151
|
miRNAs that Induce Human Cardiomyocyte Proliferation Converge on the Hippo Pathway. Cell Rep 2019; 23:2168-2174. [PMID: 29768213 PMCID: PMC6261450 DOI: 10.1016/j.celrep.2018.04.049] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 07/28/2017] [Accepted: 04/12/2018] [Indexed: 11/23/2022] Open
Abstract
Understanding the mechanisms that control human cardiomyocyte proliferation might be applicable to regenerative medicine. We screened a whole genome collection of human miRNAs, identifying 96 to be capable of increasing proliferation (DNA synthesis and cytokinesis) of human iPSC-derived cardiomyocytes. Chemical screening and computational approaches indicated that most of these miRNAs (67) target different components of the Hippo pathway and that their activity depends on the nuclear translocation of the Hippo transcriptional effector YAP. 53 of the 67 miRNAs are present in human iPSC cardiomyocytes, yet anti-miRNA screening revealed that none are individually essential for basal proliferation of hiPSC cardiomyocytes despite the importance of YAP for proliferation. We propose a model in which multiple endogenous miRNAs redundantly suppress Hippo signaling to sustain the cell cycle of immature cardiomyocytes.
Collapse
|
152
|
Shen NN, Zhang C, Li Z, Kong LC, Wang XH, Gu ZC, Wang JL. MicroRNA expression signatures of atrial fibrillation: The critical systematic review and bioinformatics analysis. Exp Biol Med (Maywood) 2019; 245:42-53. [PMID: 31766887 DOI: 10.1177/1535370219890303] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Association between microRNA (miRNA) expression signatures and atrial fibrillation has been evaluated with inconsistent findings in different studies. This study aims to identify miRNAs that actually play vital role in pathophysiological process of atrial fibrillation and explore miRNA-targeted genes and the involved pathways. Relevant studies were retrieved from the electronic databases of Embase, Medline, and Cochrane Library to determine the miRNA expression profiles between atrial fibrillation subjects and non-atrial fibrillation controls. Robustness of results was assessed using sensitivity analysis. Subgroup analyses were performed based on species, miRNA detection method, sample source, and ethnicity. Quality assessment of studies was independently conducted according to QUADAS-2. Bioinformatics analysis was applied to explore the potential genes and pathways associated with atrial fibrillation, which were targeted by differentially expressed miRNAs. Form of pooled results was shown as log10 odds ratios (logORs) with 95% confidence intervals (CI), and random-effects model was used. In total, 40 articles involving 283 differentially expressed miRNAs were reported. And 51 significantly dysregulated miRNAs were identified in consistent direction, with 22 upregulated and 29 downregulated. Among above-mentioned miRNAs, miR-223-3p (logOR 6.473; P < 0.001) was the most upregulated, while miR-1-5p (logOR 7.290; P < 0.001) was the most downregulated. Subgroup analysis confirmed 53 significantly dysregulated miRNAs (21 upregulated and 32 downregulated) in cardiac tissue, with miRNA-1-5p and miRNA-223-3p being the most upregulated and downregulated miRNAs, respectively. Additionally, miR-328 and miR-1-5p were highly blood-specific, and miR-133 was animal-specific. In the detection method sub-groups, miRNA-29b and miRNA-223-3p were differentially expressed consistently. Four miRNAs, including miRNA-223-3p, miRNA-21, miRNA-328, and miRNA-1-5p, were consistently dysregulated in both Asian and non-Asian. Results of sensitivity analysis showed that 47 out of 51 (92.16%) miRNAs were dysregulated consistently. Totally, 51 consistently dysregulated miRNAs associated with atrial fibrillation were confirmed in this study. Five important miRNAs, including miR-29b, miR-328, miR-1-5p, miR-21, and miR-223-3p may act as potential biomarkers for atrial fibrillation. Impact statement Atrial fibrillation (AF) is considered as the most common arrhythmia, and it subsequently causes serious complications including thrombosis and heart failure that increase the social burden. The definite mechanisms underlying AF pathogenesis remain complicated and unclear. Many studies attempted to discover the transcriptomic changes using microarray technologies, and the present studies for this hot topic have assessed individual miRNAs profiles for AF. However, results of different articles are controversial and not each reported miRNA is actually associated with the pathogenesis of AF. The present systematic review and meta-analysis identified that 51 consistently dysregulated miRNAs were associated with AF. Of these miRNAs, five miRNAs (miRNA-1-5p, miRNA-328, miRNA-29b, miRNA-21, and miRNA-223-3p) may act as novel biomarkers for AF. The findings could offer a better description of the biological characteristics of miRNAs, meanwhile might serve as new target for the intervention and monitoring AF in future studies.
Collapse
Affiliation(s)
- Nan-Nan Shen
- Department of Pharmacy, Affiliated Hospital of Shaoxing University, Shao Xing 312000, China.,State Key Laboratory of Oncogenes and Related Genes, Department of Pharmacy, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Chi Zhang
- State Key Laboratory of Oncogenes and Related Genes, Department of Pharmacy, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Zheng Li
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Ling-Cong Kong
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Xin-Hua Wang
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Zhi-Chun Gu
- State Key Laboratory of Oncogenes and Related Genes, Department of Pharmacy, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Jia-Liang Wang
- Department of Pharmacy, Affiliated Hospital of Shaoxing University, Shao Xing 312000, China
| |
Collapse
|
153
|
Zlabinger K, Spannbauer A, Traxler D, Gugerell A, Lukovic D, Winkler J, Mester-Tonczar J, Podesser B, Gyöngyösi M. MiR-21, MiR-29a, GATA4, and MEF2c Expression Changes in Endothelin-1 and Angiotensin II Cardiac Hypertrophy Stimulated Isl-1 +Sca-1 +c-kit + Porcine Cardiac Progenitor Cells In Vitro. Cells 2019; 8:cells8111416. [PMID: 31717562 PMCID: PMC6912367 DOI: 10.3390/cells8111416] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/06/2019] [Accepted: 11/07/2019] [Indexed: 12/16/2022] Open
Abstract
Cost- and time-intensive porcine translational disease models offer great opportunities to test drugs and therapies for pathological cardiac hypertrophy and can be supported by porcine cell culture models that provide further insights into basic disease mechanisms. Cardiac progenitor cells (CPCs) residing in the adult heart have been shown to differentiate in vitro into cardiomyocytes and could contribute to cardiac regeneration. Therefore, it is important to evaluate their changes on the cellular level caused by disease. We successfully isolated Isl1+Sca1+cKit+ porcine CPCs (pCPCs) from pig hearts and stimulated them with endothelin-1 (ET-1) and angiotensin II (Ang II) in vitro. We also performed a cardiac reprogramming transfection and tested the same conditions. Our results show that undifferentiated Isl1+Sca1+cKit+ pCPCs were significantly upregulated in GATA4, MEF2c, and miR-29a gene expressions and in BNP and MCP-1 protein expressions with Ang II stimulation, but they showed no significant changes in miR-29a and MCP-1 when stimulated with ET-1. Differentiated Isl1+Sca1+cKit+ pCPCs exhibited significantly higher levels of MEF2c, GATA4, miR-29a, and miR-21 as well as Cx43 and BNP with Ang II stimulation. pMx-MGT-transfected Isl1+Sca1+cKit+ pCPCs showed significant elevations in MEF2c, GATA4, and BNP expressions when stimulated with ET-1. Our model demonstrates that in vitro stimulation leads to successful Isl1+Sca1+cKit+ pCPC hypertrophy with upregulation of cardiac remodeling associated genes and profibrotic miRNAs and offers great possibilities for further investigations of disease mechanisms and treatment.
Collapse
Affiliation(s)
- Katrin Zlabinger
- Medical University of Vienna, Department of Cardiology, 1090 Vienna, Austria; (A.S.); (D.T.); (A.G.); (D.L.); (J.W.); (J.M.-T.)
- Correspondence: (K.Z.); (M.G.); Tel.: +43(0)-140-400-48520 (K.Z.)
| | - Andreas Spannbauer
- Medical University of Vienna, Department of Cardiology, 1090 Vienna, Austria; (A.S.); (D.T.); (A.G.); (D.L.); (J.W.); (J.M.-T.)
| | - Denise Traxler
- Medical University of Vienna, Department of Cardiology, 1090 Vienna, Austria; (A.S.); (D.T.); (A.G.); (D.L.); (J.W.); (J.M.-T.)
| | - Alfred Gugerell
- Medical University of Vienna, Department of Cardiology, 1090 Vienna, Austria; (A.S.); (D.T.); (A.G.); (D.L.); (J.W.); (J.M.-T.)
| | - Dominika Lukovic
- Medical University of Vienna, Department of Cardiology, 1090 Vienna, Austria; (A.S.); (D.T.); (A.G.); (D.L.); (J.W.); (J.M.-T.)
| | - Johannes Winkler
- Medical University of Vienna, Department of Cardiology, 1090 Vienna, Austria; (A.S.); (D.T.); (A.G.); (D.L.); (J.W.); (J.M.-T.)
| | - Julia Mester-Tonczar
- Medical University of Vienna, Department of Cardiology, 1090 Vienna, Austria; (A.S.); (D.T.); (A.G.); (D.L.); (J.W.); (J.M.-T.)
| | - Bruno Podesser
- Medical University of Vienna, Department of Biomedical Research, 1090 Vienna, Austria;
| | - Mariann Gyöngyösi
- Medical University of Vienna, Department of Cardiology, 1090 Vienna, Austria; (A.S.); (D.T.); (A.G.); (D.L.); (J.W.); (J.M.-T.)
- Correspondence: (K.Z.); (M.G.); Tel.: +43(0)-140-400-48520 (K.Z.)
| |
Collapse
|
154
|
Miklas JW, Clark E, Levy S, Detraux D, Leonard A, Beussman K, Showalter MR, Smith AT, Hofsteen P, Yang X, Macadangdang J, Manninen T, Raftery D, Madan A, Suomalainen A, Kim DH, Murry CE, Fiehn O, Sniadecki NJ, Wang Y, Ruohola-Baker H. TFPa/HADHA is required for fatty acid beta-oxidation and cardiolipin re-modeling in human cardiomyocytes. Nat Commun 2019; 10:4671. [PMID: 31604922 PMCID: PMC6789043 DOI: 10.1038/s41467-019-12482-1] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 09/10/2019] [Indexed: 02/07/2023] Open
Abstract
Mitochondrial trifunctional protein deficiency, due to mutations in hydratase subunit A (HADHA), results in sudden infant death syndrome with no cure. To reveal the disease etiology, we generated stem cell-derived cardiomyocytes from HADHA-deficient hiPSCs and accelerated their maturation via an engineered microRNA maturation cocktail that upregulated the epigenetic regulator, HOPX. Here we report, matured HADHA mutant cardiomyocytes treated with an endogenous mixture of fatty acids manifest the disease phenotype: defective calcium dynamics and repolarization kinetics which results in a pro-arrhythmic state. Single cell RNA-seq reveals a cardiomyocyte developmental intermediate, based on metabolic gene expression. This intermediate gives rise to mature-like cardiomyocytes in control cells but, mutant cells transition to a pathological state with reduced fatty acid beta-oxidation, reduced mitochondrial proton gradient, disrupted cristae structure and defective cardiolipin remodeling. This study reveals that HADHA (tri-functional protein alpha), a monolysocardiolipin acyltransferase-like enzyme, is required for fatty acid beta-oxidation and cardiolipin remodeling, essential for functional mitochondria in human cardiomyocytes.
Collapse
Affiliation(s)
- Jason W Miklas
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Elisa Clark
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Shiri Levy
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA
- Department of Biochemistry, University of Washington, School of Medicine, Seattle, WA, 98195, USA
| | - Damien Detraux
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA
- Department of Biochemistry, University of Washington, School of Medicine, Seattle, WA, 98195, USA
| | - Andrea Leonard
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA
- Department of Mechanical Engineering, University of Washington, Seattle, WA, 98195, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, 98109, USA
| | - Kevin Beussman
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA
- Department of Mechanical Engineering, University of Washington, Seattle, WA, 98195, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, 98109, USA
| | - Megan R Showalter
- NIH West Coast Metabolomics Center, University of California Davis, Davis, CA, 95616, USA
| | - Alec T Smith
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Peter Hofsteen
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, 98109, USA
- Department of Pathology, University of Washington, Seattle, WA, 98109, USA
| | - Xiulan Yang
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, 98109, USA
- Department of Pathology, University of Washington, Seattle, WA, 98109, USA
| | - Jesse Macadangdang
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Tuula Manninen
- Helsinki University Hospital, 00290, Helsinki, Finland
- Research Programs Unit, Stem Cells and Metabolism, University of Helsinki, 00290, Helsinki, Finland
| | - Daniel Raftery
- Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center, University of Washington, Seattle, WA, 98109, USA
| | - Anup Madan
- Covance Genomics Laboratory, Redmond, WA, 98052, USA
| | - Anu Suomalainen
- Helsinki University Hospital, 00290, Helsinki, Finland
- Research Programs Unit, Stem Cells and Metabolism, University of Helsinki, 00290, Helsinki, Finland
- Neuroscience Center, University of Helsinki, 00290, Helsinki, Finland
| | - Deok-Ho Kim
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Charles E Murry
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, 98109, USA
- Department of Pathology, University of Washington, Seattle, WA, 98109, USA
- Department of Medicine/Cardiology, University of Washington, Seattle, WA, 98109, USA
| | - Oliver Fiehn
- NIH West Coast Metabolomics Center, University of California Davis, Davis, CA, 95616, USA
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Nathan J Sniadecki
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
- Department of Mechanical Engineering, University of Washington, Seattle, WA, 98195, USA
- Department of Pathology, University of Washington, Seattle, WA, 98109, USA
| | - Yuliang Wang
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA
- Paul G. Allen School of Computer Science & Engineering, University of Washington, Seattle, WA, 98195, USA
| | - Hannele Ruohola-Baker
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA.
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA.
