Noncoding RNAs in Cardiac Hypertrophy

Cardiac hypertrophy is classified as pathological and physiological hypertrophy. Pathological hypertrophy typically precedes the onset of heart failure, one of the largest contributors to disease burden and deaths worldwide. In contrast, physiological hypertrophy is an adaptive response and protects against adverse cardiac remodeling. Noncoding RNAs (ncRNAs) have drawn significant attention over the last couple of decades, and their dysregulation is increasingly being linked to cardiac hypertrophy and cardiovascular diseases. In this review, we will summarize the profiling, function, and molecular mechanism of microRNAs, long noncoding RNAs, and circular RNAs in pathological cardiac hypertrophy. Additionally, we also review microRNAs responsible for physiological hypertrophy. With better understanding of ncRNAs in cardiac hypertrophy, manipulation of the important ncRNAs will offer exciting avenues for the prevention and therapy of heart failure.

[1]  D. Riesner,et al.  Viroids are single-stranded covalently closed circular RNA molecules existing as highly base-paired rod-like structures. , 1976, Proceedings of the National Academy of Sciences of the United States of America.

[2]  V. Ambros,et al.  The lin-4 regulatory RNA controls developmental timing in Caenorhabditis elegans by blocking LIN-14 protein synthesis after the initiation of translation. , 1999, Developmental biology.

[3]  R. Graham,et al.  Dissecting cardiac hypertrophy and signaling pathways: evidence for an interaction between multifunctional g proteins and prostanoids. , 2003, Circulation research.

[4]  E. Olson,et al.  Cardiac hypertrophy: the good, the bad, and the ugly. , 2003, Annual review of physiology.

[5]  Yong Zhao,et al.  Serum response factor regulates a muscle-specific microRNA that targets Hand2 during cardiogenesis , 2005, Nature.

[6]  A. Pasquinelli,et al.  Regulation by let-7 and lin-4 miRNAs Results in Target mRNA Degradation , 2005, Cell.

[7]  Jian-Fu Chen,et al.  The role of microRNA-1 and microRNA-133 in skeletal muscle proliferation and differentiation , 2006, Nature Genetics.

[8]  E. Olson,et al.  A signature pattern of stress-responsive microRNAs that can evoke cardiac hypertrophy and heart failure , 2006, Proceedings of the National Academy of Sciences.

[9]  J. Mattick,et al.  Rapid evolution of noncoding RNAs: lack of conservation does not mean lack of function. , 2006, Trends in genetics : TIG.

[10]  C. Croce,et al.  MicroRNA-133 controls cardiac hypertrophy , 2007, Nature Medicine.

[11]  Danish Sayed,et al.  MicroRNAs Play an Essential Role in the Development of Cardiac Hypertrophy , 2007, Circulation research.

[12]  Chaoqian Xu,et al.  The muscle-specific microRNA miR-1 regulates cardiac arrhythmogenic potential by targeting GJA1 and KCNJ2 , 2011, Nature Medicine.

[13]  Michael T. McManus,et al.  Dysregulation of Cardiogenesis, Cardiac Conduction, and Cell Cycle in Mice Lacking miRNA-1-2 , 2007, Cell.

[14]  E. Olson,et al.  An intragenic MEF2-dependent enhancer directs muscle-specific expression of microRNAs 1 and 133 , 2007, Proceedings of the National Academy of Sciences.

[15]  Yanjie Lu,et al.  Down-regulation of miR-1/miR-133 Contributes to Re-expression of Pacemaker Channel Genes HCN2 and HCN4 in Hypertrophic Heart*♦ , 2008, Journal of Biological Chemistry.

[16]  E. Olson,et al.  microRNA-133a regulates cardiomyocyte proliferation and suppresses smooth muscle gene expression in the heart. , 2008, Genes & development.

[17]  J. McCarthy MicroRNA-206: the skeletal muscle-specific myomiR. , 2008, Biochimica et biophysica acta.

[18]  T. Golub,et al.  MicroRNA-1 Negatively Regulates Expression of the Hypertrophy-Associated Calmodulin and Mef2a Genes , 2009, Molecular and Cellular Biology.

[19]  Jian-Fu Chen,et al.  MicroRNA-208a is a regulator of cardiac hypertrophy and conduction in mice. , 2009, The Journal of clinical investigation.

[20]  D. Catalucci,et al.  Reciprocal Regulation of MicroRNA-1 and Insulin-Like Growth Factor-1 Signal Transduction Cascade in Cardiac and Skeletal Muscle in Physiological and Pathological Conditions , 2009, Circulation.

[21]  Jiang Chang,et al.  NFATc4 is negatively regulated in miR-133a-mediated cardiomyocyte hypertrophic repression. , 2010, American journal of physiology. Heart and circulatory physiology.

