P. Anversa, C. Beghi, Y. Kikkawa, and G. Olivetti, Myocardial response to infarction in the rat Morphometric measurement of infarct size and myocyte cellular hypertrophy, Am J Pathol, vol.118, pp.484-92, 1985.

S. Dunlay and R. Vl, Gender differences in the pathophysiology, clinical presentation, and outcomes of ischemic heart failure. Current heart failure reports, pp.267-76, 2012.

Y. Matsui, J. Morimoto, and T. Uede, Role of matricellular proteins in cardiac tissue remodeling after myocardial infarction, World Journal of Biological Chemistry, vol.1, issue.5, pp.69-80, 2010.
DOI : 10.4331/wjbc.v1.i5.69

S. Mccurdy, C. Baicu, S. Heymans, and A. Bradshaw, Cardiac extracellular matrix remodeling: Fibrillar collagens and Secreted Protein Acidic and Rich in Cysteine (SPARC), Journal of Molecular and Cellular Cardiology, vol.48, issue.3, pp.544-553, 2010.
DOI : 10.1016/j.yjmcc.2009.06.018

M. Lindsey, Myofibroblasts and the extracellular matrix network in post-myocardial infarction cardiac remodeling, Pflugers Archiv : European journal of physiology, vol.466, pp.1113-1140, 2014.

N. Frangogiannis, The Mechanistic Basis of Infarct Healing, Antioxidants & Redox Signaling, vol.8, issue.11-12, pp.1907-1946, 2006.
DOI : 10.1089/ars.2006.8.1907

M. Sutton and N. Sharpe, Left Ventricular Remodeling After Myocardial Infarction : Pathophysiology and Therapy, Circulation, vol.101, issue.25, pp.2981-2989, 2000.
DOI : 10.1161/01.CIR.101.25.2981

D. Vanhoutte, M. Schellings, Y. Pinto, and S. Heymans, Relevance of matrix metalloproteinases and their inhibitors after myocardial infarction: A temporal and spatial window, Cardiovascular Research, vol.69, issue.3, pp.604-617, 2006.
DOI : 10.1016/j.cardiores.2005.10.002

E. Braunwald, Heart Failure, JACC: Heart Failure, vol.1, issue.1, pp.1-20, 2013.
DOI : 10.1016/j.jchf.2012.10.002

L. Daga, U. Kaul, and A. Mansoor, Approach to STEMI and NSTEMI. The Journal of the Association of Physicians of India, pp.19-25, 2011.

M. Elashoff, J. Wingrove, P. Beineke, S. Daniels, W. Tingley et al., Development of a blood-based gene expression algorithm for assessment of obstructive coronary artery disease in non-diabetic patients. BMC medical genomics, p.26, 2011.

E. Grube, L. Newby, and G. Ginsburg, Correlation of Peripheral-Blood Gene Expression With the Extent of Coronary Artery Stenosis, Circ Cardiovasc Genet, vol.1, pp.31-38, 2008.

Y. Devaux, F. Azuaje, M. Vausort, C. Yvorra, and D. Wagner, Integrated protein network and microarray analysis to identify potential biomarkers after myocardial infarction, Functional & Integrative Genomics, vol.358, issue.3, pp.329-366, 2010.
DOI : 10.2174/187152507779315813

D. Wagner, Transforming growth factor beta receptor 1 is a new candidate prognostic biomarker after acute myocardial infarction. BMC medical genomics, p.83, 2011.