- Department of Biochemistry, University of Washington, School of Medicine, Seattle, WA, 98195, USA.
| |
Collapse
|
155
|
Lee MJ, Fallen S, Zhou Y, Baxter D, Scherler K, Kuo MF, Wang K. The Impact of Moyamoya Disease and RNF213 Mutations on the Spectrum of Plasma Protein and MicroRNA. J Clin Med 2019; 8:jcm8101648. [PMID: 31658621 PMCID: PMC6832561 DOI: 10.3390/jcm8101648] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 09/30/2019] [Accepted: 10/08/2019] [Indexed: 12/23/2022] Open
Abstract
Moyamoya disease (MMD) is a rare cerebrovascular disorder characterized by occlusion of bilateral internal carotid and intracerebral arteries with the compensatory growth of fragile small vessels. MMD patients develop recurrent infarctions in the basal ganglia and subcortical regions. Symptoms include transient ischemic attack or stroke, seizures, and headaches, which may occur suddenly or in a stepwise progression. Mutations in Ring Finger Protein 213 (RNF213), a Zinc ring finger protein, have been identified in some MMD patients but the etiology of MMD is still largely unknown. To gain insight into the pathophysiology of MMD, we characterized the impact of the RNF213 mutations on plasma protein and RNA profiles. Isobaric tags for relative and absolute quantitation and proximity extension assay were used to characterize the plasma proteome. Next generation sequencing-based small RNAseq was used to analyze the cell-free small RNAs in whole plasma and RNA encapsulated in extracellular vesicles. The changes of miRNAs and proteins identified are associated with signaling processes including angiogenesis and immune activities which may reflect the pathology and progression of MMD.
Collapse
Affiliation(s)
- Ming-Jen Lee
- Department of Neurology, National Taiwan University Hospital, Taipei 10002, Taiwan.
| | | | - Yong Zhou
- Institute for Systems Biology, Seattle, WA 98109, USA.
| | - David Baxter
- Institute for Systems Biology, Seattle, WA 98109, USA.
| | | | - Meng-Fai Kuo
- Department of Neurosurgery, National Taiwan University Hospital, Taipei 10002, Taiwan.
| | - Kai Wang
- Institute for Systems Biology, Seattle, WA 98109, USA.
| |
Collapse
|
156
|
The Role of Exo-miRNAs in Cancer: A Focus on Therapeutic and Diagnostic Applications. Int J Mol Sci 2019; 20:ijms20194687. [PMID: 31546654 PMCID: PMC6801421 DOI: 10.3390/ijms20194687] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 09/12/2019] [Accepted: 09/19/2019] [Indexed: 12/16/2022] Open
Abstract
Exosomes are extracellular vesicles released into biological fluids where they act as carriers of various molecules, including proteins, lipids, and RNAs, between cells, modulating or perturbing specific physiological processes. Recently, it has been suggested that tumoral cells release excessive amounts of exosomes that, through their cargo, promote tumor progression, stimulating growth, angiogenesis, metastasis, insensitivity to chemotherapy, and immune evasion. Increasing evidence highlights exosomal microRNAs (exo-miRNAs) as important players in tumorigenesis. MicroRNA (miRNA) are a class of small non-coding RNA able to regulate gene expression, targeting multiple mRNAs and inducing translational repression and/or mRNA degradation. Exo-miRNAs are highly stable and easily detectable in biological fluids, and for these reasons, miRNAs are potential cancer biomarkers useful diagnostically and prognostically. Furthermore, since exosomes are natural delivery systems between cells, they can be appropriately modified to carry therapeutic miRNAs to specific recipient cells. Here we summarize the main functions of exo-miRNAs and their possible role for diagnostic and therapeutic applications.
Collapse
|
157
|
Kanbay M, Yerlikaya A, Sag AA, Ortiz A, Kuwabara M, Covic A, Wiecek A, Stenvinkel P, Afsar B. A journey from microenvironment to macroenvironment: the role of metaflammation and epigenetic changes in cardiorenal disease. Clin Kidney J 2019; 12:861-870. [PMID: 31807301 PMCID: PMC6885688 DOI: 10.1093/ckj/sfz106] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Indexed: 12/11/2022] Open
Abstract
Chronic non-communicable diseases have become a pandemic public problem in the 21st century, causing enormous burden on the economy, health and quality of life of societies. The role of a chronic inflammatory state in the pathogenesis of chronic disease has been more comprehensively recognized by recent findings. The new paradigm ‘metaflammation’ focuses on metabolism-induced (high fat or fructose-based diet or excessive calorie intake) chronic inflammation. There is a close correlation between the increased incidence of chronic kidney disease (CKD) and chronic heart failure with both increased inflammatory marker levels and western-type diet. In this review we describe the concept of metaflammation, its role in the development of CKD and chronic heart disease, the molecular and signalling pathways involved and the therapeutic consequences.
Collapse
Affiliation(s)
- Mehmet Kanbay
- Department of Medicine, Division of Nephrology, Koc University School of Medicine, Istanbul, Turkey
| | - Aslihan Yerlikaya
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Alan A Sag
- Department of Radiology, Division of Vascular and Interventional Radiology, Duke University Medical Center, Durham, NC, USA
| | - Alberto Ortiz
- Dialysis Unit, School of Medicine, IIS-Fundacion Jimenez Diaz, Universidad Autónoma de Madrid, Madrid, Spain
| | | | - Adrian Covic
- Nephrology Department, Dialysis and Renal Transplant Center, "Dr. C.I. Parhon" University Hospital, "Grigore T. Popa" University of Medicine and Pharmacy, Iasi, Romania
| | - Andrzej Wiecek
- Department of Nephrology, Transplantation and Internal Medicine, Medical University of Silesia, Katowice, Poland
| | - Peter Stenvinkel
- Department of Clinical Science Intervention and Technology, Division of Renal Medicine and Baxter Novum, Karolinska Institutet, Stockholm, Sweden
| | - Baris Afsar
- Department of Medicine, Division of Nephrology, Suleyman Demirel University School of Medicine, Isparta, Turkey
| |
Collapse
|
158
|
Lange S, Banerjee I, Carrion K, Serrano R, Habich L, Kameny R, Lengenfelder L, Dalton N, Meili R, Börgeson E, Peterson K, Ricci M, Lincoln J, Ghassemian M, Fineman J, del Álamo JC, Nigam V. miR-486 is modulated by stretch and increases ventricular growth. JCI Insight 2019; 4:125507. [PMID: 31513548 PMCID: PMC6795397 DOI: 10.1172/jci.insight.125507] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 09/04/2019] [Indexed: 12/24/2022] Open
Abstract
Perturbations in biomechanical stimuli during cardiac development contribute to congenital cardiac defects such as hypoplastic left heart syndrome (HLHS). This study sought to identify stretch-responsive pathways involved in cardiac development. miRNA-Seq identified miR-486 as being increased in cardiomyocytes exposed to cyclic stretch in vitro. The right ventricles (RVs) of patients with HLHS experienced increased stretch and had a trend toward higher miR-486 levels. Sheep RVs dilated from excessive pulmonary blood flow had 60% more miR-486 compared with control RVs. The left ventricles of newborn mice treated with miR-486 mimic were 16.9%-24.6% larger and displayed a 2.48-fold increase in cardiomyocyte proliferation. miR-486 treatment decreased FoxO1 and Smad signaling while increasing the protein levels of Stat1. Stat1 associated with Gata-4 and serum response factor (Srf), 2 key cardiac transcription factors with protein levels that increase in response to miR-486. This is the first report to our knowledge of a stretch-responsive miRNA that increases the growth of the ventricle in vivo.
Collapse
Affiliation(s)
- Stephan Lange
- Division of Cardiovascular Medicine, Department of Medicine, UCSD School of Medicine, San Diego, California, USA
- Institute of Medicine, Department of Molecular and Clinical Medicine, the Wallenberg Laboratory and Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Indroneal Banerjee
- Division of Cardiovascular Medicine, Department of Medicine, UCSD School of Medicine, San Diego, California, USA
| | - Katrina Carrion
- Division of Cardiology, Department of Pediatrics, UCSD School of Medicine, San Diego, California, USA
| | - Ricardo Serrano
- Department of Mechanical and Aerospace Engineering, UCSD, San Diego, USA
| | - Louisa Habich
- Division of Cardiovascular Medicine, Department of Medicine, UCSD School of Medicine, San Diego, California, USA
| | - Rebecca Kameny
- Department of Pediatrics, UCSF School of Medicine, San Francisco, USA
| | - Luisa Lengenfelder
- Division of Cardiovascular Medicine, Department of Medicine, UCSD School of Medicine, San Diego, California, USA
| | - Nancy Dalton
- Division of Cardiovascular Medicine, Department of Medicine, UCSD School of Medicine, San Diego, California, USA
| | - Rudolph Meili
- Department of Mechanical and Aerospace Engineering, UCSD, San Diego, USA
| | - Emma Börgeson
- Institute of Medicine, Department of Molecular and Clinical Medicine, the Wallenberg Laboratory and Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Kirk Peterson
- Division of Cardiovascular Medicine, Department of Medicine, UCSD School of Medicine, San Diego, California, USA
| | - Marco Ricci
- Division of Cardiothoracic Surgery and
- Division of Pediatric Surgery, Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Joy Lincoln
- Center for Cardiovascular Research, Nationwide Children’s Hospital, Columbus, Ohio, USA
| | | | - Jeffery Fineman
- Department of Pediatrics, UCSF School of Medicine, San Francisco, USA
| | - Juan C. del Álamo
- Department of Mechanical and Aerospace Engineering, UCSD, San Diego, USA
| | - Vishal Nigam
- Division of Cardiology, Department of Pediatrics, UCSD School of Medicine, San Diego, California, USA
- Division of Cardiology, Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington, USA
- Seattle Children’s Research Institute, Seattle, Washington, USA
| |
Collapse
|
159
|
Poon ENY, Hao B, Guan D, Jun Li M, Lu J, Yang Y, Wu B, Wu SCM, Webb SE, Liang Y, Miller AL, Yao X, Wang J, Yan B, Boheler KR. Integrated transcriptomic and regulatory network analyses identify microRNA-200c as a novel repressor of human pluripotent stem cell-derived cardiomyocyte differentiation and maturation. Cardiovasc Res 2019; 114:894-906. [PMID: 29373717 DOI: 10.1093/cvr/cvy019] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 01/22/2018] [Indexed: 11/12/2022] Open
Abstract
Aims MicroRNAs (miRNAs) are crucial for the post-transcriptional control of protein-encoding genes and together with transcription factors (TFs) regulate gene expression; however, the regulatory activities of miRNAs during cardiac development are only partially understood. In this study, we tested the hypothesis that integrative computational approaches could identify miRNAs that experimentally could be shown to regulate cardiomyogenesis. Methods and results We integrated expression profiles with bioinformatics analyses of miRNA and TF regulatory programs to identify candidate miRNAs involved with cardiac development. Expression profiling showed that miR-200c, which is not normally detected in adult heart, is progressively down-regulated both during cardiac development and in vitro differentiation of human embryonic stem cells (hESCs) to cardiomyocytes (CMs). We employed computational methodologies to predict target genes of both miR-200c and five key cardiac TFs to identify co-regulated gene networks. The inferred cardiac networks revealed that the cooperative action of miR-200c with these five key TFs, including three (GATA4, SRF and TBX5) targeted by miR-200c, should modulate key processes and pathways necessary for CM development and function. Experimentally, over-expression (OE) of miR-200c in hESC-CMs reduced the mRNA levels of GATA4, SRF and TBX5. Cardiac expression of Ca2+, K+ and Na+ ion channel genes (CACNA1C, KCNJ2 and SCN5A) were also significantly altered by knockdown or OE of miR-200c. Luciferase reporter assays validated miR-200c binding sites on the 3' untranslated region of CACNA1C. In hESC-CMs, elevated miR-200c increased beating frequency, and repressed both Ca2+ influx, mediated by the L-type Ca2+ channel and Ca2+ transients. Conclusions Our analyses demonstrate that miR-200c represses hESC-CM differentiation and maturation. The integrative computation and experimental approaches described here, when applied more broadly, will enhance our understanding of the interplays between miRNAs and TFs in controlling cardiac development and disease processes.
Collapse
Affiliation(s)
- Ellen Ngar-Yun Poon
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China.,Department of Paediatrics and Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Baixia Hao
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Daogang Guan
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Mulin Jun Li
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.,Centre of Genomics Sciences, LKS Faculty of Medicine, The University of Hong Kong. Hong Kong, China
| | - Jun Lu
- School of Biomedical Sciences, LSK Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Yong Yang
- Laboratory for Food Safety and Environmental Technology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Ave, Shenzhen, Guangdong 518055, China
| | - Binbin Wu
- Laboratory for Food Safety and Environmental Technology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Ave, Shenzhen, Guangdong 518055, China
| | - Stanley Chun-Ming Wu
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Sarah E Webb
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Yan Liang
- Laboratory for Food Safety and Environmental Technology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Ave, Shenzhen, Guangdong 518055, China
| | - Andrew L Miller
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China.,Marine Biology Laboratory, Woods Hole, MA 02543, USA
| | - Xiaoqiang Yao
- School of Biomedical Sciences, LSK Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Junwen Wang
- Centre of Genomics Sciences, LKS Faculty of Medicine, The University of Hong Kong. Hong Kong, China.,Center for Individualized Medicine, Department of Health Sciences Research, Mayo Clinic, Scottsdale, AZ 85259, USA and Department of Biomedical Informatics, Arizona State University, Scottsdale, AZ 85259, USA
| | - Bin Yan
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China.,Centre of Genomics Sciences, LKS Faculty of Medicine, The University of Hong Kong. Hong Kong, China.,Laboratory for Food Safety and Environmental Technology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Ave, Shenzhen, Guangdong 518055, China
| | - Kenneth R Boheler
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China.,Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
160
|
Li H, Wan HQ, Zhao HJ, Luan SX, Zhang CG. Identification of candidate genes and miRNAs associated with neuropathic pain induced by spared nerve injury. Int J Mol Med 2019; 44:1205-1218. [PMID: 31432094 PMCID: PMC6713433 DOI: 10.3892/ijmm.2019.4305] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 05/22/2019] [Indexed: 12/16/2022] Open
Abstract
Neuropathic pain (NP) is a complex, chronic pain condition caused by injury or dysfunction affecting the somatosensory nervous system. This study aimed to identify crucial genes and miRNAs involved in NP. Microarray data (access number GSE91396) were downloaded from the Gene Expression Omnibus (GEO). Murine RNA-seq samples from three brain regions [nucleus accumbens, (NAc); medial prefrontal cortex, (mPFC) and periaqueductal gray, (PAG)] were compared between the spared nerve injury (SNI) model and a sham surgery. After data normalization, differentially expressed RNAs were screened using the limma package and functional enrichment analysis was performed with Database for Annotation, Visualization and Integrated Discovery. The microRNA (miRNA/miR)-mRNA regulatory network and miRNA-target gene-pathway regulatory network were constructed using Cytoscape software. A total of 2,776 differentially expressed RNAs (219 miRNAs and 2,557 mRNAs) were identified in the SNI model compared with the sham surgery group. A total of two important modules (red and turquoise module) were found to be related to NP using weighed gene co-expression network analysis (WGCNA) for the 2,325 common differentially expressed RNAs in three brain regions. The differentially expressed genes (DEGs) in the miRNA-mRNA regulatory network were significantly enriched in 21 Gene Ontology terms and five pathways. A total of four important DEGs (CXCR2, IL12B, TNFSF8 and GRK1) and five miRNAs (miR-208a-5p, miR-7688-3p, miR-344f-3p, miR-135b-3p and miR-135a-2-3p) were revealed according to the miRNA-target gene-pathway regulatory network to be related to NP. Four important DEGs (CXCR2, IL12B, TNFSF8 and GRK1) and five miRNAs (miR-208a-5p, miR-7688-3p, miR-344f-3p, miR-135b-3p and miR-135a-2-3p) were differentially expressed in SNI, indicating their plausible roles in NP pathogenesis.
