 0.73, 95% CI 0.64-0.82) and the sensitivity and specificity were raised to 68.0% and 90.2%. CONCLUSION Use of miR-208a in combination with NT-proBNP may allow a more reliable method in diagnosing HFrEF patients.
Collapse
Affiliation(s)
- Dong-Mei Li
- Department of Physical Diagnosis, The Second Affiliated Hospital of Mudanjiang Medical University, Mudanjiang 157000, China
| | - Bin-Xin Li
- Operating Room, Hongqi Hospital, Mudanjiang Medical University, Mudanjiang 157000, China
| | - Li-Jie Yang
- Outpatient of General Internal Medicine, Hongqi Hospital, Mudanjiang Medical University, Mudanjiang 157000, China
| | - Peng Gao
- Emergency Department, Hongqi Hospital, Mudanjiang Medical University, Mudanjiang 157000, China
| | - Zhong-Yu Ma
- Department of Physical Diagnosis, The Second Affiliated Hospital of Mudanjiang Medical University, Mudanjiang 157000, China
| | - Zhong-Juan Li
- Emergency Department, Hongqi Hospital, Mudanjiang Medical University, Mudanjiang 157000, China.
| |
Collapse
|
122
|
Wang K, Ojamaa K, Samuels A, Gilani N, Zhang K, An S, Zhang Y, Tang YD, Askari B, Gerdes AM. BNP as a New Biomarker of Cardiac Thyroid Hormone Function. Front Physiol 2020; 11:729. [PMID: 32733267 PMCID: PMC7363952 DOI: 10.3389/fphys.2020.00729] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 06/04/2020] [Indexed: 12/16/2022] Open
Abstract
Background Cardiac re-expression of fetal genes in patients with heart failure (HF) suggests the presence of low cardiac tissue thyroid hormone (TH) function. However, serum concentrations of T3 and T4 are often normal or subclinically low, necessitating an alternative serum biomarker for low cardiac TH function to guide treatment of these patients. The clinical literature suggests that serum Brain Natriuretic Peptide (BNP) levels are inversely associated with serum triiodo-L-thyronine (T3) levels. The objective of this study was to investigate BNP as a potential serum biomarker for TH function in the heart. Methods Two animal models of thyroid hormone deficiency: (1) 8-weeks of propyl thiouracil-induced hypothyroidism (Hypo) in adult female rats were subsequently treated with oral T3 (10 μg/kg/d) for 3, 6, or 14 days; (2) HF induced by coronary artery ligation (myocardial infarction, MI) in adult female rats was treated daily with low dose oral T3 (5 μg/kg/d) for 8 or 16 wks. Results Six days of T3 treatment of Hypo rats normalized most cardiac functional parameters. Serum levels of BNP increased 5-fold in Hypo rats, while T3 treatment normalized BNP by day 14, showing a significant inverse relationship between serum BNP and free or total T3 concentrations. Myocardial BNP mRNA was increased 2.5-fold in Hypo rats and its expression was decreased to normal values by 14 days of T3 treatment. Measurements of hemodynamic function showed significant dysfunction in MI rats after 16 weeks, with serum BNP increased by 4.5-fold and serum free and total T3 decreased significantly. Treatment with T3 decreased serum BNP while increasing total T3 indicating an inverse correlation between these two biologic factors (r 2 = 0.676, p < 0.001). Myocardial BNP mRNA was increased 5-fold in MI rats which was significantly decreased by T3 over 8 to 16 week treatment periods. Conclusions Results from the two models of TH dysfunction confirmed an inverse relationship between tissue and serum T3 and BNP, such that the reduction in serum BNP could potentially be utilized to monitor efficacy and dosing of T3 treatment. Thus, serum BNP may serve as a reliable biomarker for cardiac TH function.
Collapse
Affiliation(s)
- Kaihao Wang
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, United States.,Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kaie Ojamaa
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, United States
| | - Abigail Samuels
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, United States
| | - Nimra Gilani
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, United States
| | - Kuo Zhang
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, United States.,Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shimin An
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, United States.,Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Youhua Zhang
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, United States
| | - Yi-Da Tang
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bardia Askari
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, United States
| | - Anthony Martin Gerdes
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, United States
| |
Collapse
|
123
|
Peters LJF, Biessen EAL, Hohl M, Weber C, van der Vorst EPC, Santovito D. Small Things Matter: Relevance of MicroRNAs in Cardiovascular Disease. Front Physiol 2020; 11:793. [PMID: 32733281 PMCID: PMC7358539 DOI: 10.3389/fphys.2020.00793] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 06/15/2020] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs (miRNAs) are short sequences of non-coding RNA that play an important role in the regulation of gene expression and thereby in many physiological and pathological processes. Furthermore, miRNAs are released in the extracellular space, for example in vesicles, and are detectable in various biological fluids, such as serum, plasma, and urine. Over the last years, it has been shown that miRNAs are crucial in the development of several cardiovascular diseases (CVDs). This review discusses the (patho)physiological implications of miRNAs in CVD, ranging from cardiovascular risk factors (i.e., hypertension, diabetes, dyslipidemia), to atherosclerosis, myocardial infarction, and cardiac remodeling. Moreover, the intriguing possibility of their use as disease-specific diagnostic and prognostic biomarkers for human CVDs will be discussed in detail. Finally, as several approaches have been developed to alter miRNA expression and function (i.e., mimics, antagomirs, and target-site blockers), we will highlight the miRNAs with the most promising therapeutic potential that may represent suitable candidates for therapeutic intervention in future translational studies and ultimately in clinical trials. All in all, this review gives a comprehensive overview of the most relevant miRNAs in CVD and discusses their potential use as biomarkers and even therapeutic targets.
Collapse
Affiliation(s)
- Linsey J. F. Peters
- Institute for Molecular Cardiovascular Research, RWTH Aachen University, Aachen, Germany
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, Maastricht, Netherlands
- Interdisciplinary Center for Clinical Research, RWTH Aachen University, Aachen, Germany
- German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Erik A. L. Biessen
- Institute for Molecular Cardiovascular Research, RWTH Aachen University, Aachen, Germany
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Mathias Hohl
- Klinik für Innere Medizin III, Universität des Saarlandes, Homburg, Germany
| | - Christian Weber
- German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, Maastricht, Netherlands
- Munich Cluster for Systems Neurology, Munich, Germany
| | - Emiel P. C. van der Vorst
- Institute for Molecular Cardiovascular Research, RWTH Aachen University, Aachen, Germany
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, Maastricht, Netherlands
- Interdisciplinary Center for Clinical Research, RWTH Aachen University, Aachen, Germany
- German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Donato Santovito
- German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany
| |
Collapse
|
124
|
Promoting role of circ-Jarid2/miR-129-5p/Celf1 axis in cardiac hypertrophy. Gene Ther 2020; 28:718-728. [PMID: 32632266 DOI: 10.1038/s41434-020-0165-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 05/20/2020] [Accepted: 06/02/2020] [Indexed: 11/09/2022]
Abstract
Cardiac hypertrophy is imposed much pressure on heart and threatening our live. Previous study suggested that dysregulation of Celf1 is largely connecting to neonatal cardiac dysfunction. Hence, we aimed to explore the precise function and probable regulatory mechanism upstream of Celf1in cardiac hypertrophy. Here, Ang-II treatment was implemented to stimulate hypertrophic phenotypes inH9C2 and MCM cells. Immunofluorescence assay was conducted to measure the surface area of cardiomyocytes. And qRT-PCR assay was conducted to investigate gene expression. Moreover, western blot assay was conducted to probe the protein levels. Results uncovered that Celf1 expression was increased dependent on elevated Ang-II concentration, and that inhibited Celf1 could relieve the Ang-II-caused cardiac hypertrophy. Significantly, Celf1was found to be targeted by miR-129-5p but then released via the sponging role of circ-Jarid2. Furthermore, circ-Jarid2 was found to promote cardiac hypertrophy, whereas miR-129-5p played suppressing parts in hypertrophic cardiomyocytes. Moreover, we verified circ-Jarid2 contributed to cardiac hypertrophy via miR-129-5p/Celf1 axis both in vitro and in vivo. In conclusion, circ-Jarid2/miR-129-5p/Celf1 axis aggravates cardiac hypertrophy, which provides new ideas for developing treatment strategies for patients with cardiac hypertrophy.