F. Azuaje, F. Dewey, D. Brutsaert, Y. Devaux, E. Ashley et al., Systems-Based Approaches to Cardiovascular Biomarker Discovery, Circulation: Cardiovascular Genetics, vol.5, issue.3, pp.360-367, 2012.
DOI : 10.1161/CIRCGENETICS.112.962977

E. Goretti, D. Wagner, and Y. Devaux, miRNAs as biomarkers of myocardial infarction: a step forward towards personalized medicine?, Trends in Molecular Medicine, vol.20, issue.12, pp.716-741, 2014.
DOI : 10.1016/j.molmed.2014.10.006

M. Mueller, M. Vafaie, M. Biener, E. Giannitsis, and H. Katus, Cardiac Troponin T, Circulation Journal, vol.77, issue.7, pp.1653-61, 2013.
DOI : 10.1253/circj.CJ-13-0706

M. Parmacek and R. Solaro, Biology of the troponin complex in cardiac myocytes, Progress in Cardiovascular Diseases, vol.47, issue.3, pp.159-76, 2004.
DOI : 10.1016/j.pcad.2004.07.003

E. Giannitsis, K. Kurz, K. Hallermayer, J. Jarausch, A. Jaffe et al., Analytical Validation of a High-Sensitivity Cardiac Troponin T Assay, Clinical Chemistry, vol.56, issue.2, pp.254-61, 2010.
DOI : 10.1373/clinchem.2009.132654

T. Reichlin, W. Hochholzer, S. Bassetti, S. Steuer, C. Stelzig et al., Early Diagnosis of Myocardial Infarction with Sensitive Cardiac Troponin Assays, New England Journal of Medicine, vol.361, issue.9, pp.858-67, 2009.
DOI : 10.1056/NEJMoa0900428

D. Chan and L. Ng, Biomarkers in acute myocardial infarction, BMC Medicine, vol.105, issue.1, p.34, 2010.
DOI : 10.1161/01.CIR.0000015464.18023.0A

N. Danchin, E. Durand, V. Decalf, and A. Chaib, Risk stratification of acute coronary syndromes] Annales de cardiologie et d'angeiologie, pp.16-20, 2007.

E. Braunwald, Biomarkers in Heart Failure, New England Journal of Medicine, vol.358, issue.20, pp.2148-59, 2008.
DOI : 10.1056/NEJMra0800239

/. Authors, M. Task-force, P. Steg, S. James, D. Atar et al., ESC Guidelines for the management of acute myocardial infarction in patients presenting with ST-segment elevation: The Task Force on the management of ST-segment elevation acute myocardial infarction of the European Society of Cardiology (ESC) European heart journal, pp.2569-619, 2012.

N. Hrynchyshyn, P. Jourdain, M. Desnos, B. Diebold, and F. Funck, Galectin-3: A new biomarker for the diagnosis, analysis and prognosis of acute and chronic heart failure, Archives of Cardiovascular Diseases, vol.106, issue.10, pp.541-547, 2013.
DOI : 10.1016/j.acvd.2013.06.054

H. Jiang, L. Zhang, Y. Yu, M. Liu, J. X. Zhang et al., A pilot study of angiogenin in heart failure with preserved ejection fraction: a novel potential biomarker for diagnosis and prognosis? Journal of cellular and molecular medicine, pp.2189-97, 2014.

D. Gruson, A. Buglioni, J. Burnett, and J. , PTH: Potential role in management of heart failure, Clinica Chimica Acta, vol.433, pp.290-296, 2014.
DOI : 10.1016/j.cca.2014.03.029

F. Zhu, Z. Shi, C. Qin, L. Tao, X. Liu et al., Therapeutic target database update 2012: a resource for facilitating target-oriented drug discovery, Nucleic Acids Research, vol.6, issue.10, pp.1128-1164, 2012.
DOI : 10.1038/nrd2443

S. Rajagopalan and B. Pitt, Aldosterone antagonists in the treatment of hypertension and target organ damage. Current hypertension reports, pp.240-248, 2001.