[22]  Nicholas T. Ingolia,et al.  Mammalian microRNAs predominantly act to decrease target mRNA levels , 2010, Nature.

[23]  G. Condorelli,et al.  MicroRNA-199b targets the nuclear kinase Dyrk1a in an auto-amplification loop promoting calcineurin/NFAT signalling , 2010, Nature Cell Biology.

[24]  B. Long,et al.  miR-9 and NFATc3 Regulate Myocardin in Cardiac Hypertrophy* , 2010, The Journal of Biological Chemistry.

[25]  Zhe Li,et al.  Reciprocal Repression Between MicroRNA-133 and Calcineurin Regulates Cardiac Hypertrophy: A Novel Mechanism for Progressive Cardiac Hypertrophy , 2010, Hypertension.

[26]  Hsien-Da Huang,et al.  microRNA: a master regulator of cellular processes for bioengineering systems. , 2010, Annual review of biomedical engineering.

[27]  T. Sun,et al.  Attenuation of microRNA-1 derepresses the cytoskeleton regulatory protein twinfilin-1 to provoke cardiac hypertrophy , 2010, Journal of Cell Science.

[28]  Yanjie Lu,et al.  Down-regulation of miR-1/miR-133 contributes to re-expression of pacemaker channel genes HCN2 and HCN4 in hypertrophic heart. , 2008, The Journal of Biological Chemistry.

[29]  M. Irigoyen,et al.  MicroRNAs 29 are involved in the improvement of ventricular compliance promoted by aerobic exercise training in rats. , 2011, Physiological genomics.

[30]  B. Long,et al.  Cardiac Hypertrophy Is Positively Regulated by MicroRNA miR-23a* , 2011, The Journal of Biological Chemistry.

[31]  J. Krieger,et al.  Aerobic Exercise Training–Induced Left Ventricular Hypertrophy Involves Regulatory MicroRNAs, Decreased Angiotensin-Converting Enzyme-Angiotensin II, and Synergistic Regulation of Angiotensin-Converting Enzyme 2-Angiotensin (1-7) , 2011, Hypertension.

[32]  M. Abdellatif,et al.  Thioredoxin 1 Negatively Regulates Angiotensin II–Induced Cardiac Hypertrophy Through Upregulation of miR-98/let-7 , 2011, Circulation research.

[33]  S. Kauppinen,et al.  Therapeutic inhibition of the miR-34 family attenuates pathological cardiac remodeling and improves heart function , 2012, Proceedings of the National Academy of Sciences.

[34]  M. Kyba,et al.  The H19 lincRNA is a developmental reservoir of miR-675 which suppresses growth and Igf1r , 2012, Nature Cell Biology.

[35]  Danish Sayed,et al.  GATA4 expression is primarily regulated via a miR-26b-dependent post-transcriptional mechanism during cardiac hypertrophy. , 2012, Cardiovascular research.

[36]  K. Chowdhury,et al.  The miRNA-212/132 family regulates both cardiac hypertrophy and cardiomyocyte autophagy , 2012, Nature Communications.

[37]  R. Hui,et al.  MiR‐221 promotes cardiac hypertrophy in vitro through the modulation of p27 expression , 2012, Journal of cellular biochemistry.

[38]  Qiang Sun,et al.  Cardiomyocyte overexpression of miR-27b induces cardiac hypertrophy and dysfunction in mice , 2011, Cell Research.

[39]  Da-Zhi Wang,et al.  MicroRNA-22 Regulates Cardiac Hypertrophy and Remodeling in Response to Stress , 2013, Circulation research.

[40]  J. Kjems,et al.  Natural RNA circles function as efficient microRNA sponges , 2013, Nature.

[41]  X. Chen,et al.  Reciprocal regulation of miR-23a and lysophosphatidic acid receptor signaling in cardiomyocyte hypertrophy. , 2013, Biochimica et biophysica acta.

[42]  Sebastian D. Mackowiak,et al.  Circular RNAs are a large class of animal RNAs with regulatory potency , 2013, Nature.

[43]  I. Karakikes,et al.  Therapeutic Cardiac‐Targeted Delivery of miR‐1 Reverses Pressure Overload–Induced Cardiac Hypertrophy and Attenuates Pathological Remodeling , 2013, Journal of the American Heart Association.

[44]  Michael K. Slevin,et al.  Circular RNAs are abundant, conserved, and associated with ALU repeats. , 2013, RNA.

[45]  Zhichao Ma,et al.  Swimming exercise training-induced left ventricular hypertrophy involves microRNAs and synergistic regulation of the PI3K/AKT/mTOR signaling pathway , 2013, European Journal of Applied Physiology.

[46]  Shi-ming Liu,et al.  MiR-30-Regulated Autophagy Mediates Angiotensin II-Induced Myocardial Hypertrophy , 2013, PloS one.