Collapse
|
125
|
An Overview of Non-coding RNAs and Cardiovascular System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1229:3-45. [PMID: 32285403 DOI: 10.1007/978-981-15-1671-9_1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cardiovascular disease management and timely diagnosis remain a major dilemma. Delineating molecular mechanisms of cardiovascular diseases is opening horizon in the field of molecular medicines and in the development of early diagnostic markers. Non-coding RNAs are the highly functional and vibrant nucleic acids and are known to be involved in the regulation of endothelial cells, vascular and smooth muscles cells, cardiac metabolism, ischemia, inflammation and many processes in cardiovascular system. This chapter is comprehensively focusing on the overview of the non-coding RNAs including their discovery, generation, classification and functional regulation. In addition, overview regarding different non-coding RNAs as long non-coding, siRNAs and miRNAs involvement in the cardiovascular diseases is also addressed. Detailed functional analysis of this vast group of highly regulatory molecules will be promising for shaping future drug discoveries.
Collapse
|
126
|
Sadat-Ebrahimi SR, Aslanabadi N. Role of MicroRNAs in Diagnosis, Prognosis, and Treatment of Acute Heart Failure: Ambassadors from Intracellular Zone. Galen Med J 2020; 9:e1818. [PMID: 34466598 PMCID: PMC8343948 DOI: 10.31661/gmj.v9i0.1818] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 01/24/2020] [Accepted: 02/06/2020] [Indexed: 12/12/2022] Open
Abstract
Acute heart failure (AHF) is one of the burdensome diseases affecting a considerable proportion of the population. Recently, it has been demonstrated that micro-ribonucleic acids (miRNAs) can exert diagnostic, prognostic, and therapeutic roles in a variety of conditions including AHF. These molecules play essential roles in HF-related pathophysiology, particularly, cardiac fibrosis, and hypertrophy. Some miRNAs namely miRNA-423-5p are reported to have both diagnostic and prognostic capabilities. However, some studies suggest that combination of biomarkers is a much better way to achieve the highest accuracy such as the combination of miRNAs and N-terminal pro b-type Natriuretic Peptide (NT pro-BNP). Therefore, this review discusses different views towards various roles of miRNAs in AHF.
Collapse
Affiliation(s)
- Seyyed-Reza Sadat-Ebrahimi
- Cardiovascular Research Center, Madani Heart Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Naser Aslanabadi
- Cardiovascular Research Center, Madani Heart Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Correspondence to: Naser Aslanabadi, Professor of Cardiology, Cardiovascular Research Center, Madani Heart Center, Tabriz University of Medical Sciences, Tabriz, Iran Telephone Number: +989143110844 Email Address:
| |
Collapse
|
127
|
Liu W, Yang D, Chen L, Liu Q, Wang W, Yang Z, Shang A, Quan W, Li D. Plasma Exosomal miRNA-139-3p is a Novel Biomarker of Colorectal Cancer. J Cancer 2020; 11:4899-4906. [PMID: 32626537 PMCID: PMC7330702 DOI: 10.7150/jca.45548] [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: 03/02/2020] [Accepted: 05/29/2020] [Indexed: 12/21/2022] Open
Abstract
Objectives: This study investigated plasma exosomal miRNA-139-3p as a blood-based biomarker for the early diagnosis and metastasis monitoring of colorectal cancer (CRC). Patients and Methods: Exosome-rich fractions were isolated from the plasma of 80 CRC patients, and 23 controls using a kit method. We then used real-time polymerase chain reaction (RT-qPCR) to detect miR-139-3p levels in all subjects to evaluate expression levels and the predictive value of plasma exosomal miR-139-3p in CRC. We also collected clinicopathological data to explore correlations between abnormal miR-139-3p expression and clinicopathological parameters. Results: When compared with healthy controls, exosomal miR-139-3p expression levels in CRC patients were significantly down-regulated. Furthermore, these expression levels were lower in metastatic colorectal cancer (mCRC) and submucosal patients. Receiver operating characteristic (ROC) curve analysis showed that exosomal miR-139-3p levels were differentiated between CRC patients and healthy controls, as well as between non-metastatic and metastatic patients. Conclusion: Our findings show that decreased exosomal miR-139-3p expression levels in CRC patient plasma may act as a novel biomarker for the early diagnosis and metastasis monitoring in CRC.
Collapse
Affiliation(s)
- Wanchao Liu
- Department of Clinical Laboratory, Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, 201999, China
| | - Dianyu Yang
- Department of Clinical Laboratory, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Longmei Chen
- Department of Clinical Laboratory, Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, 201999, China
| | - Qingqing Liu
- Department of Clinical Laboratory, Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, 201999, China
| | - Wenhui Wang
- Department of Clinical Laboratory, Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, 201999, China
| | - Zhenghua Yang
- Department of Clinical Laboratory, Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, 201999, China
| | - Anquan Shang
- Department of Clinical Laboratory, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Wenqiang Quan
- Department of Clinical Laboratory, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Dong Li
- Department of Clinical Laboratory, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| |
Collapse
|
128
|
Kmecova Z, Veteskova J, Lelkova-Zirova K, Bies Pivackova L, Doka G, Malikova E, Paulis L, Krenek P, Klimas J. Disease severity-related alterations of cardiac microRNAs in experimental pulmonary hypertension. J Cell Mol Med 2020; 24:6943-6951. [PMID: 32395887 PMCID: PMC7299706 DOI: 10.1111/jcmm.15352] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/24/2020] [Accepted: 04/01/2020] [Indexed: 12/16/2022] Open
Abstract
Right ventricular (RV) failure is the primary cause of death in pulmonary arterial hypertension (PAH). We hypothesized that heart‐relevant microRNAs, that is myomiRs (miR‐1, miR‐133a, miR‐208, miR‐499) and miR‐214, can have a role in the right ventricle in the development of PAH. To mimic PAH, male Wistar rats were injected with monocrotaline (MCT, 60 mg/kg, s.c.); control group received vehicle. MCT rats were divided into two groups, based on the clinical presentation: MCT group terminated 4 weeks after MCT administration and prematurely terminated group (ptMCT) displaying signs of terminal disease. Myocardial damage genes and candidate microRNAs expressions were determined by RT‐qPCR. Reduced blood oxygen saturation, breathing disturbances, RV enlargement as well as elevated levels of markers of myocardial damage confirmed PH in MCT animals and were more pronounced in ptMCT. MyomiRs (miR‐1/miR‐133a/miR‐208a/miR‐499) were decreased and the expression of miR‐214 was increased only in ptMCT group (P < 0.05). The myomiRs negatively correlated with Fulton index as a measure of RV hypertrophy in MCT group (P < 0.05), whereas miR‐214 showed a positive correlation (P < 0.05). We conclude that the expression of determined microRNAs mirrored the disease severity and targeting their pathways might represent potential future therapeutic approach in PAH.