Collapse
Affiliation(s)
- He Li
- Department of Pain Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Hong-Quan Wan
- Department of Mental Health, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Hai-Jun Zhao
- Department of Pain Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Shu-Xin Luan
- Department of Mental Health, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Chun-Guo Zhang
- Department of Pain Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
161
|
Zou JB, Chai HB, Zhang XF, Guo DY, Tai J, Wang Y, Liang YL, Wang F, Cheng JX, Wang J, Shi YJ. Reconstruction of the lncRNA-miRNA-mRNA network based on competitive endogenous RNA reveal functional lncRNAs in Cerebral Infarction. Sci Rep 2019; 9:12176. [PMID: 31434962 PMCID: PMC6704173 DOI: 10.1038/s41598-019-48435-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 08/06/2019] [Indexed: 02/06/2023] Open
Abstract
Functioning as miRNA sponges, long non-coding RNA (lncRNA) exert its pharmacological action via regulating expression of protein-coding genes. However, the lncRNA-mediated ceRNA in cerebral Infarction (CI) remains unclear. In this study, the expression recordsets of mRNA, lncRNA and miRNA of CI samples were obtained from the NCBI GEO datasets separately. The differentially expressed lncRNAs (DELs), miRNAs (DEMis) and mRNAs (DEMs) were identified by limma package in R platform. A total of 267 DELs, 26 DEMis, and 760 DEMs were identified as differentially expressed profiles, with which we constructed the ceRNA network composed of DELs-DEMis-DEMs. Further, clusterProfiler package in R platform is employed for performing Gene Ontology (GO) and KEGG pathway analysis. An aberrant ceRNA network was constructed according to node degrees in CI, including 28 DELs, 19 DEMs and 12 DEMis, from which we extracted the core network, in which 9 nodes were recognized as kernel genes including Tspan3, Eif4a2, rno-miR-208a-3p, rno-miR-194-5p, Pdpn, H3f3b, Stat3, Cd63 and Sdc4. Finally, with the DELs-DEMis-DEMs ceRNA network provided above, we can improve our understanding of the pathogenesis of CI mediated by lncRNA.
Collapse
Affiliation(s)
- Jun-Bo Zou
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research,Pharmacy College, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Hong-Bo Chai
- The first affiliated Hospital of Hunan University of Medicine, Huaihua, 410007, China
| | - Xiao-Fei Zhang
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research,Pharmacy College, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Dong-Yan Guo
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research,Pharmacy College, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Jia Tai
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research,Pharmacy College, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Yu Wang
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research,Pharmacy College, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Yu-Lin Liang
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research,Pharmacy College, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Fang Wang
- Key laboratory of Modern Prepararation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330000, China
| | - Jiang-Xue Cheng
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research,Pharmacy College, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Jing Wang
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research,Pharmacy College, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Ya-Jun Shi
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research,Pharmacy College, Shaanxi University of Chinese Medicine, Xianyang, 712046, China.
| |
Collapse
|
162
|
Wu MJ, Andreasson JOL, Kladwang W, Greenleaf W, Das R. Automated Design of Diverse Stand-Alone Riboswitches. ACS Synth Biol 2019; 8:1838-1846. [PMID: 31298841 PMCID: PMC6703183 DOI: 10.1021/acssynbio.9b00142] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
![]()
Riboswitches that couple binding
of ligands to conformational changes
offer sensors and control elements for RNA synthetic biology and medical
biotechnology. However, design of these riboswitches has required
expert intuition or software specialized to transcription or translation
outputs; design has been particularly challenging for applications
in which the riboswitch output cannot be amplified by other molecular
machinery. We present a fully automated design method called RiboLogic
for such “stand-alone” riboswitches and test it via high-throughput experiments on 2875 molecules using
RNA-MaP (RNA on a massively parallel array) technology. These molecules
consistently modulate their affinity to the MS2 bacteriophage coat
protein upon binding of flavin mononucleotide, tryptophan, theophylline,
and microRNA miR-208a, achieving activation ratios of up to 20 and
significantly better performance than control designs. By encompassing
a wide diversity of stand-alone switches and highly quantitative data,
the resulting ribologic-solves experimental data
set provides a rich resource for further improvement of riboswitch
models and design methods.
Collapse
|
163
|
Zaleta-Rivera K, Dainis A, Ribeiro AJS, Cordero P, Rubio G, Shang C, Liu J, Finsterbach T, Parikh VN, Sutton S, Seo K, Sinha N, Jain N, Huang Y, Hajjar RJ, Kay MA, Szczesna-Cordary D, Pruitt BL, Wheeler MT, Ashley EA. Allele-Specific Silencing Ameliorates Restrictive Cardiomyopathy Attributable to a Human Myosin Regulatory Light Chain Mutation. Circulation 2019; 140:765-778. [PMID: 31315475 DOI: 10.1161/circulationaha.118.036965] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Restrictive cardiomyopathy is a rare heart disease associated with mutations in sarcomeric genes and with phenotypic overlap with hypertrophic cardiomyopathy. There is no approved therapy directed at the underlying cause. Here, we explore the potential of an interfering RNA (RNAi) therapeutic for a human sarcomeric mutation in MYL2 causative of restrictive cardiomyopathy in a mouse model. METHODS A short hairpin RNA (M7.8L) was selected from a pool for specificity and efficacy. Two groups of myosin regulatory light chain N47K transgenic mice were injected with M7.8L packaged in adeno-associated virus 9 at 3 days of age and 60 days of age. Mice were subjected to treadmill exercise and echocardiography after treatment to determine maximal oxygen uptake and left ventricular mass. At the end of treatment, heart, lung, liver, and kidney tissue was harvested to determine viral tropism and for transcriptomic and proteomic analysis. Cardiomyocytes were isolated for single-cell studies. RESULTS A one-time injection of AAV9-M7.8L RNAi in 3-day-old humanized regulatory light chain mutant transgenic mice silenced the mutated allele (RLC-47K) with minimal effects on the normal allele (RLC-47N) assayed at 16 weeks postinjection. AAV9-M7.8L RNAi suppressed the expression of hypertrophic biomarkers, reduced heart weight, and attenuated a pathological increase in left ventricular mass. Single adult cardiac myocytes from mice treated with AAV9-M7.8L showed partial restoration of contraction, relaxation, and calcium kinetics. In addition, cardiac stress protein biomarkers, such as calmodulin-dependent protein kinase II and the transcription activator Brg1 were reduced, suggesting recovery toward a healthy myocardium. Transcriptome analyses further revealed no significant changes of argonaute (AGO1, AGO2) and endoribonuclease dicer (DICER1) transcripts, and endogenous microRNAs were preserved, suggesting that the RNAi pathway was not saturated. CONCLUSIONS Our results show the feasibility, efficacy, and safety of RNAi therapeutics directed towards human restrictive cardiomyopathy. This is a promising step toward targeted therapy for a prevalent human disease.
Collapse
Affiliation(s)
- Kathia Zaleta-Rivera
- Division of Cardiovascular Medicine (K.Z.-R., A.D., P.C., G.R., C.S., J.L., T.F., W.N.P., S.S., K.S., N.S., N.J., Y.H., M.T.W., E.A.A.), Stanford University School of Medicine, CA
| | - Alexandra Dainis
- Division of Cardiovascular Medicine (K.Z.-R., A.D., P.C., G.R., C.S., J.L., T.F., W.N.P., S.S., K.S., N.S., N.J., Y.H., M.T.W., E.A.A.), Stanford University School of Medicine, CA
| | | | - Pablo Cordero
- Division of Cardiovascular Medicine (K.Z.-R., A.D., P.C., G.R., C.S., J.L., T.F., W.N.P., S.S., K.S., N.S., N.J., Y.H., M.T.W., E.A.A.), Stanford University School of Medicine, CA
| | - Gabriel Rubio
- Division of Cardiovascular Medicine (K.Z.-R., A.D., P.C., G.R., C.S., J.L., T.F., W.N.P., S.S., K.S., N.S., N.J., Y.H., M.T.W., E.A.A.), Stanford University School of Medicine, CA
| | - Ching Shang
- Division of Cardiovascular Medicine (K.Z.-R., A.D., P.C., G.R., C.S., J.L., T.F., W.N.P., S.S., K.S., N.S., N.J., Y.H., M.T.W., E.A.A.), Stanford University School of Medicine, CA
| | - Jing Liu
- Division of Cardiovascular Medicine (K.Z.-R., A.D., P.C., G.R., C.S., J.L., T.F., W.N.P., S.S., K.S., N.S., N.J., Y.H., M.T.W., E.A.A.), Stanford University School of Medicine, CA
| | - Thomas Finsterbach
- Division of Cardiovascular Medicine (K.Z.-R., A.D., P.C., G.R., C.S., J.L., T.F., W.N.P., S.S., K.S., N.S., N.J., Y.H., M.T.W., E.A.A.), Stanford University School of Medicine, CA
| | - Victoria N Parikh
- Division of Cardiovascular Medicine (K.Z.-R., A.D., P.C., G.R., C.S., J.L., T.F., W.N.P., S.S., K.S., N.S., N.J., Y.H., M.T.W., E.A.A.), Stanford University School of Medicine, CA
| | - Shirley Sutton
- Division of Cardiovascular Medicine (K.Z.-R., A.D., P.C., G.R., C.S., J.L., T.F., W.N.P., S.S., K.S., N.S., N.J., Y.H., M.T.W., E.A.A.), Stanford University School of Medicine, CA
| | - Kinya Seo
- Division of Cardiovascular Medicine (K.Z.-R., A.D., P.C., G.R., C.S., J.L., T.F., W.N.P., S.S., K.S., N.S., N.J., Y.H., M.T.W., E.A.A.), Stanford University School of Medicine, CA
| | - Nikita Sinha
- Division of Cardiovascular Medicine (K.Z.-R., A.D., P.C., G.R., C.S., J.L., T.F., W.N.P., S.S., K.S., N.S., N.J., Y.H., M.T.W., E.A.A.), Stanford University School of Medicine, CA
| | - Nikhil Jain
- Division of Cardiovascular Medicine (K.Z.-R., A.D., P.C., G.R., C.S., J.L., T.F., W.N.P., S.S., K.S., N.S., N.J., Y.H., M.T.W., E.A.A.), Stanford University School of Medicine, CA
| | - Yong Huang
- Division of Cardiovascular Medicine (K.Z.-R., A.D., P.C., G.R., C.S., J.L., T.F., W.N.P., S.S., K.S., N.S., N.J., Y.H., M.T.W., E.A.A.), Stanford University School of Medicine, CA
| | - Roger J Hajjar
- Cardiovascular Institute, Cardiovascular Research Center at Icahn School of Medicine at Mount Sinai, New York, NY (R.J.H.)
| | - Mark A Kay
- Department of Genetics (M.A.K., E.A.A.), Stanford University School of Medicine, CA
- Department of Pediatrics (M.A.K.), Stanford University School of Medicine, CA
| | - Danuta Szczesna-Cordary
- Department of Molecular and Cellular Pharmacology, University of Miami Leonard M. Miller School of Medicine, FL (D.S.-C.)
| | - Beth L Pruitt
- Department of Mechanical Engineering, Stanford University, CA (A.J.S.R., B.L.P.)
| | - Matthew T Wheeler
- Division of Cardiovascular Medicine (K.Z.-R., A.D., P.C., G.R., C.S., J.L., T.F., W.N.P., S.S., K.S., N.S., N.J., Y.H., M.T.W., E.A.A.), Stanford University School of Medicine, CA
| | - Euan A Ashley
- Division of Cardiovascular Medicine (K.Z.-R., A.D., P.C., G.R., C.S., J.L., T.F., W.N.P., S.S., K.S., N.S., N.J., Y.H., M.T.W., E.A.A.), Stanford University School of Medicine, CA
- Department of Genetics (M.A.K., E.A.A.), Stanford University School of Medicine, CA
| |
Collapse
|
164
|
MicroRNAs: Emerging biomarkers for atrial fibrillation. J Cardiol 2019; 74:475-482. [PMID: 31324570 DOI: 10.1016/j.jjcc.2019.05.018] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 05/20/2019] [Accepted: 05/20/2019] [Indexed: 12/15/2022]
Abstract
Atrial fibrillation (AF) causes severe cardiac dysrhythmia among patients with cardiovascular diseases. AF increases the risk of stroke and heart failure and is a growing public health concern. AF is also associated with various disease conditions such as hypertension, coronary artery disease, aging, and diabetes mellitus. The mechanism underlying AF is not completely understood due to its complexity. However, experimental and clinical data have revealed that the prevalence of this disease is associated with atrial arrhythmogenic remodeling. Currently, there are no biomarkers that are available for the early diagnosis of AF. Several studies have proposed microRNAs (miRNAs) as useful biomarkers for the diagnosis of AF due to their stability and easy availability both in atrial tissue and circulating blood. miRNAs play an important role in the development of the heart. The dysregulation of miRNA expression is associated with cardiac remodeling. Genetic factors strongly contribute to the pathogenesis of AF. Recently, single nucleotide polymorphisms (SNPs) in various genes and miRNAs have been reported to be associated with AF. The aim of this review was to discuss the correlation between SNPs in miRNAs and AF, including those miRNAs that are commonly reported as potential biomarkers for AF.