C. Stier, J. Chander, P. Rocha, and R. , Aldosterone as a Mediator in Cardiovascular Injury, Cardiology in Review, vol.10, issue.2, pp.97-107, 2002.
DOI : 10.1097/00045415-200203000-00008

D. Bicket, Using ACE inhibitors appropriately, American family physician, vol.66, pp.461-469, 2002.

F. Spencer and R. Becker, Novel inhibitors of factor X for use in cardiovascular diseases, Current Cardiology Reports, vol.292, issue.Suppl 1, pp.395-404, 2000.
DOI : 10.1161/01.CIR.92.3.485

H. Lal, F. Ahmad, J. Woodgett, and T. Force, The GSK-3 family as therapeutic target for myocardial diseases. Circulation research, pp.138-187, 2015.

M. Jessup, B. Greenberg, D. Mancini, T. Cappola, D. Pauly et al., Calcium Upregulation by Percutaneous Administration of Gene Therapy in Cardiac Disease (CUPID): A Phase 2 Trial of Intracoronary Gene Therapy of Sarcoplasmic Reticulum Ca2+-ATPase in Patients With Advanced Heart Failure, Circulation, vol.124, issue.3, pp.304-317, 2011.
DOI : 10.1161/CIRCULATIONAHA.111.022889

M. Penn, F. Mendelsohn, G. Schaer, W. Sherman, M. Farr et al., An Open-Label Dose Escalation Study to Evaluate the Safety of Administration of Nonviral Stromal Cell-Derived Factor-1 Plasmid to Treat Symptomatic Ischemic Heart Failure, Circulation Research, vol.112, issue.5, pp.816-841, 2013.
DOI : 10.1161/CIRCRESAHA.111.300440

J. Wolfram and J. Donahue, Gene Therapy to Treat Cardiovascular Disease, Journal of the American Heart Association, vol.2, issue.4, p.119, 2013.
DOI : 10.1161/JAHA.113.000119

D. Prazeres, G. Ferreira, G. Monteiro, C. Cooney, and J. Cabral, Large-scale production of pharmaceutical-grade plasmid DNA for gene therapy: problems and bottlenecks, Trends in Biotechnology, vol.17, issue.4, pp.169-74, 1999.
DOI : 10.1016/S0167-7799(98)01291-8

G. Buchlis, G. Podsakoff, A. Radu, S. Hawk, A. Flake et al., Factor IX expression in skeletal muscle of a severe hemophilia B patient 10 years after AAV-mediated gene transfer, Blood, vol.119, issue.13, pp.3038-3079, 2012.
DOI : 10.1182/blood-2011-09-382317

A. Kass-eisler, E. Falck-pedersen, A. M. Rivera, J. Buttrick, P. Wittenberg et al., Quantitative determination of adenovirus-mediated gene delivery to rat cardiac myocytes in vitro and in vivo., Proceedings of the National Academy of Sciences, vol.90, issue.24, pp.11498-502, 1993.
DOI : 10.1073/pnas.90.24.11498

J. Donahue, K. Kikkawa, A. Thomas, E. Marban, and J. Lawrence, Acceleration of widespread adenoviral gene transfer to intact rabbit hearts by coronary perfusion with low calcium and serotonin, Gene Therapy, vol.5, issue.5, pp.630-634, 1998.
DOI : 10.1038/sj.gt.3300649

K. Kikuchi, A. Mcdonald, T. Sasano, and J. Donahue, Targeted Modification of Atrial Electrophysiology by Homogeneous Transmural Atrial Gene Transfer, Circulation, vol.111, issue.3, pp.264-70, 2005.
DOI : 10.1161/01.CIR.0000153338.47507.83

D. Comings, The structure and function of chromatin Advances in human genetics, pp.237-431, 1972.

V. Erdmann, M. Szymanski, A. Hochberg, N. De-groot, and J. Barciszewski, Collection of mRNA-like non-coding RNAs, Nucleic Acids Research, vol.32, issue.5, pp.192-197, 1999.
DOI : 10.1007/BF00020478

S. Frietze and J. Harrow, An integrated encyclopedia of DNA elements in the human genome, Nature, vol.489, pp.57-74, 2012.