Collapse
Affiliation(s)
- Zuzana Kmecova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, Bratislava, Slovakia
| | - Jana Veteskova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, Bratislava, Slovakia
| | - Katarina Lelkova-Zirova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, Bratislava, Slovakia
| | - Lenka Bies Pivackova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, Bratislava, Slovakia
| | - Gabriel Doka
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, Bratislava, Slovakia
| | - Eva Malikova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, Bratislava, Slovakia
| | - Ludovit Paulis
- Institute of Pathophysiology, Faculty of Medicine, Comenius University, Bratislava, Slovakia.,Institute of Normal and Pathological Physiology, Centre of Experimental Medicine, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Peter Krenek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, Bratislava, Slovakia
| | - Jan Klimas
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, Bratislava, Slovakia
| |
Collapse
|
129
|
Larupa Santos J, Rodríguez I, S. Olesen M, Hjorth Bentzen B, Schmitt N. Investigating gene-microRNA networks in atrial fibrillation patients with mitral valve regurgitation. PLoS One 2020; 15:e0232719. [PMID: 32392228 PMCID: PMC7213724 DOI: 10.1371/journal.pone.0232719] [Citation(s) in RCA: 12] [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: 12/17/2019] [Accepted: 04/20/2020] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Atrial fibrillation (AF) is predicted to affect around 17.9 million individuals in Europe by 2060. The disease is associated with severe electrical and structural remodelling of the heart, and increased the risk of stroke and heart failure. In order to improve treatment and find new drug targets, the field needs to better comprehend the exact molecular mechanisms in these remodelling processes. OBJECTIVES This study aims to identify gene and miRNA networks involved in the remodelling of AF hearts in AF patients with mitral valve regurgitation (MVR). METHODS Total RNA was extracted from right atrial biopsies from patients undergoing surgery for mitral valve replacement or repair with AF and without history of AF to test for differentially expressed genes and miRNAs using RNA-sequencing and miRNA microarray. In silico predictions were used to construct a mRNA-miRNA network including differentially expressed mRNAs and miRNAs. Gene and chromosome enrichment analysis were used to identify molecular pathways and high-density AF loci. RESULTS We found 644 genes and 43 miRNAs differentially expressed in AF patients compared to controls. From these lists, we identified 905 pairs of putative miRNA-mRNA interactions, including 37 miRNAs and 295 genes. Of particular note, AF-associated miR-130b-3p, miR-338-5p and miR-208a-3p were differentially expressed in our AF tissue samples. These miRNAs are predicted regulators of several differentially expressed genes associated with cardiac conduction and fibrosis. We identified two high-density AF loci in chromosomes 14q11.2 and 6p21.3. CONCLUSIONS AF in MVR patients is associated with down-regulation of ion channel genes and up-regulation of extracellular matrix genes. Other AF related genes are dysregulated and several are predicted to be targeted by miRNAs. Our novel miRNA-mRNA regulatory network provides new insights into the mechanisms of AF.
Collapse
Affiliation(s)
- Joana Larupa Santos
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N, Denmark
| | - Ismael Rodríguez
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N, Denmark
| | - Morten S. Olesen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N, Denmark
- Department of Cardiology, Laboratory for Molecular Cardiology, The Heart Centre, Rigshospitalet, University Hospital of Copenhagen, Copenhagen Ø, Denmark
| | - Bo Hjorth Bentzen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N, Denmark
| | - Nicole Schmitt
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N, Denmark
| |
Collapse
|
130
|
Tang R, Long T, Lui KO, Chen Y, Huang ZP. A Roadmap for Fixing the Heart: RNA Regulatory Networks in Cardiac Disease. MOLECULAR THERAPY-NUCLEIC ACIDS 2020; 20:673-686. [PMID: 32380417 PMCID: PMC7210385 DOI: 10.1016/j.omtn.2020.04.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 03/16/2020] [Accepted: 04/21/2020] [Indexed: 02/07/2023]
Abstract
With the continuous development of RNA biology and massive genome-wide transcriptome analysis, more and more RNA molecules and their functions have been explored in the last decade. Increasing evidence has demonstrated that RNA-related regulatory networks play an important role in a variety of human diseases, including cardiovascular diseases. In this review, we focus on RNA regulatory networks in heart disease, most of which are devastating conditions with no known cure. We systemically summarize recent discoveries of important new components of RNA regulatory networks, including microRNAs, long non-coding RNAs, and circular RNAs, as well as multiple regulators that affect the activity of these networks in cardiac physiology and pathology. In addition, this review covers emerging micropeptides, which represent short open reading frames (sORFs) in long non-coding RNA transcripts that may modulate cardiac physiology. Based on the current knowledge of RNA regulatory networks, we think that ongoing discoveries will not only provide us a better understanding of the molecular mechanisms that underlie heart disease, but will also identify novel biomarkers and therapeutic targets for the diagnosis and treatment of cardiac disease.
Collapse
Affiliation(s)
- Rong Tang
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Tianxin Long
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Kathy O Lui
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR 999077, China
| | - Yili Chen
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Zhan-Peng Huang
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
131
|
Cassani M, Fernandes S, Vrbsky J, Ergir E, Cavalieri F, Forte G. Combining Nanomaterials and Developmental Pathways to Design New Treatments for Cardiac Regeneration: The Pulsing Heart of Advanced Therapies. Front Bioeng Biotechnol 2020; 8:323. [PMID: 32391340 PMCID: PMC7193099 DOI: 10.3389/fbioe.2020.00323] [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: 10/21/2019] [Accepted: 03/24/2020] [Indexed: 12/12/2022] Open
Abstract
The research for heart therapies is challenged by the limited intrinsic regenerative capacity of the adult heart. Moreover, it has been hampered by the poor results obtained by tissue engineering and regenerative medicine attempts at generating functional beating constructs able to integrate with the host tissue. For this reason, organ transplantation remains the elective treatment for end-stage heart failure, while novel strategies aiming to promote cardiac regeneration or repair lag behind. The recent discovery that adult cardiomyocytes can be ectopically induced to enter the cell cycle and proliferate by a combination of microRNAs and cardioprotective drugs, like anti-oxidant, anti-inflammatory, anti-coagulants and anti-platelets agents, fueled the quest for new strategies suited to foster cardiac repair. While proposing a revolutionary approach for heart regeneration, these studies raised serious issues regarding the efficient controlled delivery of the therapeutic cargo, as well as its timely removal or metabolic inactivation from the site of action. Especially, there is need for innovative treatment because of evidence of severe side effects caused by pleiotropic drugs. Biocompatible nanoparticles possess unique physico-chemical properties that have been extensively exploited for overcoming the limitations of standard medical therapies. Researchers have put great efforts into the optimization of the nanoparticles synthesis and functionalization, to control their interactions with the biological milieu and use as a viable alternative to traditional approaches. Nanoparticles can be used for diagnosis and deliver therapies in a personalized and targeted fashion. Regarding the treatment of cardiovascular diseases, nanoparticles-based strategies have provided very promising outcomes, in preclinical studies, during the last years. Efficient encapsulation of a large variety of cargos, specific release at the desired site and improvement of cardiac function are some of the main achievements reached so far by nanoparticle-based treatments in animal models. This work offers an overview on the recent nanomedical applications for cardiac regeneration and highlights how the versatility of nanomaterials can be combined with the newest molecular biology discoveries to advance cardiac regeneration therapies.