Collapse
|
165
|
Zhang Y, Wang Y, Yanni J, Qureshi MA, Logantha SJRJ, Kassab S, Boyett MR, Gardiner NJ, Sun H, Howarth FC, Dobrzynski H. Electrical Conduction System Remodeling in Streptozotocin-Induced Diabetes Mellitus Rat Heart. Front Physiol 2019; 10:826. [PMID: 31338036 PMCID: PMC6628866 DOI: 10.3389/fphys.2019.00826] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 06/13/2019] [Indexed: 12/15/2022] Open
Abstract
Cardiovascular complications are common in type 1 diabetes mellitus (TIDM) and there is an increased risk of arrhythmias as a result of dysfunction of the cardiac conduction system (CCS). We have previously shown that, in vivo, there is a decrease in the heart rate and prolongation of the QRS complex in streptozotocin-induced type 1 diabetic rats indicating dysfunction of the CCS. The aim of this study was to investigate the function of the ex vivo CCS and key proteins that are involved in pacemaker mechanisms in TIDM. RR interval, PR interval and QRS complex duration were significantly increased in diabetic rats. The beating rate of the isolated sinoatrial node (SAN) preparation was significantly decreased in diabetic rats. The funny current density and cell capacitance were significantly decreased in diabetic nodal cells. Western blot showed that proteins involved in the function of the CCS were significantly decreased in diabetic rats, namely: HCN4, Cav1.3, Cav3.1, Cx45, and NCX1 in the SAN; RyR2 and NCX1 in the atrioventricular junction and Cx40, Cx43, Cx45, and RyR2 in the Purkinje network. We conclude that there are complex functional and cellular changes in the CCS in TIDM. The changes in the proteins involved in the function of this electrical system are expected to adversely affect action potential generation and propagation, and these changes are likely to be arrhythmogenic.
Collapse
Affiliation(s)
- Yu Zhang
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester, United Kingdom.,Department of Physiology, Xuzhou Medical University, Xuzhou, China
| | - Yanwen Wang
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester, United Kingdom
| | - Joseph Yanni
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester, United Kingdom
| | - Mohammed Anwar Qureshi
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Sunil Jit R J Logantha
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester, United Kingdom
| | - Sarah Kassab
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester, United Kingdom
| | - Mark R Boyett
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester, United Kingdom
| | - Natalie J Gardiner
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester, United Kingdom
| | - Hong Sun
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
| | - Frank Christopher Howarth
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Halina Dobrzynski
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
166
|
Filardi T, Ghinassi B, Di Baldassarre A, Tanzilli G, Morano S, Lenzi A, Basili S, Crescioli C. Cardiomyopathy Associated with Diabetes: The Central Role of the Cardiomyocyte. Int J Mol Sci 2019; 20:ijms20133299. [PMID: 31284374 PMCID: PMC6651183 DOI: 10.3390/ijms20133299] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 07/02/2019] [Accepted: 07/03/2019] [Indexed: 12/18/2022] Open
Abstract
The term diabetic cardiomyopathy (DCM) labels an abnormal cardiac structure and performance due to intrinsic heart muscle malfunction, independently of other vascular co-morbidity. DCM, accounting for 50%–80% of deaths in diabetic patients, represents a worldwide problem for human health and related economics. Optimal glycemic control is not sufficient to prevent DCM, which derives from heart remodeling and geometrical changes, with both consequences of critical events initially occurring at the cardiomyocyte level. Cardiac cells, under hyperglycemia, very early undergo metabolic abnormalities and contribute to T helper (Th)-driven inflammatory perturbation, behaving as immunoactive units capable of releasing critical biomediators, such as cytokines and chemokines. This paper aims to focus onto the role of cardiomyocytes, no longer considered as “passive” targets but as “active” units participating in the inflammatory dialogue between local and systemic counterparts underlying DCM development and maintenance. Some of the main biomolecular/metabolic/inflammatory processes triggered within cardiac cells by high glucose are overviewed; particular attention is addressed to early inflammatory cytokines and chemokines, representing potential therapeutic targets for a prompt early intervention when no signs or symptoms of DCM are manifesting yet. DCM clinical management still represents a challenge and further translational investigations, including studies at female/male cell level, are warranted.
Collapse
Affiliation(s)
- Tiziana Filardi
- Department of Experimental Medicine, "Sapienza" University, Viale del Policlinico 155, 00161 Rome, Italy
| | - Barbara Ghinassi
- Department of Medicine and Aging Sciences, "G. D'Annunzio" University of Chieti and Pescara, Via dei Vestini 31, 66100 Chieti, Italy
| | - Angela Di Baldassarre
- Department of Medicine and Aging Sciences, "G. D'Annunzio" University of Chieti and Pescara, Via dei Vestini 31, 66100 Chieti, Italy
| | - Gaetano Tanzilli
- Department of Cardiovascular Sciences, "Sapienza" University, Viale del Policlinico 155, 00161 Rome, Italy
| | - Susanna Morano
- Department of Experimental Medicine, "Sapienza" University, Viale del Policlinico 155, 00161 Rome, Italy
| | - Andrea Lenzi
- Department of Experimental Medicine, "Sapienza" University, Viale del Policlinico 155, 00161 Rome, Italy
| | - Stefania Basili
- Department of Translational and Precision Medicine, "Sapienza" University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Clara Crescioli
- Department of Movement, Human and Health Sciences, Section of Health Sciences, University of Rome "Foro Italico", Piazza L. de Bosis 6, 00135 Rome, Italy.
| |
Collapse
|
167
|
Xiao Y, Zhao J, Tuazon JP, Borlongan CV, Yu G. MicroRNA-133a and Myocardial Infarction. Cell Transplant 2019; 28:831-838. [PMID: 30983393 PMCID: PMC6719504 DOI: 10.1177/0963689719843806] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 02/27/2019] [Accepted: 03/12/2019] [Indexed: 12/12/2022] Open
Abstract
Myocardial infarction (MI) is the leading cause of morbidity and mortality in the world. The infarcted heart displays typical cell death cascades characterized by a loss of cells and fibrotic scarring in the myocardium. Cardiac hypertrophy and fibrosis largely contribute to ventricular wall thickening and stiffening, altogether defining an adverse cardiac remodeling that ultimately leads to impaired cardiac function and subsequent heart failure. Finding a strategy to promote therapeutic, instead of detrimental, cardiac remodeling may pose as a potent MI treatment. Accumulating evidence shows that microRNAs (miRNAs) may play an essential role in cardiovascular diseases. In particular, microRNA-133a (miR-133a) is one of the most abundant miRNAs in the heart. Multiple studies have demonstrated that miR-133a participates in the early pathology of MI, as well as in subsequent cardiac remodeling. In this review, we summarize recent research progress highlighting the regulatory effects of miR-133a in ischemic myocardial diseases, such as inhibiting angiogenesis, apoptosis, fibrosis, hypertrophy, and inflammation, while promoting therapeutic cardiac remodeling. The goal is to elicit a critical discussion on the translational direction of miRNA-mediated treatments towards a safe and effective MI therapy.
Collapse
Affiliation(s)
- Yi Xiao
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, PR China
| | - Jiling Zhao
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, PR China
| | - Julian P. Tuazon
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Cesar V. Borlongan
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Guolong Yu
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, PR China
| |
Collapse
|
168
|
Shen NN, Zhang ZL, Li Z, Zhang C, Li H, Wang JL, Wang J, Gu ZC. Identification of microRNA biomarkers in atrial fibrillation: A protocol for systematic review and bioinformatics analysis. Medicine (Baltimore) 2019; 98:e16538. [PMID: 31348272 PMCID: PMC6708903 DOI: 10.1097/md.0000000000016538] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Atrial fibrillation (AF) is recognized as the most prevalent arrhythmia, and its subsequently serious complications of heart failure and thromboembolism always raise the social attention. To date, the molecular pathogenesis of AF has largely remained unclear. Publications of contemporary studies have evaluated individual miRNAs expression signatures for AF, and findings of different studies are inconsistent and not all miRNAs reported are actually important in the pathogenesis of AF. METHODS Medline, Embase, and Cochrane Library databases will be comprehensively searched (up to April 30, 2019) for studies identifying miRNA expression profiling in subjects with and without AF. Log10 odds ratios (logORs) and associated 95% confidence interval (95%CI) will be calculated using random-effects models. Subgroup analysis will be performed according to miRNA detecting methods, species, sample types, and ethnicities. Sensitivity analysis will be conducted to detect the robustness of the findings. The methodological quality of studies will be independently assessed using criteria adopted from the Quality Assessment of Diagnostic Accuracy Studies (QUADAS-2). Furthermore, bioinformatics analysis will be performed to identify the potential target genes in AF and the corresponding pathways of dysregulated miRNAs. Two reviewers will independently screen potential studies and extract data in a structured eligibility items, with any disagreements being resolved by consensus. RESULTS The present systematic review will identify potential biomarkers by pooling all differentially expressed miRNAs in AF studies, as well as to predict miRNA-target interactions and to identify the potential biometric functions using bioinformatics analysis. CONCLUSIONS This systematic review and bioinformatics analysis will identify several miRNAs as potential biomarkers for AF, and explore the biological pathways regulated by the eligible miRNAs. PROSPERO REGISTRATION NUMBER CRD42019127594.
Collapse
Affiliation(s)
- Nan-Nan Shen
- Department of Pharmacy
- Department of Geriatrics, Affiliated Hospital of Shaoxing University, Shao Xing, Zhejiang Province
- Department of Pharmacy
| | | | - Zheng Li
- Department of Cardiology, Renji Hospital
| | | | - Hao Li
- Department of Pharmacy, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Jia-Liang Wang
- Department of Pharmacy
- Department of Geriatrics, Affiliated Hospital of Shaoxing University, Shao Xing, Zhejiang Province
| | - Jun Wang
- Department of Pharmacy
- Department of Geriatrics, Affiliated Hospital of Shaoxing University, Shao Xing, Zhejiang Province
| | | |
Collapse
|
169
|
Di Mauro V, Crasto S, Colombo FS, Di Pasquale E, Catalucci D. Wnt signalling mediates miR-133a nuclear re-localization for the transcriptional control of Dnmt3b in cardiac cells. Sci Rep 2019; 9:9320. [PMID: 31249372 PMCID: PMC6597717 DOI: 10.1038/s41598-019-45818-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 06/14/2019] [Indexed: 12/14/2022] Open
Abstract
MiR-133a is a muscle-enriched miRNA, which plays a key role for proper skeletal and cardiac muscle function via regulation of transduction cascades, including the Wnt signalling. MiR-133a modulates its targets via canonical mRNA repression, a process that has been largely demonstrated to occur within the cytoplasm. However, recent evidence has shown that miRNAs play additional roles in other sub-cellular compartments, such as nuclei. Here, we show that miR-133a translocates to the nucleus of cardiac cells following inactivation of the canonical Wnt pathway. The nuclear miR-133a/AGO2 complex binds to a complementary miR-133a target site within the promoter of the de novo DNA methyltransferase 3B (Dnmt3b) gene, leading to its transcriptional repression, which is mediated by DNMT3B itself. Altogether, these data show an unconventional role of miR-133a that upon its relocalization to the nucleus is responsible for epigenetic repression of its target gene Dnmt3b via a DNMT3B self-regulatory negative feedback loop.
Collapse
Affiliation(s)
- Vittoria Di Mauro
- University of Milan Bicocca, Piazza dell'Ateneo Nuovo 1, 20126, Milan, Italy
- CNR-IRGB UOS Milan, Via Fantoli 15/16, 20138, Milan, Italy
- Humanitas Clinical and Research Center, via Alessandro Manzoni 113, 20089, Rozzano, Milan, Italy
| | - Silvia Crasto
- CNR-IRGB UOS Milan, Via Fantoli 15/16, 20138, Milan, Italy
- Humanitas Clinical and Research Center, via Alessandro Manzoni 113, 20089, Rozzano, Milan, Italy
| | - Federico Simone Colombo
- Humanitas Clinical and Research Center, via Alessandro Manzoni 113, 20089, Rozzano, Milan, Italy
| | - Elisa Di Pasquale
- CNR-IRGB UOS Milan, Via Fantoli 15/16, 20138, Milan, Italy
- Humanitas Clinical and Research Center, via Alessandro Manzoni 113, 20089, Rozzano, Milan, Italy
| | - Daniele Catalucci
- CNR-IRGB UOS Milan, Via Fantoli 15/16, 20138, Milan, Italy.
- Humanitas Clinical and Research Center, via Alessandro Manzoni 113, 20089, Rozzano, Milan, Italy.
| |
Collapse
|
170
|
MicroRNA-19a/b-3p protect the heart from hypertension-induced pathological cardiac hypertrophy through PDE5A. J Hypertens 2019; 36:1847-1857. [PMID: 29664809 PMCID: PMC6080882 DOI: 10.1097/hjh.0000000000001769] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
AIM PDE5A is a leading factor contributing to cGMP signaling and cardiac hypertrophy. However, microRNA-mediated posttranscriptional regulation of PDE5A has not been reported. The aim of this study is to screen the microRNAs that are able to regulate PDE5A and explore the function of the microRNAs in cardiac hypertrophy and remodeling. METHODS AND RESULTS Although miR-19a/b-3p (microRNA-19a-3p and microRNA-19b-3p) have been reported to be differentially expressed during cardiac hypertrophy, the direct targets and the functions of this microRNA family for regulation of cardiac hypertrophy have not yet been investigated. The present study identified some direct targets and the underlying functions of miR-19a/b-3p by using bioinformatics tools and gene manipulations within mouse neonatal cardiomyocytes. Transfection of miR-19a/b-3p down-regulated endogenous expressions of PDE5A at both mRNA and protein levels with real-time PCR and western blot. Luciferase reporter assays showed that PDE5A was a direct target of miR-19a/b-3p. In mouse models of cardiac hypertrophy, we found that miR-19a/b-3p was expressed in cardiomyocytes and that its expression was reduced in pressure overload-induced hypertrophic hearts. miR-19a/b-3p transgenic mice prevented the progress of cardiac hypertrophy and cardiac remodeling in response to angiotensin II infusion with echocardiographic assessment and pressure-volume relation analysis. CONCLUSION Our study elucidates that PDE5A is a novel direct target of miR-19a/b-3p, and demonstrates that antihypertrophic roles of the miR-19a/b-3p family in Ang II-induced hypertrophy and cardiac remodeling, suggests that endogenous miR-19a/b-3p might have clinical potential to suppress cardiac hypertrophy and heart failure.