[47]  Shanshan Zhu,et al.  Circular intronic long noncoding RNAs. , 2013, Molecular cell.

[48]  L. Zentilin,et al.  MiR-378 Controls Cardiac Hypertrophy by Combined Repression of Mitogen-Activated Protein Kinase Pathway Factors , 2013, Circulation.

[49]  D. Guan,et al.  MicroRNA-350 induces pathological heart hypertrophy by repressing both p38 and JNK pathways. , 2013, Biochimica et biophysica acta.

[50]  Yue Jiang,et al.  miR‐145 inhibits isoproterenol‐induced cardiomyocyte hypertrophy by targeting the expression and localization of GATA6 , 2013, FEBS letters.

[51]  G. Ewald,et al.  Deep RNA Sequencing Reveals Dynamic Regulation of Myocardial Noncoding RNAs in Failing Human Heart and Remodeling With Mechanical Circulatory Support , 2014, Circulation.

[52]  Chaoqian Xu,et al.  MicroRNA-328 as a regulator of cardiac hypertrophy. , 2014, International journal of cardiology.

[53]  Jing Ye,et al.  miR-34a Modulates Angiotensin II-Induced Myocardial Hypertrophy by Direct Inhibition of ATG9A Expression and Autophagic Activity , 2014, PloS one.

[54]  R. Parker,et al.  Circular RNAs: diversity of form and function , 2014, RNA.

[55]  K. Santos,et al.  An Analysis of the Global Expression of MicroRNAs in an Experimental Model of Physiological Left Ventricular Hypertrophy , 2014, PloS one.

[56]  Xiaoke Yin,et al.  Cardiac fibroblast-derived microRNA passenger strand-enriched exosomes mediate cardiomyocyte hypertrophy. , 2014, The Journal of clinical investigation.

[57]  Ling-Ling Chen,et al.  Complementary Sequence-Mediated Exon Circularization , 2014, Cell.

[58]  N. Rajewsky,et al.  circRNA biogenesis competes with pre-mRNA splicing. , 2014, Molecular cell.

[59]  C. Feschotte,et al.  Volatile evolution of long noncoding RNA repertoires: mechanisms and biological implications. , 2014, Trends in genetics : TIG.

[60]  E. Ashley,et al.  A long non-coding RNA protects the heart from pathological hypertrophy , 2014, Nature.

[61]  E. Bronze-da-Rocha MicroRNAs Expression Profiles in Cardiovascular Diseases , 2014, BioMed research international.

[62]  Y. Zhang,et al.  Expression profile of long non-coding RNAs in a mouse model of cardiac hypertrophy. , 2014, International journal of cardiology.

[63]  F. Liu,et al.  The Long Noncoding RNA CHRF Regulates Cardiac Hypertrophy by Targeting miR-489 , 2014, Circulation research.

[64]  G. Velmurugan,et al.  MiRNAs with Apoptosis Regulating Potential Are Differentially Expressed in Chronic Exercise-Induced Physiologically Hypertrophied Hearts , 2015, PloS one.

[65]  K. Hong,et al.  MicroRNA-223 Displays a Protective Role Against Cardiomyocyte Hypertrophy by Targeting Cardiac Troponin I-Interacting Kinase , 2015, Cellular Physiology and Biochemistry.

[66]  Jin Ock Kim,et al.  miR-185 Plays an Anti-Hypertrophic Role in the Heart via Multiple Targets in the Calcium-Signaling Pathways , 2015, PloS one.

[67]  J. Zhong,et al.  The ACE2/Apelin Signaling, MicroRNAs, and Hypertension , 2015, International journal of hypertension.

[68]  Jie-ning Zhu,et al.  CDK6 mediates the effect of attenuation of miR-1 on provoking cardiomyocyte hypertrophy , 2015, Molecular and Cellular Biochemistry.

[69]  Vassilios J. Bezzerides,et al.  Abstract 238: miR-222 is Necessary for Exercise-induced Cardiac Growth and Protects Against Pathological Cardiac Remodeling , 2014 .

[70]  C. Chen,et al.  MicroRNA regulation of unfolded protein response transcription factor XBP1 in the progression of cardiac hypertrophy and heart failure in vivo , 2015, Journal of Translational Medicine.

[71]  R. Guigó,et al.  Genome-wide profiling of the cardiac transcriptome after myocardial infarction identifies novel heart-specific long non-coding RNAs , 2014, European heart journal.

[72]  B. Schroen,et al.  Long noncoding RNAs in cardiac development and ageing , 2015, Nature Reviews Cardiology.

[73]  W. Gong,et al.  miR-21-3p regulates cardiac hypertrophic response by targeting histone deacetylase-8. , 2015, Cardiovascular research.