Collapse
Affiliation(s)
- Marco Cassani
- International Clinical Research Center, St Anne’s University Hospital, Brno, Czechia
| | - Soraia Fernandes
- International Clinical Research Center, St Anne’s University Hospital, Brno, Czechia
| | - Jan Vrbsky
- International Clinical Research Center, St Anne’s University Hospital, Brno, Czechia
| | - Ece Ergir
- International Clinical Research Center, St Anne’s University Hospital, Brno, Czechia
- Faculty of Technical Chemistry, Institute of Applied Synthetic Chemistry and Institute of Chemical Technologies and Analytics, Vienna University of Technology, Vienna, Austria
| | - Francesca Cavalieri
- School of Science, RMIT University, Melbourne, VIC, Australia
- Dipartimento di Scienze e Tecnologie Chimiche, Università di Roma “Tor Vergata”, Via Della Ricerca Scientifica, Rome, Italy
| | - Giancarlo Forte
- International Clinical Research Center, St Anne’s University Hospital, Brno, Czechia
| |
Collapse
|
132
|
Comprehensive Overview of Non-coding RNAs in Cardiac Development. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1229:197-211. [PMID: 32285413 DOI: 10.1007/978-981-15-1671-9_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Cardiac development in the human embryo is characterized by the interactions of several transcription and growth factors leading the heart from a primordial linear tube into a synchronous contractile four-chamber organ. Studies on cardiogenesis showed that cell proliferation, differentiation, fate specification and morphogenesis are spatiotemporally coordinated by cell-cell interactions and intracellular signalling cross-talks. In recent years, research has focused on a class of inter- and intra-cellular modulators called non-coding RNAs (ncRNAs), transcribed from the noncoding portion of the DNA and involved in the proper formation of the heart. In this chapter, we will summarize the current state of the art on the roles of three major forms of ncRNAs [microRNAs (miRNAs), long ncRNAs (lncRNAs) and circular RNAs (circRNAs)] in orchestrating the four sequential phases of cardiac organogenesis.
Collapse
|
133
|
Ghosh A, Shcherbik N. Effects of Oxidative Stress on Protein Translation: Implications for Cardiovascular Diseases. Int J Mol Sci 2020; 21:E2661. [PMID: 32290431 PMCID: PMC7215667 DOI: 10.3390/ijms21082661] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 04/08/2020] [Accepted: 04/09/2020] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular diseases (CVDs) are a group of disorders that affect the heart and blood vessels. Due to their multifactorial nature and wide variation, CVDs are the leading cause of death worldwide. Understanding the molecular alterations leading to the development of heart and vessel pathologies is crucial for successfully treating and preventing CVDs. One of the causative factors of CVD etiology and progression is acute oxidative stress, a toxic condition characterized by elevated intracellular levels of reactive oxygen species (ROS). Left unabated, ROS can damage virtually any cellular component and affect essential biological processes, including protein synthesis. Defective or insufficient protein translation results in production of faulty protein products and disturbances of protein homeostasis, thus promoting pathologies. The relationships between translational dysregulation, ROS, and cardiovascular disorders will be examined in this review.
Collapse
Affiliation(s)
- Arnab Ghosh
- Department for Cell Biology and Neuroscience, School of Osteopathic Medicine, Rowan University, 2 Medical Center Drive, Stratford, NJ 08084, USA
| | - Natalia Shcherbik
- Department for Cell Biology and Neuroscience, School of Osteopathic Medicine, Rowan University, 2 Medical Center Drive, Stratford, NJ 08084, USA
| |
Collapse
|
134
|
Fulgencio-Covián A, Alonso-Barroso E, Guenzel AJ, Rivera-Barahona A, Ugarte M, Pérez B, Barry MA, Pérez-Cerdá C, Richard E, Desviat LR. Pathogenic implications of dysregulated miRNAs in propionic acidemia related cardiomyopathy. Transl Res 2020; 218:43-56. [PMID: 31951825 DOI: 10.1016/j.trsl.2019.12.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 12/03/2019] [Accepted: 12/23/2019] [Indexed: 12/25/2022]
Abstract
Cardiac alterations (hypertrophic/dilated cardiomyopathy, and rhythm alterations) are one of the major causes of mortality and morbidity in propionic acidemia (PA), caused by the deficiency of the mitochondrial enzyme propionyl-CoA carboxylase (PCC), involved in the catabolism of branched-chain amino acids, cholesterol, and odd-chain fatty acids. Impaired mitochondrial oxidative phosphorylation has been documented in heart biopsies of PA patients, as well as in the hypomorphic Pcca-/-(A138T) mouse model, in the latter correlating with increased oxidative damage and elevated expression of cardiac dysfunction biomarkers atrial and brain natriuretic peptides (ANP and BNP) and beta-myosin heavy chain (β-MHC). Here we characterize the cardiac phenotype in the PA mouse model by histological and echocardiography studies and identify a series of upregulated cardiac-enriched microRNAs (miRNAs) in the PA mouse heart, some of them also altered as circulating miRNAs in PA patients' plasma samples. In PA mice hearts, we show alterations in signaling pathways regulated by the identified miRNAs, which could be contributing to cardiac remodeling and dysfunction; notably, an activation of the mammalian target of rapamycin (mTOR) pathway and a decrease in autophagy, which are reverted by rapamycin treatment. In vitro studies in HL-1 cardiomyocytes indicate that propionate, the major toxic metabolite accumulating in the disease, triggers the increase in expression levels of miRNAs, BNP, and β-MHC, concomitant with an increase in reactive oxygen species. Our results highlight miRNAs and signaling alterations in the PCC-deficient heart which may contribute to the development of PA-associated cardiomyopathy and provide a basis to identify new targets for therapeutic intervention.
Collapse
Affiliation(s)
- Alejandro Fulgencio-Covián
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa CSIC-UAM, Universidad Autónoma de Madrid, Madrid, Spain; Centro de Diagnóstico de Enfermedades Moleculares (CEDEM), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain; Instituto de Investigación Sanitaria Hospital La Paz (IdiPaz), ISCIII, Madrid, Spain
| | - Esmeralda Alonso-Barroso
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa CSIC-UAM, Universidad Autónoma de Madrid, Madrid, Spain; Centro de Diagnóstico de Enfermedades Moleculares (CEDEM), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain; Instituto de Investigación Sanitaria Hospital La Paz (IdiPaz), ISCIII, Madrid, Spain
| | | | - Ana Rivera-Barahona
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa CSIC-UAM, Universidad Autónoma de Madrid, Madrid, Spain; Centro de Diagnóstico de Enfermedades Moleculares (CEDEM), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain; Instituto de Investigación Sanitaria Hospital La Paz (IdiPaz), ISCIII, Madrid, Spain
| | - Magdalena Ugarte
- Centro de Diagnóstico de Enfermedades Moleculares (CEDEM), Madrid, Spain
| | - Belén Pérez
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa CSIC-UAM, Universidad Autónoma de Madrid, Madrid, Spain; Centro de Diagnóstico de Enfermedades Moleculares (CEDEM), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain; Instituto de Investigación Sanitaria Hospital La Paz (IdiPaz), ISCIII, Madrid, Spain
| | | | - Celia Pérez-Cerdá
- Centro de Diagnóstico de Enfermedades Moleculares (CEDEM), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain; Instituto de Investigación Sanitaria Hospital La Paz (IdiPaz), ISCIII, Madrid, Spain
| | - Eva Richard
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa CSIC-UAM, Universidad Autónoma de Madrid, Madrid, Spain; Centro de Diagnóstico de Enfermedades Moleculares (CEDEM), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain; Instituto de Investigación Sanitaria Hospital La Paz (IdiPaz), ISCIII, Madrid, Spain
| | - Lourdes R Desviat
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa CSIC-UAM, Universidad Autónoma de Madrid, Madrid, Spain; Centro de Diagnóstico de Enfermedades Moleculares (CEDEM), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain; Instituto de Investigación Sanitaria Hospital La Paz (IdiPaz), ISCIII, Madrid, Spain.