Collapse
|
171
|
Zhang W, Xu W, Feng Y, Zhou X. Non-coding RNA involvement in the pathogenesis of diabetic cardiomyopathy. J Cell Mol Med 2019; 23:5859-5867. [PMID: 31240820 PMCID: PMC6714214 DOI: 10.1111/jcmm.14510] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 06/02/2019] [Accepted: 06/11/2019] [Indexed: 12/19/2022] Open
Abstract
In recent years, the incidence of diabetes has been increasing rapidly, which seriously endangers human health. Diabetic cardiomyopathy, an important cardiovascular complication of diabetes, is characterized by myocardial fibrosis, ventricular remodelling and cardiac dysfunction. It has been documented that mitochondrial dysfunction, oxidative stress, inflammatory response, autophagy, apoptosis, diabetic microangiopathy and myocardial fibrosis are implicated in the pathogenesis of diabetic cardiomyopathy. With the development of molecular biology technology, accumulating evidence demonstrates that non‐coding RNAs (ncRNAs) are critically involved in the molecular mechanisms of diabetic cardiomyopathy. In this review, we summarize the pathological roles of three types of ncRNAs (microRNA, long ncRNA and circular RNA) in the progression of diabetic cardiomyopathy, which may provide valuable insights into the pathogenesis of diabetic cardiovascular complications.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Cardiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Weiting Xu
- Department of Cardiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yu Feng
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiang Zhou
- Department of Cardiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
172
|
Nagy O, Baráth S, Ujfalusi A. The role of microRNAs in congenital heart disease. EJIFCC 2019; 30:165-178. [PMID: 31263391 PMCID: PMC6599193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Congenital heart diseases (CHDs) are the leading inherited cause of perinatal and infant mortality. CHD refers to structural anomalies of the heart and blood vessels that arise during cardiac development and represents a broad spectrum of malformations, including septal and valve defects, lesions affecting the outflow tract and ventricules. Advanced treatment strategies have greatly improved life expectancy and led to expanded population of adult patients with CHD. Thus, a better understanding of the pathogenesis and molecular mechanisms underlying CHDs is essential to improve the diagnosis and prognosis of patients. The etiology of CHD is largely unknown, genetic and environmental factors may contribute to the disease. In addition to the mutations affecting genomic DNA, epigenetic changes are being increasingly acknowledged as key factors in the development and progression of CHDs. The posttranscriptional regulation of gene expression by microRNAs (miRs) controls the highly complex multi-cell lineage process of cardiac tissue formation. In recent years, multiplex experimental models have provided evidence that changes in expression levels of miRs are associated with human cardiovascular disease, including CHD. The newly described correlations between miRs and heart development suggest the potential importance of miRs as diagnostic markers in human cardiovascular diseases. In the future, more intensive research is likely to be carried out to clarify their contribution to personalized management and treatment of CHD patients. In this paper, we discuss the current knowledge on the causative role of miRs in cardiac development and CHDs.
Collapse
Affiliation(s)
| | | | - Anikó Ujfalusi
- Corresponding author: Anikó Ujfalusi Department of Laboratory Medicine Faculty of Medicine University of Debrecen Nagyerdei krt. 98. Debrecen H-4032 Hungary Phone: +36 52 340 006 Fax: +36 52 417 631 E-mail:
| |
Collapse
|
173
|
Zhi Y, Xu C, Sui D, Du J, Xu F, Li Y. Effective Delivery of Hypertrophic miRNA Inhibitor by Cholesterol-Containing Nanocarriers for Preventing Pressure Overload Induced Cardiac Hypertrophy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1900023. [PMID: 31179215 PMCID: PMC6548964 DOI: 10.1002/advs.201900023] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 03/04/2019] [Indexed: 05/15/2023]
Abstract
Persistent cardiac hypertrophy causes heart failure and sudden death. Gene therapy is a promising intervention for this disease, but is limited by the lack of effective delivery systems. Herein, it is reported that CHO-PGEA (cholesterol (CHO)-terminated ethanolamine-aminated poly(glycidyl methacrylate)) can efficiently condense small RNAs into nanosystems for preventing cardiac hypertrophy. CHO-PGEA contains two features: 1) lipophilic cholesterol groups enhance transfection efficiency in cardiomyocytes, 2) abundant hydrophilic hydroxyl groups benefit biocompatibility. miR-182, which is known to downregulate forkhead box O3, is selected as an intervention target and can be blocked by synthetic small RNA inhibitor of miR-182 (miR-182-in). CHO-PGEA can efficiently deliver miR-182-in into hearts. In the mice with aortic coarctation, CHO-PEGA/miR-182-in significantly suppresses cardiac hypertrophy without organ injury. This work demonstrates that CHO-PGEA/miRNA nanosystems are very promising for RNA-based therapeutics to treat heart diseases.
Collapse
Affiliation(s)
- Ying Zhi
- Beijing Anzhen HospitalCapital Medical UniversityThe Key Laboratory of Remodeling‐Related Cardiovascular DiseasesMinistry of EducationBeijing Institute of Heart Lung and Blood Vessel DiseasesBeijing100029China
| | - Chen Xu
- Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology)Ministry of EducationBeijing Laboratory of Biomedical MaterialsBeijing Advanced Innovation Center for Soft Matter Science and EngineeringBeijing University of Chemical TechnologyBeijing100029China
| | - Dandan Sui
- Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology)Ministry of EducationBeijing Laboratory of Biomedical MaterialsBeijing Advanced Innovation Center for Soft Matter Science and EngineeringBeijing University of Chemical TechnologyBeijing100029China
| | - Jie Du
- Beijing Anzhen HospitalCapital Medical UniversityThe Key Laboratory of Remodeling‐Related Cardiovascular DiseasesMinistry of EducationBeijing Institute of Heart Lung and Blood Vessel DiseasesBeijing100029China
| | - Fu‐Jian Xu
- Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology)Ministry of EducationBeijing Laboratory of Biomedical MaterialsBeijing Advanced Innovation Center for Soft Matter Science and EngineeringBeijing University of Chemical TechnologyBeijing100029China
| | - Yulin Li
- Beijing Anzhen HospitalCapital Medical UniversityThe Key Laboratory of Remodeling‐Related Cardiovascular DiseasesMinistry of EducationBeijing Institute of Heart Lung and Blood Vessel DiseasesBeijing100029China
| |
Collapse
|
174
|
Saucerman JJ, Tan PM, Buchholz KS, McCulloch AD, Omens JH. Mechanical regulation of gene expression in cardiac myocytes and fibroblasts. Nat Rev Cardiol 2019; 16:361-378. [PMID: 30683889 PMCID: PMC6525041 DOI: 10.1038/s41569-019-0155-8] [Citation(s) in RCA: 140] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The intact heart undergoes complex and multiscale remodelling processes in response to altered mechanical cues. Remodelling of the myocardium is regulated by a combination of myocyte and non-myocyte responses to mechanosensitive pathways, which can alter gene expression and therefore function in these cells. Cellular mechanotransduction and its downstream effects on gene expression are initially compensatory mechanisms during adaptations to the altered mechanical environment, but under prolonged and abnormal loading conditions, they can become maladaptive, leading to impaired function and cardiac pathologies. In this Review, we summarize mechanoregulated pathways in cardiac myocytes and fibroblasts that lead to altered gene expression and cell remodelling under physiological and pathophysiological conditions. Developments in systems modelling of the networks that regulate gene expression in response to mechanical stimuli should improve integrative understanding of their roles in vivo and help to discover new combinations of drugs and device therapies targeting mechanosignalling in heart disease.
Collapse
Affiliation(s)
- Jeffrey J Saucerman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Philip M Tan
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Kyle S Buchholz
- Departments of Bioengineering and Medicine, University of California San Diego, La Jolla, CA, USA
| | - Andrew D McCulloch
- Departments of Bioengineering and Medicine, University of California San Diego, La Jolla, CA, USA.
| | - Jeffrey H Omens
- Departments of Bioengineering and Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
175
|
Abstract
Heritable cardiomyopathies are a class of heart diseases caused by variations in a number of genetic loci. Genetic variants on one allele lead to either a degraded protein, which causes a haploinsufficiency of that protein, or a nonfunctioning protein that subverts the molecular system within which the protein works. Over years, both of these mechanisms eventually lead to diseased heart tissue and symptoms of a failing heart. Most cardiomyopathy treatments repurpose heart failure drugs to manage these symptoms and avoid adverse outcomes. There are few therapies that correct the underlying pathogenic genetic or molecular mechanism. This review will reflect on this unmet clinical need in genetic cardiomyopathies and consider a variety of therapies that address the mechanism of disease rather than patient symptoms. These therapies are genetic, targeting a defective gene or transcript, or ameliorating a genetic insufficiency. However, there are also a number of small molecules under exploration that modulate downstream faulty protein products affected in cardiomyopathies.
Collapse
Affiliation(s)
- Giuliana G Repetti
- From the Department of Genetics, Harvard Medical School, Boston, MA (G.G.R., C.N.T., J.G.S., C.E.S.)
| | - Christopher N Toepfer
- From the Department of Genetics, Harvard Medical School, Boston, MA (G.G.R., C.N.T., J.G.S., C.E.S.)
- Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, United Kingdom (C.N.T.)
- Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (C.N.T., C.E.S.)
| | - Jonathan G Seidman
- From the Department of Genetics, Harvard Medical School, Boston, MA (G.G.R., C.N.T., J.G.S., C.E.S.)
| | - Christine E Seidman
- From the Department of Genetics, Harvard Medical School, Boston, MA (G.G.R., C.N.T., J.G.S., C.E.S.)
- Howard Hughes Medical Institute, Chevy Chase, MD (C.E.S.)
| |
Collapse
|
176
|
Briasoulis A, Sharma S, Telila T, Mallikethi-Reddy S, Papageorgiou N, Oikonomou E, Tousoulis D. MicroRNAs in Atrial Fibrillation. Curr Med Chem 2019; 26:855-863. [PMID: 28933293 DOI: 10.2174/0929867324666170920151024] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 11/20/2016] [Accepted: 11/29/2016] [Indexed: 12/20/2022]
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs, involved in regulation of post-transcriptional gene expression. They exert key role not only in physiology and normal development of the cardiovascular system but also in cardiovascular disease development and progression. Recent animal and human studies of tissue specific miRNAs have suggested a role in structural and electrical remodeling in atrial fibrillation (AF). Their emerging role as biomarkers and potential therapeutic targets in patients with AF is discussed in this review.
Collapse
Affiliation(s)
- Alexandros Briasoulis
- Wayne State University, Division of Cardiology, Detroit, Michigan, MI, United States
| | - Shikha Sharma
- University of Michigan, Division of Cardiology, Arrhythmia services, Ann Arbor, Michigan, MI, United States
| | - Tesfaye Telila
- Wayne State University, Division of Cardiology, Detroit, Michigan, MI, United States
| | | | | | - Evangelos Oikonomou
- 1st Department of Cardiology, University of Athens Medical School, Athens, Greece
| | - Dimitris Tousoulis
- 1st Department of Cardiology, University of Athens Medical School, Athens, Greece
| |
Collapse
|
177
|
Groenendyk J, Fan X, Peng Z, Kurgan L, Michalak M. Endoplasmic reticulum and the microRNA environment in the cardiovascular system 1. Can J Physiol Pharmacol 2019; 97:515-527. [PMID: 31063413 DOI: 10.1139/cjpp-2018-0720] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Stress responses are important to human physiology and pathology, and the inability to adapt to cellular stress leads to cell death. To mitigate cellular stress and re-establish homeostasis, cells, including those in the cardiovascular system, activate stress coping response mechanisms. The endoplasmic reticulum, a component of the cellular reticular network in cardiac cells, mobilizes so-called endoplasmic reticulum stress coping responses, such as the unfolded protein response. MicroRNAs play an important part in the maintenance of cellular and tissue homeostasis, perform a central role in the biology of the cardiac myocyte, and are involved in pathological cardiac function and remodeling. In this paper, we review a link between endoplasmic reticulum homeostasis and microRNA with an emphasis on the impact on stress responses in the cardiovascular system.
Collapse
Affiliation(s)
- Jody Groenendyk
- a Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2S7, Canada
| | - Xiao Fan
- b Irving Medical Center, Columbia University, New York, NY 10032, USA
| | - Zhenling Peng
- c Center for Applied Mathematics, Tianjin University, Tianjin 300072, China
| | - Lukasz Kurgan
- d Department of Computer Science, Virginia Commonwealth University, Richmond, VA 23284, USA.,e Department of Electrical and Computer Engineering, University of Alberta, Edmonton, AB T6G 2V4, Canada
| | - Marek Michalak
- a Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2S7, Canada
| |
Collapse
|
178
|
Zhu X, Zhao Y, Hou W, Guo L. MiR-153 regulates cardiomyocyte apoptosis by targeting Nrf2/HO-1 signaling. Chromosome Res 2019; 27:167-178. [DOI: 10.1007/s10577-019-09608-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 04/15/2019] [Accepted: 04/15/2019] [Indexed: 12/21/2022]
|
179
|
Fung EC, Butt AN, Eastwood J, Swaminathan R, Sodi R. Circulating microRNA in cardiovascular disease. Adv Clin Chem 2019; 91:99-122. [PMID: 31331491 DOI: 10.1016/bs.acc.2019.03.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Acute myocardial infarction (AMI) and heart failure (HF) are two major causes of cardiovascular mortality and morbidity. Early diagnosis of these conditions is essential to instigate immediate treatment that may result in improved outcomes. Traditional biomarkers of AMI include cardiac troponins and other proteins released from the injured myocardium but there are a number of limitations with these biomarkers especially with regard to specificity. In the past few years circulating nucleic acids, notably microRNA that are small non-coding RNAs that regulate various cellular processes, have been investigated as biomarkers of disease offering improved sensitivity and specificity in the diagnosis and prognostication of various conditions. In this review, the role of microRNAs as biomarkers used in the diagnosis of AMI and HF is discussed, their advantage over traditional biomarkers is outlined and the potential for their implementation in clinical practice is critically assessed.