[74]  Yolan J. Reckman,et al.  RBM20 Regulates Circular RNA Production From the Titin Gene. , 2016, Circulation research.

[75]  T. Meitinger,et al.  Characterization of circular RNAs in human, mouse and rat hearts. , 2016, Journal of molecular and cellular cardiology.

[76]  J. Salzman,et al.  Detecting circular RNAs: bioinformatic and experimental challenges , 2016, Nature Reviews Genetics.

[77]  A. Schambach,et al.  Long noncoding RNA Chast promotes cardiac remodeling , 2016, Science Translational Medicine.

[78]  P. Zhang,et al.  A Long Non-Coding RNA Defines an Epigenetic Checkpoint in Cardiac Hypertrophy , 2016 .

[79]  He Huang,et al.  MiR-155 Knockout in Fibroblasts Improves Cardiac Remodeling by Targeting Tumor Protein p53-Inducible Nuclear Protein 1 , 2016, Journal of cardiovascular pharmacology and therapeutics.

[80]  Jian Wang,et al.  miR-199-sponge transgenic mice develop physiological cardiac hypertrophy. , 2016, Cardiovascular research.

[81]  Yan Li,et al.  Circular RNA profiling reveals an abundant circHIPK3 that regulates cell growth by sponging multiple miRNAs , 2016, Nature Communications.

[82]  T. Minamino,et al.  Physiological and pathological cardiac hypertrophy. , 2016, Journal of molecular and cellular cardiology.

[83]  Jian Wang,et al.  The H19 long noncoding RNA is a novel negative regulator of cardiomyocyte hypertrophy. , 2016, Cardiovascular research.

[84]  Christoph Dieterich,et al.  Profiling and Validation of the Circular RNA Repertoire in Adult Murine Hearts , 2016, Genom. Proteom. Bioinform..

[85]  Xiaohong Wang,et al.  Overexpression of miR-223 Tips the Balance of Pro- and Anti-hypertrophic Signaling Cascades toward Physiologic Cardiac Hypertrophy* , 2016, The Journal of Biological Chemistry.

[86]  X-H Zhu,et al.  LncRNA MIAT enhances cardiac hypertrophy partly through sponging miR-150. , 2016, European review for medical and pharmacological sciences.

[87]  Wei Wu,et al.  NONCODE 2016: an informative and valuable data source of long non-coding RNAs , 2015, Nucleic Acids Res..

[88]  B. Zhao,et al.  A circular RNA protects the heart from pathological hypertrophy and heart failure by targeting miR-223. , 2016, European heart journal.

[89]  Jing Huang,et al.  Long Non-Coding RNA-ROR Mediates the Reprogramming in Cardiac Hypertrophy , 2016, PloS one.

[90]  Xiaoying Li,et al.  Unraveling the Expression Profiles of Long Noncoding RNAs in Rat Cardiac Hypertrophy and Functions of lncRNA BC088254 in Cardiac Hypertrophy Induced by Transverse Aortic Constriction , 2016, Cardiology.

[91]  Jin Ock Kim,et al.  MicroRNA‐101b attenuates cardiomyocyte hypertrophy by inhibiting protein kinase C epsilon signaling , 2017, FEBS letters.

[92]  K. See,et al.  A landscape of circular RNA expression in the human heart , 2017, Cardiovascular research.

[93]  J. Sluijter,et al.  miR-17-3 p Contributes to Exercise-Induced Cardiac Growth and Protects against Myocardial Ischemia-Reperfusion Injury , 2017 .

[94]  J. Sluijter,et al.  miR-17-3p Contributes to Exercise-Induced Cardiac Growth and Protects against Myocardial Ischemia-Reperfusion Injury , 2017, Theranostics.

[95]  J. Kjems,et al.  The emerging landscape of circular RNA in life processes , 2016, RNA biology.

[96]  A. Ciccodicola,et al.  Heart failure: Pilot transcriptomic analysis of cardiac tissue by RNA-sequencing. , 2017, Cardiology journal.

[97]  C. Zhuo,et al.  LncRNA TINCR attenuates cardiac hypertrophy by epigenetically silencing CaMKII , 2017, Oncotarget.

[98]  S. Zhuang,et al.  HOTAIR functions as a competing endogenous RNA to regulate PTEN expression by inhibiting miR-19 in cardiac hypertrophy , 2017, Molecular and Cellular Biochemistry.

[99]  E. Chen,et al.  MicroRNA-195 Regulates Metabolism in Failing Myocardium Via Alterations in Sirtuin 3 Expression and Mitochondrial Protein Acetylation , 2018, Circulation.

[100]  Chunquan Li,et al.  Identification and analysis of a key long non‐coding RNAs (lncRNAs)‐associated module reveal functional lncRNAs in cardiac hypertrophy , 2017, Journal of cellular and molecular medicine.