| |
Collapse
|
135
|
Williams JL, Paudyal A, Awad S, Nicholson J, Grzesik D, Botta J, Meimaridou E, Maharaj AV, Stewart M, Tinker A, Cox RD, Metherell LA. Mylk3 null C57BL/6N mice develop cardiomyopathy, whereas Nnt null C57BL/6J mice do not. Life Sci Alliance 2020; 3:3/4/e201900593. [PMID: 32213617 PMCID: PMC7103425 DOI: 10.26508/lsa.201900593] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 03/10/2020] [Accepted: 03/10/2020] [Indexed: 12/30/2022] Open
Abstract
The C57BL/6J and C57BL/6N mice have well-documented phenotypic and genotypic differences, including the infamous nicotinamide nucleotide transhydrogenase (Nnt) null mutation in the C57BL/6J substrain, which has been linked to cardiovascular traits in mice and cardiomyopathy in humans. To assess whether Nnt loss alone causes a cardiovascular phenotype, we investigated the C57BL/6N, C57BL/6J mice and a C57BL/6J-BAC transgenic rescuing NNT expression, at 3, 12, and 18 mo. We identified a modest dilated cardiomyopathy in the C57BL/6N mice, absent in the two B6J substrains. Immunofluorescent staining of cardiomyocytes revealed eccentric hypertrophy in these mice, with defects in sarcomere organisation. RNAseq analysis identified differential expression of a number of cardiac remodelling genes commonly associated with cardiac disease segregating with the phenotype. Variant calling from RNAseq data identified a myosin light chain kinase 3 (Mylk3) mutation in C57BL/6N mice, which abolishes MYLK3 protein expression. These results indicate the C57BL/6J Nnt-null mice do not develop cardiomyopathy; however, we identified a null mutation in Mylk3 as a credible cause of the cardiomyopathy phenotype in the C57BL/6N.
Collapse
Affiliation(s)
- Jack L Williams
- Centre for Endocrinology, William Harvey Research Institute, Charterhouse Square, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Anju Paudyal
- Medical Research Council Harwell Institute, Mary Lyon Centre, Harwell Campus, Oxfordshire, UK
| | - Sherine Awad
- Centre for Endocrinology, William Harvey Research Institute, Charterhouse Square, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - James Nicholson
- Centre for Endocrinology, William Harvey Research Institute, Charterhouse Square, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Dominika Grzesik
- Centre for Endocrinology, William Harvey Research Institute, Charterhouse Square, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Joaquin Botta
- Centre for Endocrinology, William Harvey Research Institute, Charterhouse Square, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Eirini Meimaridou
- School of Human Sciences, London Metropolitan University, London, UK
| | - Avinaash V Maharaj
- Centre for Endocrinology, William Harvey Research Institute, Charterhouse Square, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Michelle Stewart
- Medical Research Council Harwell Institute, Mary Lyon Centre, Harwell Campus, Oxfordshire, UK
| | - Andrew Tinker
- William Harvey Heart Centre, William Harvey Research Institute, Charterhouse Square, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Roger D Cox
- Medical Research Council Harwell Institute, Mammalian Genetics Unit, Harwell Campus, Oxfordshire, UK
| | - Lou A Metherell
- Centre for Endocrinology, William Harvey Research Institute, Charterhouse Square, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
136
|
The Therapeutic Potential of MicroRNAs in Atrial Fibrillation. Mediators Inflamm 2020; 2020:3053520. [PMID: 32256190 PMCID: PMC7091547 DOI: 10.1155/2020/3053520] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 03/03/2020] [Indexed: 12/12/2022] Open
Abstract
One of the most globally prevalent supraventricular arrhythmias is atrial fibrillation (AF). Knowledge of the structures and functions of messenger RNA (mRNA) has recently increased. It is no longer viewed as solely an intermediate molecule between DNA and proteins but has come to be seen as a dynamic and modifiable gene regulator. This new perspective on mRNA has led to rising interest in it and its presence in research into new therapeutic schemes. This paper, therefore, focuses on microRNAs (miRNAs), which are small noncoding RNAs that regulate posttranscriptional gene expression and play a vital role in the physiology and normative development of cardiovascular systems. This means they play an equally vital role in the development and progression of cardiovascular diseases. In recent years, multiple studies have pinpointed particular miRNA expression profiles as being associated with varying histological features of AF. These studies have been carried out in both animal models and AF patients. The emergence of miRNAs as biomarkers and their therapeutic potential in AF patients will be discussed in the body of this paper.
Collapse
|
137
|
Barreto-Chaves MLM, Senger N, Fevereiro MR, Parletta AC, Takano APC. Impact of hyperthyroidism on cardiac hypertrophy. Endocr Connect 2020; 9:R59-R69. [PMID: 32101527 PMCID: PMC7159257 DOI: 10.1530/ec-19-0543] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 02/25/2020] [Indexed: 12/14/2022]
Abstract
The cardiac growth process (hypertrophy) is a crucial phenomenon conserved across a wide array of species and is critically involved in the maintenance of cardiac homeostasis. This process enables an organism to adapt to changes in systemic demand and occurs due to a plethora of responses, depending on the type of signal or stimuli received. The growth of cardiac muscle cells in response to environmental conditions depends on the type, strength and duration of stimuli, and results in adaptive physiological responses or non-adaptive pathological responses. Thyroid hormones (TH) have a direct effect on the heart and induce a cardiac hypertrophy phenotype, which may evolve to heart failure. In this review, we summarize the literature on TH function in the heart by presenting results from experimental studies. We discuss the mechanistic aspects of TH associated with cardiac myocyte hypertrophy, increased cardiac myocyte contractility and electrical remodeling, as well as the associated signaling pathways. In addition to classical crosstalk with the sympathetic nervous system (SNS), emerging work pointing to the new endocrine interaction between TH and the renin-angiotensin system (RAS) is also explored. Given the inflammatory potential of the angiotensin II peptide, this new interaction may open the door for new therapeutic approaches which target the key mechanisms responsible for TH-induced cardiac hypertrophy.
Collapse
Affiliation(s)
- M L M Barreto-Chaves
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - N Senger
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - M R Fevereiro
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - A C Parletta
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - A P C Takano
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
138
|
Al-Attar R, Storey KB. Suspended in time: Molecular responses to hibernation also promote longevity. Exp Gerontol 2020; 134:110889. [PMID: 32114078 DOI: 10.1016/j.exger.2020.110889] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 02/20/2020] [Accepted: 02/21/2020] [Indexed: 12/16/2022]
Abstract
Aging in most animals is an inevitable process that causes or is a result of physiological, biochemical, and molecular changes in the body, and has a strong influence on an organism's lifespan. Although advancement in medicine has allowed humans to live longer, the prevalence of age-associated medical complications is continuously burdening older adults worldwide. Current animal models used in research to study aging have provided novel information that has helped investigators understand the aging process; however, these models are limiting. Aging is a complex process that is regulated at multiple biological levels, and while a single manipulation in these models can provide information on a process, it is not enough to understand the global regulation of aging. Some mammalian hibernators live up to 9.8-times higher than their expected average lifespan, and new research attributes this increase to their ability to hibernate. A common theme amongst these mammalian hibernators is their ability to greatly reduce their metabolic rate to a fraction of their normal rate and initiate cytoprotective responses that enable their survival. Metabolic rate depression is strictly regulated at different biological levels in order to enable the animal to not only survive, but to also do so by relying mainly on their limited internal fuels. As such, understanding both the global and specific regulatory mechanisms used to promote survival during hibernation could, in theory, allow investigators to have a better understanding of the aging process. This can also allow pharmaceutical industries to find therapeutics that could delay or reverse age-associated medical complications and promote healthy aging and longevity in humans.