Collapse
Affiliation(s)
- En C Fung
- Department of Laboratory Services, Raja Isteri Pengiran Anak Saleha (RIPAS) Hospital, Bandar Seri Begawan, Brunei Darussalam
| | - Asif N Butt
- Department of Clinical Biochemistry, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Jarlath Eastwood
- Aberdeen Royal Infirmary, NHS Grampian, Aberdeen, United Kingdom
| | - Ramasamyiyer Swaminathan
- Department of Clinical Biochemistry, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Ravinder Sodi
- Department of Blood Sciences, University Hospitals of Morecambe Bay NHS Foundation Trust, Lancaster, United Kingdom; Lancaster Medical School, Lancaster University, Lancaster, United Kingdom.
| |
Collapse
|
180
|
Collagen XXV promotes myoblast fusion during myogenic differentiation and muscle formation. Sci Rep 2019; 9:5878. [PMID: 30971718 PMCID: PMC6458142 DOI: 10.1038/s41598-019-42296-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 03/27/2019] [Indexed: 11/25/2022] Open
Abstract
Fusion of myoblasts into multinucleated myofibers is crucial for skeletal muscle development and regeneration. However, the mechanisms controlling this process remain to be determined. Here we identified the involvement of a new extracellular matrix protein in myoblast fusion. Collagen XXV is a transmembrane-type collagen highly transcribed during early myogenesis when primary myofibers form. Limb muscles of E12.5 and E14.5 Col25a1−/− embryos show a clear defect in the formation of multinucleated myofibers. In cell culture, the cleaved soluble extracellular domain of the collagen XXV is sufficient to promote the formation of highly multinucleated myofibers. Col25a1 is transiently expressed during myogenic differentiation and Col25a1 transcripts are down-regulated in multinucleated myofibers by a muscle-specific microRNA, miR-499. Altogether, these findings indicate that collagen XXV is required in vivo and in vitro for the fusion of myoblasts into myofibers and give further evidence that microRNAs participate to the regulation of this process.
Collapse
|
181
|
Ramón Y Cajal S, Segura MF, Hümmer S. Interplay Between ncRNAs and Cellular Communication: A Proposal for Understanding Cell-Specific Signaling Pathways. Front Genet 2019; 10:281. [PMID: 31001323 PMCID: PMC6454836 DOI: 10.3389/fgene.2019.00281] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 03/14/2019] [Indexed: 01/09/2023] Open
Abstract
Intercellular communication is essential for the development of specialized cells, tissues, and organs and is critical in a variety of diseases including cancer. Current knowledge states that different cell types communicate by ligand–receptor interactions: hormones, growth factors, and cytokines are released into the extracellular space and act on receptors, which are often expressed in a cell-type-specific manner. Non-coding RNAs (ncRNAs) are emerging as newly identified communicating factors in both physiological and pathological states. This class of RNA encompasses microRNAs (miRNAs, well-studied post-transcriptional regulators of gene expression), long non-coding RNAs (lncRNAs) and other ncRNAs. lncRNAs are diverse in length, sequence, and structure (linear or circular), and their functions are described as transcriptional regulation, induction of epigenetic changes and even direct regulation of protein activity. They have also been reported to act as miRNA sponges, interacting with miRNA and modulating its availability to endogenous mRNA targets. Importantly, lncRNAs may have a cell-type-specific expression pattern. In this paper, we propose that lncRNA–miRNA interactions, analogous to receptor–ligand interactions, are responsible for cell-type-specific outcomes. Specific binding of miRNAs to lncRNAs may drive cell-type-specific signaling cascades and modulate biochemical feedback loops that ultimately determine cell identity and response to stress factors.
Collapse
Affiliation(s)
- Santiago Ramón Y Cajal
- Department of Pathology, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain.,Translational Molecular Pathology, Vall d'Hebron Research Institute, Barcelona, Spain.,Spanish Biomedical Research Network Centre in Oncology (CIBERONC), Barcelona, Spain
| | - Miguel F Segura
- Group of Translational Research in Child and Adolescent Cancer, Vall d'Hebron Research Institute, Barcelona, Spain
| | - Stefan Hümmer
- Translational Molecular Pathology, Vall d'Hebron Research Institute, Barcelona, Spain.,Spanish Biomedical Research Network Centre in Oncology (CIBERONC), Barcelona, Spain
| |
Collapse
|
182
|
Wang M, Ling W, Xiong C, Xie D, Chu X, Li Y, Qiu X, Li Y, Xiao X. Potential Strategies for Cardiac Diseases: Lineage Reprogramming of Somatic Cells into Induced Cardiomyocytes. Cell Reprogram 2019; 21:63-77. [DOI: 10.1089/cell.2018.0052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Mingyu Wang
- Department of Animal Science, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Wenhui Ling
- Department of Animal Science, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Chunxia Xiong
- Department of Animal Science, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Dengfeng Xie
- Department of Animal Science, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Xinyue Chu
- Department of Animal Science, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Yunxin Li
- Department of Animal Science, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Xiaoyan Qiu
- Department of Animal Science, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Yuemin Li
- Department of Animal Science, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Xiong Xiao
- Department of Animal Science, College of Animal Science and Technology, Southwest University, Chongqing, China
| |
Collapse
|
183
|
Dueñas A, Expósito A, Aranega A, Franco D. The Role of Non-Coding RNA in Congenital Heart Diseases. J Cardiovasc Dev Dis 2019; 6:E15. [PMID: 30939839 PMCID: PMC6616598 DOI: 10.3390/jcdd6020015] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 03/24/2019] [Accepted: 03/26/2019] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular development is a complex developmental process starting with the formation of an early straight heart tube, followed by a rightward looping and the configuration of atrial and ventricular chambers. The subsequent step allows the separation of these cardiac chambers leading to the formation of a four-chambered organ. Impairment in any of these developmental processes invariably leads to cardiac defects. Importantly, our understanding of the developmental defects causing cardiac congenital heart diseases has largely increased over the last decades. The advent of the molecular era allowed to bridge morphogenetic with genetic defects and therefore our current understanding of the transcriptional regulation of cardiac morphogenesis has enormously increased. Moreover, the impact of environmental agents to genetic cascades has been demonstrated as well as of novel genomic mechanisms modulating gene regulation such as post-transcriptional regulatory mechanisms. Among post-transcriptional regulatory mechanisms, non-coding RNAs, including therein microRNAs and lncRNAs, are emerging to play pivotal roles. In this review, we summarize current knowledge on the functional role of non-coding RNAs in distinct congenital heart diseases, with particular emphasis on microRNAs and long non-coding RNAs.
Collapse
Affiliation(s)
- Angel Dueñas
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain.
| | - Almudena Expósito
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain.
| | - Amelia Aranega
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain.
| | - Diego Franco
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain.
| |
Collapse
|
184
|
Xie S, Li X, Qian L, Cai C, Xiao G, Jiang S, Li B, Gao T, Cui W. An integrated analysis of mRNA and miRNA in skeletal muscle from myostatin-edited Meishan pigs. Genome 2019; 62:305-315. [PMID: 30913397 DOI: 10.1139/gen-2018-0110] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Myostatin (MSTN) is a key muscle factor that negatively regulates skeletal muscle growth and development. Our laboratory recently produced genetically engineered Meishan pigs containing a ZFN-edited MSTN loss-of-function mutation (MSTN-/-, MKO) that led to the hypertrophy of skeletal muscles. In this study, we performed transcriptome sequencing and miRNA sequencing in skeletal muscle samples from MKO and wildtype Meishan (MWT) pigs to investigate the effect of MSTN-/- on expression of mRNA and miRNA. Our results indicated that, compared to MWT pigs, there were 200 genes and 4 miRNAs being significantly up-regulated, and 238 genes and 5 miRNAs being significantly down-regulated in MKO pigs. Analysis by GO and KEGG pathways revealed that differentially expressed miRNAs and their target genes of those differentially expressed miRNAs were involved in the signal pathways of skeletal muscle growth and development such as AMPK, mTOR, and TGF-beta. An integrated analysis of the correlation between miRNA-mRNA and transcriptome predicated that XK and METTL8 were target genes for miR-499-5p, while LRP4 was a target gene for miR-490-3p. Our results provide important clues to help us further investigate MSTN's regulatory mechanisms during skeletal muscle growth and development.
Collapse
Affiliation(s)
- Shanshan Xie
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, P.R. China
| | - Xiang Li
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, P.R. China
| | - Lili Qian
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, P.R. China.,Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, P.R. China
| | - Chunbo Cai
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, P.R. China.,Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, P.R. China
| | - Gaojun Xiao
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, P.R. China.,Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, P.R. China
| | - Shengwang Jiang
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, P.R. China.,Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, P.R. China
| | - Biao Li
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, P.R. China.,Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, P.R. China
| | - Ting Gao
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, P.R. China.,Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, P.R. China
| | - Wentao Cui
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, P.R. China.,Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, P.R. China
| |
Collapse
|
185
|
Keepers B, Liu J, Qian L. What's in a cardiomyocyte - And how do we make one through reprogramming? BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1867:118464. [PMID: 30922868 DOI: 10.1016/j.bbamcr.2019.03.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 03/10/2019] [Accepted: 03/21/2019] [Indexed: 12/19/2022]
Abstract
Substantial progress is being made in the field cardiac reprogramming, and those in the field are hopeful that the technology will be formulated for therapeutic use. Beyond the excitement around generating a revolutionary new approach for treating ischemic heart diseases, cardiac reprogramming has delivered provocative findings that challenge common notions of cell fate and cell identity. Have we really made de novo cardiomyocytes? To answer this question, the essential characteristics of this unique and important cell type must first be defined. In this review, we walk through the history of scientific inquiry into cardiomyocytes, and then we examine the core features of cardiomyocytes as detailed in modern definitions. Informed by this, we turn to cardiac reprogramming to analyze the various screening approaches and ultimate factor combinations used in each study. We follow this with a dissection of the evidence used to support the authors' claims of successfully creating cardiomyocytes, and we end by discussing what is known about the molecular mechanisms of cardiac reprogramming. Through this analysis, we find interesting differences between the study designs and their results, but it becomes clear that the field at large is generating cells that closely match the textbook definition cardiomyocyte. However, the differences noted between the results of each study are largely unexplained, reflecting the need for further research in both cardiac reprogramming and in native cardiomyocyte biology. Knowledge gained from future research will help move the field towards better reprogramming techniques and technologies.
Collapse
Affiliation(s)
- Benjamin Keepers
- McAllister Heart Institute, Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jiandong Liu
- McAllister Heart Institute, Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Li Qian
- McAllister Heart Institute, Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|
186
|
Sun X, Wang Y, Xia B, Li Z, Dai J, Qiu P, Ma A, Lin Z, Huang J, Wang J, Xie WB, Wang J. Methamphetamine produces cardiac damage and apoptosis by decreasing melusin. Toxicol Appl Pharmacol 2019; 378:114543. [PMID: 30904475 DOI: 10.1016/j.taap.2019.03.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 03/18/2019] [Accepted: 03/19/2019] [Indexed: 12/17/2022]
Abstract
Methamphetamine (METH) is an amphetamine-type drug that is highly addictive and widely abused. Many studies have shown that METH exposure causes severe damage not only to the nervous system but also to the cardiovascular system. Melusin protein is a mechanotransducer that plays an important role in maintaining normal heart function. However, the role of melusin in METH-induced cardiotoxicity has not yet been reported. We hypothesized that methamphetamine can produce cardiac damage and apoptosis by decreasing the quantity of melusin. To test this hypothesis, we determined the protein expression of melusin and apoptosis markers in METH-treated rats and primary rat cardiomyocytes. We also established a melusin-overexpressing cell model to assess the importance of melusin in maintaining antiapoptotic pathways. To confirm our findings from the in vitro and animal models, we also evaluated the apoptotic index of cardiomyocytes and the protein expression of apoptotic markers in postmortem heart tissues from deceased METH abusers and age-matched control subjects. The results showed that the apoptosis of cardiomyocytes was increased significantly and that the protein expression of melusin was decreased after exposure to METH in primary rat cardiomyocytes, in rats and in humans. METH treatment also decreased the expression of the downstream proteins FAK, IQGAP1, p-AKT, p-GSK3β, and p-ERK in primary rat cardiomyocytes and in vivo. After overexpression of melusin, the above effects were partially reversed in primary rat cardiomyocytes. We conclude that METH can produce cardiac damage and apoptosis by decreasing melusin, while melusin-activated signaling by phosphorylated AKT, phosphorylated GSK3β, and ERK may be resistant to methamphetamine-induced myocardial apoptosis.