Collapse
Affiliation(s)
- Rasha Al-Attar
- Institute of Biochemistry and Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S 5B6, Canada.
| | - Kenneth B Storey
- Institute of Biochemistry and Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S 5B6, Canada.
| |
Collapse
|
139
|
Chromatin accessibility is associated with the changed expression of miRNAs that target members of the Hippo pathway during myoblast differentiation. Cell Death Dis 2020; 11:148. [PMID: 32094347 PMCID: PMC7039994 DOI: 10.1038/s41419-020-2341-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 02/08/2020] [Accepted: 02/10/2020] [Indexed: 12/11/2022]
Abstract
miRNAs reportedly participate in various biological processes, such as skeletal muscle proliferation and differentiation. However, the regulation of differentially expressed (DE) miRNAs and their function in myogenesis remain unclear. Herein, miRNA expression profiles and regulation during C2C12 differentiation were analyzed in relation to chromatin states by RNA-seq, ATAC-seq, and ChIP-seq. We identified 19 known and nine novel differentially expressed miRNAs at days 0, 1, 2, and 4. The expression of the differentially expressed miRNAs was related to the chromatin states of the 113 surrounding open chromatin regions defined by ATAC-seq peaks. Of these open chromatin regions, 44.25% were colocalized with MyoD/MyoG binding sites. The remainder of the above open chromatin regions were enriched with motifs of the myoblast-expressed AP-1 family, Ctcf, and Bach2 transcription factors (TFs). Additionally, the target genes of the above differentially expressed miRNAs were enriched primarily in muscle growth and development pathways, especially the Hippo signaling pathway. Moreover, via combining a loss-of-function assay with Q-PCR, western blotting, and immunofluorescence, we confirmed that the Hippo signaling pathway was responsible for C2C12 myoblast differentiation. Thus, our results showed that these differentially expressed miRNAs were regulated by chromatin states and affected muscle differentiation through the Hippo signaling pathway. Our findings provide new insights into the function of these differentially expressed miRNAs and the regulation of their expression during myoblast differentiation.
Collapse
|
140
|
Suzuki O, Koura M, Uchio-Yamada K, Sasaki M. Analysis of the transgene insertion pattern in a transgenic mouse strain using long-read sequencing. Exp Anim 2020; 69:279-286. [PMID: 32051389 PMCID: PMC7445054 DOI: 10.1538/expanim.19-0118] [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] [Indexed: 01/26/2023] Open
Abstract
Transgene insertion patterns are critical for the analysis of transgenic animals because
the influence of transgenes may change depending on the insertion pattern (such as copy
numbers and orientations of concatenations) and the insertion position in the genome. We
previously reported a genomic walking strategy to locate transgenes in the genomes of
transgenic mice (Exp. Anim. 53: 103–111, 2004) and to analyze transgene insertion patterns
(Exp. Anim. 55: 65–69, 2006). With such strategies, however, we could not determine the
copy number of transgenes or global genome modification induced by transgene insertion due
to read-length limitation. In this study, we used a long-read sequencer (MinION, Oxford
Nanopore Technologies) to overcome this limitation. We obtained 922,210 reads using MinION
with genomic DNA from a transgenic mouse strain (4C30, Proc. Jpn. Acad. Ser. B. Phys.
Biol. Sci. 87: 550–562, 2011). Among the reads, we found one 21,457-bp read containing the
transgene using a local BLAST search. Nucleotide dot plot analysis revealed that the
transgene was inserted in the genome as a tandem concatemer with an almost entire
construct (15–3,508 of 3,508 bp) and a partial fragment (4–660, 657 bp). Ensembl’s BLAST
search against the C57BL/6N genome revealed a 9,388-bp deletion at the insertion position
in the intron of the Sgcd gene, confirming that mutations such as a large
genomic deletion could occur at the time of transgene insertion. Thus, long-read
sequencers are useful tools for the analysis of transgene insertion patterns.
Collapse
Affiliation(s)
- Osamu Suzuki
- Laboratory of Animal Models for Human Diseases, National Institutes of Biomedical Innovation, Health and Nutrition,7-6-8 Saito-Asagi, Ibaraki, Ibaraki, Osaka 568-0085, Japan
| | - Minako Koura
- Laboratory of Animal Models for Human Diseases, National Institutes of Biomedical Innovation, Health and Nutrition,7-6-8 Saito-Asagi, Ibaraki, Ibaraki, Osaka 568-0085, Japan
| | - Kozue Uchio-Yamada
- Laboratory of Animal Models for Human Diseases, National Institutes of Biomedical Innovation, Health and Nutrition,7-6-8 Saito-Asagi, Ibaraki, Ibaraki, Osaka 568-0085, Japan
| | - Mitsuho Sasaki
- Laboratory of Animal Models for Human Diseases, National Institutes of Biomedical Innovation, Health and Nutrition,7-6-8 Saito-Asagi, Ibaraki, Ibaraki, Osaka 568-0085, Japan
| |
Collapse
|
141
|
Zhao X, Wang Y, Sun X. The functions of microRNA-208 in the heart. Diabetes Res Clin Pract 2020; 160:108004. [PMID: 31911250 DOI: 10.1016/j.diabres.2020.108004] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 12/15/2019] [Accepted: 12/31/2019] [Indexed: 02/07/2023]
Abstract
Cardiovascular disease is a major chronic complication of obesity and diabetes. Due to several patients with obesity and diabetes, it is necessary to urgently explore early diagnostic biomarkers and innovative therapeutic strategies to prevent the progression of cardiovascular diseases. Recently, microRNAs (also known as miRNAs) have emerged as important players in heart disease and energy regulation. MiRNAs are a group of small, highly conserved non-coding RNA molecules that regulate gene expression by suppressing the translation of messenger RNA of target genes or by promoting mRNA degradation. These act as a class of potential biomarkers and may provide key information in diagnosing common diseases such as tumors, tissue damage, and autoimmune diseases. Among all the known miRNAs, microRNA-208 (miR-208) is specifically expressed in myocardial cells and showed close association with the development of cardiac diseases, such as myocardial hypertrophy, cardiac fibrosis, myocardial infarction, arrhythmia, and heart failure. However, the functions and underlying mechanisms of miR-208 in heart are still unclear. In this review, we highlighted the novel insights of miR-208 functions and associated mechanisms in the regulation of cardiac diseases.
Collapse
Affiliation(s)
- Xin Zhao
- Department of Outpatient Clinic, ShanDong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yao Wang
- Shandong Institute of Endocrine and Metabolic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Xianglan Sun
- Department of Geriatrics, Department of Geriatric Endocrinology, ShanDong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.
| |
Collapse
|
142
|
Mirzadeh Azad F, Arabian M, Maleki M, Malakootian M. Small Molecules with Big Impacts on Cardiovascular Diseases. Biochem Genet 2020; 58:359-383. [PMID: 31997044 DOI: 10.1007/s10528-020-09948-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Accepted: 01/13/2020] [Indexed: 12/22/2022]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of morbidity and mortality worldwide. Although in recent years there has been a significant progress in the diagnosis, treatment, and prognosis of CVD, but due to their complex pathobiology, developing novel biomarkers and therapeutic interventions are still in need. MicroRNAs (miRNAs) are a fraction of non-coding RNAs that act as micro-regulators of gene expression. Mounting evidences over the last decade confirmed that microRNAs were deregulated in several CVDs and manipulating their expression could affect homeostasis, differentiation, and function of cardiovascular system. Here, we review the current knowledge concerning the roles of miRNAs in cardiovascular diseases with more details on cardiac remodeling, arrhythmias, and atherosclerosis. In addition, we discuss the latest findings on the potential therapeutic applications of miRNAs in cardiovascular diseases.