Collapse
Affiliation(s)
- Xiaoyu Sun
- Department of Forensic Medicine, Guizhou Medical University, Guiyang 550004, China
| | - Yu Wang
- Department of Forensic Medicine, Guizhou Medical University, Guiyang 550004, China
| | - Bing Xia
- Department of Forensic Medicine, Guizhou Medical University, Guiyang 550004, China
| | - Zhu Li
- Department of Forensic Medicine, Guizhou Medical University, Guiyang 550004, China
| | - Jialin Dai
- Department of Forensic Medicine, Guizhou Medical University, Guiyang 550004, China
| | - Pingming Qiu
- School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Ande Ma
- Department of Hygiene Inspection & Quarantine Science, Guangdong Province Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Zhoumeng Lin
- Institute of Computational Comparative Medicine and Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - Jiang Huang
- Department of Forensic Medicine, Guizhou Medical University, Guiyang 550004, China
| | - Jiawen Wang
- Department of Forensic Medicine, Guizhou Medical University, Guiyang 550004, China
| | - Wei-Bing Xie
- School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China.
| | - Jie Wang
- Department of Forensic Medicine, Guizhou Medical University, Guiyang 550004, China.
| |
Collapse
|
187
|
Rawal S, Nagesh PT, Coffey S, Van Hout I, Galvin IF, Bunton RW, Davis P, Williams MJA, Katare R. Early dysregulation of cardiac-specific microRNA-208a is linked to maladaptive cardiac remodelling in diabetic myocardium. Cardiovasc Diabetol 2019; 18:13. [PMID: 30696455 PMCID: PMC6352455 DOI: 10.1186/s12933-019-0814-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Accepted: 01/14/2019] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND The diabetic heart undergoes remodelling contributing to an increased incidence of heart failure in individuals with diabetes at a later stage. The molecular regulators that drive this process in the diabetic heart are still unknown. METHODS Real-time (RT) PCR analysis was performed to determine the expression of cardiac specific microRNA-208a in right atrial appendage (RAA) and left ventricular (LV) biopsy tissues collected from diabetic and non-diabetic patients undergoing coronary artery bypass graft surgery. To determine the time-dependent changes, cardiac tissue were collected from type 2 diabetic mice at different age groups. A western blotting analysis was conducted to determine the expression of contractile proteins α- and β-myosin heavy chain (MHC) and thyroid hormone receptor-α (TR-α), the negative regulator of β-MHC. To determine the beneficial effects of therapeutic modulation of miR-208a, high glucose treated adult mouse HL-1 cardiomyocytes were transfected with anti-miR-208a. RESULTS RT-PCR analysis showed marked upregulation of miR-208a from early stages of diabetes in type 2 diabetic mouse heart, which was associated with a marked increase in the expression of pro-hypertrophic β-MHC and downregulation of TR-α. Interestingly, upregulation of miR-208a preceded the switch of α-/β-MHC isoforms and the development of diastolic and systolic dysfunction. We also observed significant upregulation of miR-208a and modulation of miR-208a associated proteins in the type 2 human diabetic heart. Therapeutic inhibition of miR-208a activity in high glucose treated HL-1 cardiomyocytes prevented the activation of β-MHC and hence the hypertrophic response. CONCLUSION Our results provide the first evidence that early modulation of miR-208a in the diabetic heart induces alterations in the downstream signaling pathway leading to cardiac remodelling and that therapeutic inhibition of miR-208a may be beneficial in preventing diabetes-induced adverse remodelling of the heart.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- Animals
- Cell Line
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/metabolism
- Diabetic Cardiomyopathies/etiology
- Diabetic Cardiomyopathies/genetics
- Diabetic Cardiomyopathies/metabolism
- Diabetic Cardiomyopathies/physiopathology
- Disease Models, Animal
- Female
- Gene Expression Regulation
- Heart Ventricles/metabolism
- Heart Ventricles/physiopathology
- Humans
- Hypertrophy, Left Ventricular/etiology
- Hypertrophy, Left Ventricular/genetics
- Hypertrophy, Left Ventricular/metabolism
- Hypertrophy, Left Ventricular/physiopathology
- Male
- Mice, Inbred C57BL
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Middle Aged
- Myocytes, Cardiac/metabolism
- Myosin Heavy Chains/genetics
- Myosin Heavy Chains/metabolism
- Signal Transduction
- Time Factors
- Ventricular Function, Left
- Ventricular Myosins/genetics
- Ventricular Myosins/metabolism
- Ventricular Remodeling
Collapse
Affiliation(s)
- Shruti Rawal
- Department of Physiology-HeartOtago, Otago School of Medical Sciences, University of Otago, 270, Great King Street, Dunedin, 9010 New Zealand
- Present Address: New York University, New York, USA
| | - Prashanth Thevakar Nagesh
- Department of Microbiology & Immunology, Otago School of Medical Sciences, University of Otago, Dunedin, New Zealand
- Present Address: New York University, New York, USA
| | - Sean Coffey
- Department of Medicine, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Isabelle Van Hout
- Department of Physiology-HeartOtago, Otago School of Medical Sciences, University of Otago, 270, Great King Street, Dunedin, 9010 New Zealand
| | - Ivor F. Galvin
- Department of Cardiothoracic Surgery, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Richard W. Bunton
- Department of Cardiothoracic Surgery, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Philip Davis
- Department of Cardiothoracic Surgery, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Michael J. A. Williams
- Department of Medicine, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Rajesh Katare
- Department of Physiology-HeartOtago, Otago School of Medical Sciences, University of Otago, 270, Great King Street, Dunedin, 9010 New Zealand
| |
Collapse
|
188
|
Abstract
Cardiotoxicity is a well-known side effect of doxorubicin (DOX), but the mechanisms leading to this phenomenon are still not completely clear. Prediction of drug-induced dysfunction onset is difficult and is still largely based on detection of cardiac troponin (cTn), a circulating marker of heart damage. In the last years, several investigations focused on the possible involvement of microRNAs (miRNAs) in DOX-induced toxicity in vitro, with contrasting results. Recently, several groups employed animal models to mimic patient’s condition, investigate the biological pathways perturbed by DOX, and identify diagnostic markers of cardiotoxicity. We reviewed the results from several studies investigating cardiac miRNAs expression in rodent models of DOX-treatment. We also discussed the data from two publications indicating the possible use of circulating miRNA as biomarkers of DOX-induced cardiotoxicity. Unfortunately, limited information was derived from these studies, as selection methods of candidate-miRNAs and heterogeneity in cardiotoxicity assessment greatly hampered the novelty and robustness of the findings. Nevertheless, at least one circulating miRNA, miR-1, showed a good potential as early biomarker of drug-mediated cardiac dysfunction onset. The use of animal models to investigate DOX-induced cardiotoxicity surely helps narrowing the gap between basic research and clinical practice. Despite this, several issues, including selection of relevant miRNAs and less-than-optimal assessment of cardiotoxicity, greatly limited the results obtained so far. Nonetheless, the association of patients-based studies with the use of preclinical models may be the key to address the many unanswered questions regarding the pathophysiology and early detection of cardiotoxicity.
Collapse
|
189
|
Forini F, Nicolini G, Pitto L, Iervasi G. Novel Insight Into the Epigenetic and Post-transcriptional Control of Cardiac Gene Expression by Thyroid Hormone. Front Endocrinol (Lausanne) 2019; 10:601. [PMID: 31555215 PMCID: PMC6727178 DOI: 10.3389/fendo.2019.00601] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 08/16/2019] [Indexed: 12/17/2022] Open
Abstract
Thyroid hormone (TH) signaling is critically involved in the regulation of cardiovascular physiology. Even mild reductions of myocardial TH levels, as occur in hypothyroidism or low T3 state conditions, are thought to play a role in the progression of cardiac disorders. Due to recent advances in molecular mechanisms underlying TH action, it is now accepted that TH-dependent modulation of gene expression is achieved at multiple transcriptional and post-transcriptional levels and involves the cooperation of many processes. Among them, the epigenetic remodeling of chromatin structure and the interplay with non-coding RNA have emerged as novel TH-dependent pathways that add further degrees of complexity and broaden the network of genes controlled by TH signaling. Increasing experimental and clinical findings indicate that aberrant function of these regulatory mechanisms promotes the evolution of cardiac disorders such as post-ischemic injury, pathological hypertrophy, and heart failure, which may be reversed by the correction of the underlying TH dyshomeostasis. To encourage the clinical implementation of a TH replacement strategy in cardiac disease, here we discuss the crucial effect of epigenetic modifications and control of non-coding RNA in TH-dependent regulation of biological processes relevant for cardiac disease evolution.
Collapse
|
190
|
Huang K, Hu S, Cheng K. A New Era of Cardiac Cell Therapy: Opportunities and Challenges. Adv Healthc Mater 2019; 8:e1801011. [PMID: 30548836 PMCID: PMC6368830 DOI: 10.1002/adhm.201801011] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 11/05/2018] [Indexed: 12/15/2022]
Abstract
Myocardial infarction (MI), caused by coronary heart disease (CHD), remains one of the most common causes of death in the United States. Over the last few decades, scientists have invested considerable resources on the study and development of cell therapies for myocardial regeneration after MI. However, due to a number of limitations, they are not yet readily available for clinical applications. Mounting evidence supports the theory that paracrine products are the main contributors to the regenerative effects attributed to these cell therapies. The next generation of cell-based MI therapies will identify and isolate cell products and derivatives, integrate them with biocompatible materials and technologies, and use them for the regeneration of damaged myocardial tissue. This review discusses the progress made thus far in pursuit of this new generation of cell therapies. Their fundamental regenerative mechanisms, their potential to combine with other therapeutic products, and their role in shaping new clinical approaches for heart tissue engineering, are addressed.
Collapse
Affiliation(s)
- Ke Huang
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, 27607, USA
| | - Shiqi Hu
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, 27607, USA
| | - Ke Cheng
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, 27607, USA
- Pharmacoengineeirng and Molecular Pharmaceutics Division, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| |
Collapse
|
191
|
Wadley GD, Lamon S, Alexander SE, McMullen JR, Bernardo BC. Noncoding RNAs regulating cardiac muscle mass. J Appl Physiol (1985) 2018; 127:633-644. [PMID: 30571279 DOI: 10.1152/japplphysiol.00904.2018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Noncoding RNAs, including microRNAs (miRNAs), long noncoding RNAs (lncRNAs), and circular RNAs (circRNAs) play roles in the development and homeostasis of nearly every tissue of the body, including the regulation of processes underlying heart growth. Cardiac hypertrophy can be classified as either physiological (beneficial heart growth) or pathological (detrimental heart growth), the latter of which results in impaired cardiac function and heart failure and is predictive of a higher incidence of death due to cardiovascular disease. Several miRNAs have a functional role in exercise-induced cardiac hypertrophy, while both miRNAs and lncRNAs are heavily involved in pathological heart growth and heart failure. The latter have the potential to act as an endogenous sponge RNA and interact with specific miRNAs to control cardiac hypertrophy, adding another level of complexity to our understanding of the regulation of cardiac muscle mass. In addition to tissue-specific effects, ncRNA-mediated tissue cross talk occurs via exosomes. In particular, miRNAs can be internalized in exosomes and secreted from various cardiac and vascular cell types to promote angiogenesis, as well as protection and repair of ischemic tissues. ncRNAs hold promising therapeutic potential to protect the heart against ischemic injury and aid in regeneration. Numerous preclinical studies have demonstrated the therapeutic potential of ncRNAs, specifically miRNAs, for the treatment of cardiovascular disease. Most of these studies employ antisense oligonucleotides to inhibit miRNAs of interest; however, off-target effects often limit their potential to be translated to the clinic. In this context, approaches using viral and nonviral delivery tools are promising means to provide targeted delivery in vivo.
Collapse
Affiliation(s)
- Glenn D Wadley
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Burwood, Australia
| | - Séverine Lamon
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Burwood, Australia
| | - Sarah E Alexander
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Burwood, Australia
| | - Julie R McMullen
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.,Department of Diabetes, Central Clinical School, Monash University, Clayton, Victoria, Australia.,Department of Medicine, Monash University, Clayton, Victoria, Australia.,Department of Physiology, Monash University, Clayton, Victoria, Australia.,Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, Victoria, Australia
| | - Bianca C Bernardo
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.,Department of Diabetes, Central Clinical School, Monash University, Clayton, Victoria, Australia.,Department of Paediatrics, University of Melbourne, Victoria, Australia
| |
Collapse
|
192
|
Screening for Circulating MiR-208a and -b in Different Cardiac Arrhythmias of Dogs. J Vet Res 2018; 62:359-363. [PMID: 30584617 PMCID: PMC6296005 DOI: 10.2478/jvetres-2018-0051] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 09/26/2018] [Indexed: 01/20/2023] Open
Abstract
Introduction In recent years, the high sensitivity and specificity of novel miRNA biomarkers have been utilised for early diagnosis and treatment monitoring of various diseases. Previous reports showed that abnormal expression of miR-208 in mice resulted in the development of an aberrant cardiac conduction system and consecutive arrhythmias. On the other hand, a study on infarcted human heart tissue showed upregulation of miR-208a in subjects with ventricular tachyarrhythmias compared to healthy controls. We prospectively investigated the expression of miR-208a and -208b in the serum of dogs presenting different cardiac arrhythmias. Material and Methods A total of 28 dogs with atrial fibrillation (n = 8), ventricular premature contractions (n=6), conduction system disturbances (n = 7), and free of heart conditions (as controls) (n = 7) were enrolled in the study. Total RNA was extracted from serum samples and miR-208a and -b, miR-16 as well as a cel-miR-39-5p spike-in were analysed with qPCR and ddPCR. Results miR-208a and miR-208b were not expressed in any of the samples. The calculated ddPCR miR-16 relative expression (normalised with cel-miR-39 spike-in) showed a good correlation (r = 0.82; P < 0.001) with the qPCR results. Conclusion This outcome warrants further investigation, possibly focusing on tissue expression of miR-208 in the canine heart.
Collapse
|
193
|
Lock MC, Tellam RL, Botting KJ, Wang KCW, Selvanayagam JB, Brooks DA, Seed M, Morrison JL. The role of miRNA regulation in fetal cardiomyocytes, cardiac maturation and the risk of heart disease in adults. J Physiol 2018; 596:5625-5640. [PMID: 29785790 PMCID: PMC6265572 DOI: 10.1113/jp276072] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 05/15/2018] [Indexed: 12/14/2022] Open
Abstract
Myocardial infarction is a primary contributor towards the global burden of cardiovascular disease. Rather than repairing the existing damage of myocardial infarction, current treatments only address the symptoms of the disease and reducing the risk of a secondary infarction. Cardiac regenerative capacity is dependent on cardiomyocyte proliferation, which concludes soon after birth in humans and precocial species such as sheep. Human fetal cardiac tissue has some ability to repair following tissue damage, whereas a fully matured human heart has minimal capacity for cellular regeneration. This is in contrast to neonatal mice and adult zebrafish hearts, which retain the ability to undergo cardiomyocyte proliferation and can regenerate cardiac tissue after birth. In mice and zebrafish models, microRNAs (miRNAs) have been implicated in the regulation of genes involved in cardiac cell cycle progression and regeneration. However, the significance of miRNA regulation in cardiomyocyte proliferation for humans and other large mammals, where the timing of heart development in relation to birth is similar, remains unclear. miRNAs may be valuable targets for therapies that promote cardiac repair after injury. Therefore, elucidating the role of specific miRNAs in large animals, where heart development closely resembles that of humans, remains vitally important for identifying therapeutic targets that may be translated into clinical practice focused on tissue repair.