Collapse
Affiliation(s)
- Fatemeh Mirzadeh Azad
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Maedeh Arabian
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Majid Maleki
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mahshid Malakootian
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
143
|
Condrat CE, Thompson DC, Barbu MG, Bugnar OL, Boboc A, Cretoiu D, Suciu N, Cretoiu SM, Voinea SC. miRNAs as Biomarkers in Disease: Latest Findings Regarding Their Role in Diagnosis and Prognosis. Cells 2020; 9:E276. [PMID: 31979244 PMCID: PMC7072450 DOI: 10.3390/cells9020276] [Citation(s) in RCA: 829] [Impact Index Per Article: 165.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 01/14/2020] [Accepted: 01/21/2020] [Indexed: 02/06/2023] Open
Abstract
MicroRNAs (miRNAs) represent a class of small, non-coding RNAs with the main roles of regulating mRNA through its degradation and adjusting protein levels. In recent years, extraordinary progress has been made in terms of identifying the origin and exact functions of miRNA, focusing on their potential use in both the research and the clinical field. This review aims at improving the current understanding of these molecules and their applicability in the medical field. A thorough analysis of the literature consulting resources available in online databases such as NCBI, PubMed, Medline, ScienceDirect, and UpToDate was performed. There is promising evidence that in spite of the lack of standardized protocols regarding the use of miRNAs in current clinical practice, they constitute a reliable tool for future use. These molecules meet most of the required criteria for being an ideal biomarker, such as accessibility, high specificity, and sensitivity. Despite present limitations, miRNAs as biomarkers for various conditions remain an impressive research field. As current techniques evolve, we anticipate that miRNAs will become a routine approach in the development of personalized patient profiles, thus permitting more specific therapeutic interventions.
Collapse
Affiliation(s)
- Carmen Elena Condrat
- Alessandrescu-Rusescu National Institute for Mother and Child Health, Fetal Medicine Excellence Research Center, 020395 Bucharest, Romania; (C.E.C.); (D.C.T.); (M.G.B.); (O.L.B.); (A.B.); (D.C.); (N.S.)
| | - Dana Claudia Thompson
- Alessandrescu-Rusescu National Institute for Mother and Child Health, Fetal Medicine Excellence Research Center, 020395 Bucharest, Romania; (C.E.C.); (D.C.T.); (M.G.B.); (O.L.B.); (A.B.); (D.C.); (N.S.)
| | - Madalina Gabriela Barbu
- Alessandrescu-Rusescu National Institute for Mother and Child Health, Fetal Medicine Excellence Research Center, 020395 Bucharest, Romania; (C.E.C.); (D.C.T.); (M.G.B.); (O.L.B.); (A.B.); (D.C.); (N.S.)
| | - Oana Larisa Bugnar
- Alessandrescu-Rusescu National Institute for Mother and Child Health, Fetal Medicine Excellence Research Center, 020395 Bucharest, Romania; (C.E.C.); (D.C.T.); (M.G.B.); (O.L.B.); (A.B.); (D.C.); (N.S.)
| | - Andreea Boboc
- Alessandrescu-Rusescu National Institute for Mother and Child Health, Fetal Medicine Excellence Research Center, 020395 Bucharest, Romania; (C.E.C.); (D.C.T.); (M.G.B.); (O.L.B.); (A.B.); (D.C.); (N.S.)
| | - Dragos Cretoiu
- Alessandrescu-Rusescu National Institute for Mother and Child Health, Fetal Medicine Excellence Research Center, 020395 Bucharest, Romania; (C.E.C.); (D.C.T.); (M.G.B.); (O.L.B.); (A.B.); (D.C.); (N.S.)
- Department of Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd., 050474 Bucharest, Romania
| | - Nicolae Suciu
- Alessandrescu-Rusescu National Institute for Mother and Child Health, Fetal Medicine Excellence Research Center, 020395 Bucharest, Romania; (C.E.C.); (D.C.T.); (M.G.B.); (O.L.B.); (A.B.); (D.C.); (N.S.)
- Division of Obstetrics, Gynecology and Neonatology, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd., 050474 Bucharest, Romania
- Department of Obstetrics and Gynecology, Polizu Clinical Hospital, Alessandrescu-Rusescu National Institute for Mother and Child Health, 020395 Bucharest, Romania
| | - Sanda Maria Cretoiu
- Department of Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd., 050474 Bucharest, Romania
| | - Silviu Cristian Voinea
- Department of Surgical Oncology, Prof. Dr. Alexandru Trestioreanu Oncology Institute, Carol Davila University of Medicine and Pharmacy, 252 Fundeni Rd., 022328 Bucharest, Romania;
| |
Collapse
|
144
|
Ferrero KM, Koch WJ. Metabolic Crosstalk between the Heart and Fat. Korean Circ J 2020; 50:379-394. [PMID: 32096362 PMCID: PMC7098822 DOI: 10.4070/kcj.2019.0400] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 12/27/2019] [Indexed: 12/14/2022] Open
Abstract
It is now recognized that the heart can behave as a true endocrine organ, which can modulate the function of other tissues. Emerging evidence has shown that visceral fat is one such distant organ the heart communicates with. In fact, it appears that bi-directional crosstalk between adipose tissue and the myocardium is crucial to maintenance of normal function in both organs. In particular, factors secreted from the heart are now known to influence the metabolic activity of adipose tissue and other organs, as well as modulate the release of metabolic substrates and signaling molecules from the periphery. This review summarizes current knowledge regarding primary cardiokines and adipokines involved in heart-fat crosstalk, as well as implications of their dysregulation for cardiovascular health.
Collapse
Affiliation(s)
- Kimberly M Ferrero
- Center for Translational Medicine, Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Walter J Koch
- Center for Translational Medicine, Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
145
|
Yang M, Zhang J, Jin X, Li C, Zhou G, Feng J. NRF1-enhanced miR-4458 alleviates cardiac hypertrophy through releasing TTP-inhibited TFAM. In Vitro Cell Dev Biol Anim 2020; 56:120-128. [PMID: 31942725 DOI: 10.1007/s11626-019-00419-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 11/04/2019] [Indexed: 01/18/2023]
Abstract
Growing evidence suggests the crucial role of microRNAs (miRNAs) in regulating basic cell functions, and therefore participating in the pathologic development of diverse human diseases, including cardiac hypertrophy. Herein, we explained that miR-4458 was distinctly stimulated in Ang II-stimulated hypertrophic H9c2 cells. Intriguingly, miR-4458 inhibition led to exacerbated hypertrophic phenotypes in Ang II-treated H9c2 cells. In addition, the compensatory upregulation of miR-4458 in Ang II-treated H9c2 cells was ascribed to its transcriptional enhancement by NRF1, a transcription factor previously identified to be activated in early cardiac hypertrophy. Moreover, we discovered that miR-4458 served as a negative modulator in cardiac hypertrophy by prompting TFAM, a well-recognized myocardial protective protein. TTP, a RBP that always leads to degradation of recognized mRNAs, was predicted to interact with both miR-4458 and TFAM mRNA. Importantly, we verified that miR-4458 facilitated TFAM expression in cardiomyocytes by directly targeting TTP and releasing TTP-destabilized TFAM mRNA. On the whole, these findings demonstrated that NRF1-induced miR-4458 boosted TFAM via targeting TTP to dampen the exacerbation of cardiac hypertrophy, which indicates miR-4458 as a promising biomarker for the cardiac hypertrophy treatment.
Collapse
Affiliation(s)
- Mengsi Yang
- Department of Cardiology, The Second People's Hospital in Hefei City, Hefei, 241000, China
| | - Jing Zhang
- Department of Cardiology, The Second People's Hospital in Hefei City, Hefei, 241000, China
| | - Xiaoqin Jin
- Department of Cardiology, The Second People's Hospital in Hefei City, Hefei, 241000, China
| | - Chao Li
- Department of Cardiology, The Second People's Hospital in Hefei City, Hefei, 241000, China
| | - Gaoliang Zhou
- Department of Cardiology, The Second People's Hospital in Hefei City, Hefei, 241000, China
| | - Jun Feng
- Department of Cardiology, The Second People's Hospital in Hefei City, Hefei, 241000, China.