Collapse
Affiliation(s)
- Mitchell C. Lock
- Early Origins of Adult Health Research Group, School of Pharmacy & Medical SciencesUniversity of South AustraliaAdelaideSA 5001Australia
| | - Ross L. Tellam
- Early Origins of Adult Health Research Group, School of Pharmacy & Medical SciencesUniversity of South AustraliaAdelaideSA 5001Australia
| | - Kimberley J. Botting
- Early Origins of Adult Health Research Group, School of Pharmacy & Medical SciencesUniversity of South AustraliaAdelaideSA 5001Australia
| | - Kimberley C. W. Wang
- Early Origins of Adult Health Research Group, School of Pharmacy & Medical SciencesUniversity of South AustraliaAdelaideSA 5001Australia
- School of Human SciencesUniversity of Western AustraliaCrawleyWA 6009Australia
| | - Joseph B. Selvanayagam
- Cardiac Imaging Research Group, Department of Heart HealthSouth Australian Health & Medical Research Institute, and Flinders UniversityGPO Box 2100AdelaideSA 5001Australia
| | - Doug A. Brooks
- Mechanisms in Cell Biology and Disease Research Group, School of Pharmacy & Medical SciencesUniversity of South AustraliaAdelaideSA 5001Australia
| | - Mike Seed
- Hospital for Sick Children, Division of Cardiology555 University AvenueTorontoON M5G 1X8Canada
| | - Janna L. Morrison
- Early Origins of Adult Health Research Group, School of Pharmacy & Medical SciencesUniversity of South AustraliaAdelaideSA 5001Australia
| |
Collapse
|
194
|
Polasik D, Tyra M, Żak G, Terman A. An analysis of MYH7 single nucleotide polymorphism (g.7:75667956G>A) in relation to growth and carcass traits in pigs. JOURNAL OF ANIMAL AND FEED SCIENCES 2018. [DOI: 10.22358/jafs/98929/2018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
195
|
Abstract
The development of new medicines is one of the priority areas of translational medicine. A significant role for biomarkers (BM) that assess the safety and efficacy of new drugs. The right choice of BM reduces the time and costs necessary for the development of drugs and transfer them to the clinic. The review is devoted to the analysis of modern scientific literature on the role of previously known and newly discovered BM in translational research. Translational BM (TBM) established during preclinical studies and are applicable at all stages of the study. TBM should have a high sensitivity and specificity, be easily measured in real time in an easily accessible biological fluids, to evaluate the same process in different species of animals (including humans), make it possible to compare the results of clinical trials with preclinical. The main role of the TBM toxicity to predict, identify and monitor the toxicity of drugs at all stages of their study. The international consortium (Predictive Safety Testing Consortium, PSTC) whose main task is the qualification of new TBM toxicity and the search for new, more advanced than existing methods for testing markers, was established. Under PSTC formed 6 working groups, each of which coordinates research for the study and selection of TBM toxicity caused by the administration of drugs in the liver, kidney, heart and blood vessels, skeletal muscle, testes. The first qualified consortium markers were 7 contained in the urine markers for preclinical studies on rats with the goal of establishing early lesions in the kidney induced by drugs. Only a small number of BM used in the study of new drugs, can be translational.
Collapse
Affiliation(s)
- T. V. Osipova
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia
| | - V. M. Bukhman
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia
| |
Collapse
|
196
|
Cerf ME. High Fat Programming and Cardiovascular Disease. MEDICINA (KAUNAS, LITHUANIA) 2018; 54:E86. [PMID: 30428585 PMCID: PMC6262472 DOI: 10.3390/medicina54050086] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 10/31/2018] [Accepted: 11/08/2018] [Indexed: 02/06/2023]
Abstract
Programming is triggered through events during critical developmental phases that alter offspring health outcomes. High fat programming is defined as the maintenance on a high fat diet during fetal and/or early postnatal life that induces metabolic and physiological alterations that compromise health. The maternal nutritional status, including the dietary fatty acid composition, during gestation and/or lactation, are key determinants of fetal and postnatal development. A maternal high fat diet and obesity during gestation compromises the maternal metabolic state and, through high fat programming, presents an unfavorable intrauterine milieu for fetal growth and development thereby conferring adverse cardiac outcomes to offspring. Stressors on the heart, such as a maternal high fat diet and obesity, alter the expression of cardiac-specific factors that alter cardiac structure and function. The proper nutritional balance, including the fatty acid balance, particularly during developmental windows, are critical for maintaining cardiac structure, preserving cardiac function and enhancing the cardiac response to metabolic challenges.
Collapse
Affiliation(s)
- Marlon E Cerf
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa.
- Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Stellenbosch, Tygerberg 7505, South Africa.
| |
Collapse
|
197
|
Wang J, Song C, Cao X, Li H, Cai H, Ma Y, Huang Y, Lan X, Lei C, Ma Y, Bai Y, Lin F, Chen H. MiR-208b regulates cell cycle and promotes skeletal muscle cell proliferation by targeting CDKN1A. J Cell Physiol 2018; 234:3720-3729. [PMID: 30317561 DOI: 10.1002/jcp.27146] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 07/05/2018] [Indexed: 01/18/2023]
Abstract
Skeletal muscle is the most abundant tissue in the body. The development of skeletal muscle cell is complex and affected by many factors. A sea of microRNAs (miRNAs) have been identified as critical regulators of myogenesis. MiR-208b, a muscle-specific miRNA, was reported to have a connection with fiber type determination. However, whether miR-208b has effect on proliferation of muscle cell was under ascertained. In our study, cyclin-dependent kinase inhibitor 1A (CDKN1A), which participates in cell cycle regulation, was predicted and then validated as one target gene of miR-208b. We found that overexpression of miR-208b increased the expression of cyclin D1, cyclin E1, and cyclin-dependent kinase 2 at the levels of messenger RNA and protein in cattle primary myoblasts in vivo and in vitro. Flow cytometry showed that forced expression of miR-208b increased the percentage of cells at the S phase and decreased the percentage of cells at the G0/G1 phase. These results indicated that miR-208b participates in the cell cycle regulation of cattle primary myoblast cells. 5-Ethynyl-20-deoxyuridine and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assays showed that overexpression of miR-208b promoted the proliferation of cattle primary myoblasts. Therefore, we conclude that miR-208b participates in the cell cycle and proliferation regulation of cattle primary skeletal muscle cell through the posttranscriptional downregulation of CDKN1A.
Collapse
Affiliation(s)
- Jian Wang
- Shaanxi Key Laboratory of Agricultural Molecular Biology, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Chengchuang Song
- Shaanxi Key Laboratory of Agricultural Molecular Biology, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Xiukai Cao
- Shaanxi Key Laboratory of Agricultural Molecular Biology, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Hui Li
- Shaanxi Key Laboratory of Agricultural Molecular Biology, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Hanfang Cai
- Shaanxi Key Laboratory of Agricultural Molecular Biology, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Yilei Ma
- Shaanxi Key Laboratory of Agricultural Molecular Biology, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Yongzhen Huang
- Shaanxi Key Laboratory of Agricultural Molecular Biology, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Xianyong Lan
- Shaanxi Key Laboratory of Agricultural Molecular Biology, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Chuzhao Lei
- Shaanxi Key Laboratory of Agricultural Molecular Biology, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Yun Ma
- College of Life Sciences, Institute for Conservation and Utilization of Agro-Bioresources in Dabie Mountains, Xinyang Normal University, Xinyang, China
| | - Yueyu Bai
- Animal Health Supervision in Henan Province, Zhengzhou, China
| | - Fengpeng Lin
- Bureau of Animal Husbandry of Biyang County, Biyang, China
| | - Hong Chen
- Shaanxi Key Laboratory of Agricultural Molecular Biology, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| |
Collapse
|
198
|
Koyama R, Mannic T, Ito J, Amar L, Zennaro MC, Rossier MF, Maturana AD. MicroRNA-204 Is Necessary for Aldosterone-Stimulated T-Type Calcium Channel Expression in Cardiomyocytes. Int J Mol Sci 2018; 19:E2941. [PMID: 30262720 PMCID: PMC6212903 DOI: 10.3390/ijms19102941] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 09/14/2018] [Accepted: 09/19/2018] [Indexed: 01/03/2023] Open
Abstract
Activation of the mineralocorticoid receptor (MR) in the heart is considered to be a cardiovascular risk factor. MR activation leads to heart hypertrophy and arrhythmia. In ventricular cardiomyocytes, aldosterone induces a profound remodeling of ion channel expression, in particular, an increase in the expression and activity of T-type voltage-gated calcium channels (T-channels). The molecular mechanisms immediately downstream from MR activation, which lead to the increased expression of T-channels and, consecutively, to an acceleration of spontaneous cell contractions in vitro, remain poorly investigated. Here, we investigated the putative role of a specific microRNA in linking MR activation to the regulation of T-channel expression and cardiomyocyte beating frequency. A screening assay identified microRNA 204 (miR-204) as one of the major upregulated microRNAs after aldosterone stimulation of isolated neonatal rat cardiomyocytes. Aldosterone significantly increased the level of miR-204, an effect blocked by the MR antagonist spironolactone. When miR-204 was overexpressed in isolated cardiomyocytes, their spontaneous beating frequency was significantly increased after 24 h, like upon aldosterone stimulation, and messenger RNAs coding T-channels (CaV3.1 and CaV3.2) were increased. Concomitantly, T-type calcium currents were significantly increased upon miR-204 overexpression. Specifically repressing the expression of miR-204 abolished the aldosterone-induced increase of CaV3.1 and CaV3.2 mRNAs, as well as T-type calcium currents. Finally, aldosterone and miR-204 overexpression were found to reduce REST-NRSF, a known transcriptional repressor of CaV3.2 T-type calcium channels. Our study thus strongly suggests that miR-204 expression stimulated by aldosterone promotes the expression of T-channels in isolated rat ventricular cardiomyocytes, and therefore, increases the frequency of the cell spontaneous contractions, presumably through the inhibition of REST-NRSF protein.
Collapse
Affiliation(s)
- Riko Koyama
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan.
| | - Tiphaine Mannic
- Department of Human Protein Science, University of Geneva, CH-1211 Geneva, Switzerland.
| | - Jumpei Ito
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan.
| | - Laurence Amar
- Inserm, UMRS_970, Paris Cardiovascular Research Center, 75015 Paris, France.
- Université Paris Descartes, Sorbonne Paris Cité, 75015 Paris, France.
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Unité Hypertension artérielle, 75015 Paris, France.
| | - Maria-Christina Zennaro
- Inserm, UMRS_970, Paris Cardiovascular Research Center, 75015 Paris, France.
- Université Paris Descartes, Sorbonne Paris Cité, 75015 Paris, France.
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service de Génétique, 75015 Paris, France.
| | - Michel Florian Rossier
- Department of Human Protein Science, University of Geneva, CH-1211 Geneva, Switzerland.
- Central Institute of Hospitals, Hospital of Valais, CH-1951 Sion, Switzerland.
| | | |
Collapse
|
199
|
|
200
|
Wang X, Yang C, Liu X, Yang P. Ghrelin Alleviates Angiotensin II-Induced H9c2 Apoptosis: Impact of the miR-208 Family. Med Sci Monit 2018; 24:6707-6716. [PMID: 30244257 PMCID: PMC6178878 DOI: 10.12659/msm.908096] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Ghrelin is a novel peptide with abundant cardioprotective effects. The miR-208 family, consisting of cardiac-specifically expressed microRNAs, are not only involved in hypertrophy and fibrosis, but are also closely related with myocyte apoptosis. This study explored the role of the miR-208 family in the protective effect of ghrelin on angiotensin II (Ang II)-induced apoptosis. MATERIAL AND METHODS H9c2 cells were exposed to Ang II with or without ghrelin. Cell viability was detected by MTT assay and the percentage of apoptotic cells was confirmed by flow cytometry. miRNAs expression levels were measured by qRT-PCR. Then, cells transfected with miR-208 negative control, mimics, and inhibitors were treated with Ang II and ghrelin, followed by flow cytometry. PCR array was performed to explore the pathways affected by miR-208. RESULTS The miR-208 level was reduced in Ang II-treated H9c2 cells, accompanied with increased cell apoptosis, which were both reversed by ghrelin administration. Flow cytometry revealed that miR-208 inhibitors clearly upregulated the apoptotic percentage, whereas miR-208 mimics showed the opposite effects in the Ang II group. miR-208a further alleviated apoptosis when treated with ghrelin. miR-208 mainly affected caspase, inflammatory-related genes, and several signaling pathways. CONCLUSIONS We provide new evidence that the miR-208 family is regulated by Ang II and ghrelin. Overexpression of miR-208 family alleviated Ang II-induced cell apoptosis and miR-208a assisted in the protective effect of ghrelin. Several apoptosis pathways affected by miR-208 family were found. These findings suggest the pathogenesis of cardiomyocyte apoptosis and the protective mechanism of ghrelin.
Collapse
Affiliation(s)
- Xiaotong Wang
- Department of Cardiololgy, China-Japan Union Hospital, Jilin University, Changchun, Jilin, China (mainland)
| | - Chunyan Yang
- Department of Cardiology, China-Japan Union Hospital, Jilin University, Changchun, Jilin, China (mainland)
| | - Xueyan Liu
- Department of Cardiology, China-Japan Union Hospital, Jilin University, Changchun, Jilin, China (mainland)
| | - Ping Yang
- Department of Cardiology, China-Japan Union Hospital, Jilin University, Changchun, Jilin, China (mainland)
| |
Collapse
|