| |
Collapse
|
146
|
Wang C, He R, Liu X, Li J, Pan R. miR-29a activates expression of α-cardiac myosin heavy chain via β1 thyroid hormone receptor in neonatal rats. Arch Med Sci 2020; 20:641-654. [PMID: 38757019 PMCID: PMC11094831 DOI: 10.5114/aoms.2019.91495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 11/24/2019] [Indexed: 05/18/2024] Open
Abstract
Introduction MicroRNAs (miRs) are small noncoding RNAs which are regulators of gene expression and also regulate the genes in heart tissues. The aim of the study was to evaluate the effect of miRs on the expression level of myosin heavy chain (MHC), which is responsible for regulation of cardiac functions in neonatal rat ventricular myocytes and mice. Material and methods The miRs were suppressed in neonatal rat ventricular myocytes using small interfering RNAs (siRNAs) against Dicer followed by evaluation of MHC levels. For in vivo study the C57 black/6 Jacksonian mice were subjected to the transverse aortic constriction (TAC) procedure. Results The Dicer siRNA suppressed the endogenous miRs and the α-MHC gene but failed to down-regulate the β-MHC. Among the 17 selected miRs, miR-29a was found to up-regulate the α-MHC gene significantly but not β-MHC. The expression of α-MHC was suppressed by silencing the expression of miR-29a. Bioinformatics study done by TargetScan suggested thyroid hormone receptor-β1 (TR-β1) as a potential target of miR-29a. Additionally, miR-29a was found to regulate the expression of α-MHC via TR-β1 signaling. Conclusions The findings of the present study indicated that miR-29a modulates expression of α-the MHC gene by targeting TR-β1 in cardiac cells. The study may provide a new direction for treating cardiac failure and cardiac hypertrophy.
Collapse
Affiliation(s)
- Chengbin Wang
- Departments of Pediatrics, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Ru He
- Departments of Pediatrics, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Xiaohui Liu
- Departments of Pediatrics, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Junhua Li
- Departments of Pediatrics, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Rui Pan
- Departments of Pediatrics, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| |
Collapse
|
147
|
Kapodistrias N, Theocharopoulou G, Vlamos P. A Hypothesis of Circulating MicroRNAs' Implication in High Incidence of Atrial Fibrillation and Other Electrocardiographic Abnormalities in Cancer Patients. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1196:1-9. [PMID: 32468302 DOI: 10.1007/978-3-030-32637-1_1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
MicroRNAs are short non-coding RNA molecules that control posttranscriptional gene expression and are present in tissues cells but also circulate in biological fluids in various forms (exosome, connected with proteins, apoptotic bodies, etc.). The roles that circulated extracellular serum microRNAs possess in cancer development, like in the delivery from a recipient cell to distant tissues and the repression of host genes resulting in the impairment of critical functions, are still undetermined. Disturbances, such as the higher incidence of atrial fibrillation in cancer patients, could be analyzed in the frame of suppressive action of circulated microRNAs in genes that control cardiac conduction in atrium. More precisely, mir-21 overexpression in tissues promotes atrium fibrosis and impairs conductibility. A possible hypothesis is that the high levels of circulating microRNA in cancer may exert the same effect. Further experiments are necessary to corroborate the hypothesis.
Collapse
|
148
|
Jiang W, Agrawal DK, Boosani CS. Non-coding RNAs as Epigenetic Gene Regulators in Cardiovascular Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1229:133-148. [PMID: 32285409 DOI: 10.1007/978-981-15-1671-9_7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Epigenetic gene regulations can be considered as de-novo initiation of abnormal molecular signaling events whose regulation is otherwise required during normal or specific developmental stages of the organisms. Primarily, three different mechanisms have been identified to participate in epigenetic gene regulations which include, DNA methylation, non-coding RNA species (microRNAs [miRNA], and long non-coding RNAs [LNC-RNA]) and histone modifications. These de-novo epigenetic mechanisms have been associated with altered normal cellular functions which eventually facilitate normal cells to transition into an abnormal phenotype. Among the three modes of regulation, RNA species which are usually considered to be less stable, can be speculated to initiate instant alterations in gene expression compared to DNA methylation or histone modifications. However, LNC-RNAs appear to be more stable in the cells than the other RNA species. Moreover, there is increasing literature which clearly suggests that a single specific LNC-RNA can regulate multiple mechanisms and disease phenotypes. With specific focus on cardiovascular diseases, here we attempt to provide UpToDate information on the functional role of miRNAs and LNC-RNAs. Here we discuss the role of these epigenetic mediators in different components of cardiovascular disease which include physiopathological heart development, athersclerosis, retenosis, diabetic hearts, myocardial infarction, ischemia-reperfusion, heart valve disease, aortic aneurysm, osteogenesis, angiogenesis and hypoxia in the heart. While there is abundant literature support that shows the involvement of many LNC-RNAs and miRNAs in cardiovascular diseases, very few RNA species have been identified which regulate epigenetic mechanisms which is the current focus in this article. Understanding the role of these RNA species in regulating epigenetic mechanisms in different cell types causing cardiovascular disease, would advance the field and promote disease prevention approaches that are aimed to target epigenetic mechanisms.
Collapse
Affiliation(s)
- Wanlin Jiang
- Department of Clinical & Translational Research, Western University of Health Sciences, Pomona, CA, USA
| | - Devendra K Agrawal
- Department of Clinical & Translational Research, Western University of Health Sciences, Pomona, CA, USA
| | - Chandra Shekhar Boosani
- Department of Clinical & Translational Research, Western University of Health Sciences, Pomona, CA, USA.
| |
Collapse
|
149
|
Shao J, Lin W, Lin B, Wang Q, Chen Y, Fan C. MiR-377 accelerates cardiac hypertrophy by inhibiting autophagy via targeting PPAR γ. ALL LIFE 2020. [DOI: 10.1080/26895293.2020.1808083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Affiliation(s)
- Jianzhi Shao
- Department of Cardiology, The First People’s Hospital of Wenling, Wenling, People’s Republic of China
| | - Wenhui Lin
- Department of Cardiology, The First People’s Hospital of Wenling, Wenling, People’s Republic of China
| | - Bin Lin
- Department of Cardiology, The First People’s Hospital of Wenling, Wenling, People’s Republic of China
| | - Qizeng Wang
- Department of Cardiology, The First People’s Hospital of Wenling, Wenling, People’s Republic of China
| | - Yunpeng Chen
- Department of Cardiology, The First People’s Hospital of Wenling, Wenling, People’s Republic of China
| | - Chenrong Fan
- Department of Cardiology, The First People’s Hospital of Wenling, Wenling, People’s Republic of China
| |
Collapse
|
150
|
Šustr F, Stárek Z, Souček M, Novák J. Non-coding RNAs and Cardiac Arrhythmias. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1229:287-300. [PMID: 32285419 DOI: 10.1007/978-981-15-1671-9_17] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/26/2023]
Abstract
Cardiac arrhythmias represent wide and heterogenic group of disturbances in the cardiac rhythm. Pathophysiology of individual arrhythmias is highly complex and dysfunction in ion channels/currents involved in generation or spreading of action potential is usually documented. Non-coding RNAs (ncRNAs) represent highly variable group of molecules regulating the heart expression program, including regulation of the expression of individual ion channels and intercellular connection proteins, e.g. connexins.Within this chapter, we will describe basic electrophysiological properties of the myocardium. We will focus on action potential generation and spreading in pacemaker and non-pacemaker cells, including description of individual ion channels (natrium, potassium and calcium) and their ncRNA-mediated regulation. Most of the studies have so far focused on microRNAs, thus, their regulatory function will be described into greater detail. Clinical consequences of altered ncRNA regulatory function will also be described together with potential future directions of the research in the field.
Collapse
Affiliation(s)
- Filip Šustr
- Second Department of Internal Medicine of St. Anne's University Hospital in Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Zdeněk Stárek
- First Department of Internal Medicine and Cardioangiology of St. Anne's University Hospital in Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Miroslav Souček
- Second Department of Internal Medicine of St. Anne's University Hospital in Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Jan Novák
- Second Department of Internal Medicine of St. Anne's University Hospital in Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic.
- CEITEC - Central European Institute for Technology, Masaryk University, Brno, Czech Republic.
| |
Collapse
|