T. Mercer, M. Dinger, and J. Mattick, Long non-coding RNAs: insights into functions, Nature Reviews Genetics, vol.72, issue.3
DOI : 10.1093/oxfordjournals.molbev.a003951

J. Lu, G. Getz, E. Miska, E. Alvarez-saavedra, J. Lamb et al., MicroRNA expression profiles classify human cancers, Nature, vol.1, issue.7043, pp.834-842, 2005.
DOI : 10.1016/S1535-6108(02)00018-1

G. Swaminathan, S. Navas-martin, M. , and J. , MicroRNAs and HIV-1 Infection: Antiviral Activities and Beyond, Journal of Molecular Biology, vol.426, issue.6, pp.1178-97, 2014.
DOI : 10.1016/j.jmb.2013.12.017

S. Maciotta, M. Meregalli, and Y. Torrente, The involvement of microRNAs in neurodegenerative diseases, Frontiers in Cellular Neuroscience, vol.7, p.265, 2013.
DOI : 10.3389/fncel.2013.00265

T. Thum, D. Catalucci, and J. Bauersachs, MicroRNAs: novel regulators in cardiac development and disease. Cardiovascular research, pp.562-70, 2008.

E. Bronze-da-rocha, MicroRNAs expression profiles in cardiovascular diseases BioMed research international, p.985408, 2014.

S. Vasudevan, Y. Tong, and J. Steitz, Switching from Repression to Activation: MicroRNAs Can Up-Regulate Translation, Science, vol.35, issue.7, pp.1931-1935, 2007.
DOI : 10.1093/nar/gkm133

J. Winter, S. Jung, S. Keller, R. Gregory, and S. Diederichs, Many roads to maturity: microRNA biogenesis pathways and their regulation, Nature Cell Biology, vol.11, issue.3, pp.228-262, 2009.
DOI : 10.1126/science.1138050

C. Mall, D. Rocke, B. Durbin-johnson, and R. Weiss, Stability of miRNA in human urine supports its biomarker potential, Biomarkers in Medicine, vol.3, issue.4, pp.623-654, 2013.
DOI : 10.1073/pnas.0403453101

P. Mitchell, R. Parkin, E. Kroh, B. Fritz, S. Wyman et al., Circulating microRNAs as stable blood-based markers for cancer detection, Proceedings of the National Academy of Sciences, vol.579, issue.26, pp.10513-10521, 2008.
DOI : 10.1016/j.febslet.2005.09.039

J. Winter and S. Diederichs, Argonaute proteins regulate microRNA stability: ???Increased microRNA abundance by Argonaute proteins is due to microRNA stabilization, RNA Biology, vol.8, issue.6, pp.1149-57, 2011.
DOI : 10.4161/rna.8.6.17665

J. Arroyo, J. Chevillet, E. Kroh, I. Ruf, C. Pritchard et al., Argonaute2 complexes carry a population of circulating microRNAs independent of vesicles in human plasma, Proceedings of the National Academy of Sciences, vol.16, issue.8, pp.5003-5011, 2011.
DOI : 10.1105/tpc.104.023614

K. Vickers, B. Palmisano, B. Shoucri, R. Shamburek, and A. Remaley, MicroRNAs are transported in plasma and delivered to recipient cells by high-density lipoproteins, Nature Cell Biology, vol.257, issue.4, pp.423-456, 2011.
DOI : 10.1073/pnas.011404098

A. Montecalvo, A. Larregina, W. Shufesky, D. Stolz, M. Sullivan et al., Mechanism of transfer of functional microRNAs between mouse dendritic cells via exosomes, Blood, vol.119, issue.3, pp.756-66, 2012.
DOI : 10.1182/blood-2011-02-338004

M. Hunter, N. Ismail, X. Zhang, B. Aguda, E. Lee et al., Detection of microRNA Expression in Human Peripheral Blood Microvesicles, PLoS ONE, vol.15, issue.11, p.3694, 2008.
DOI : 10.1371/journal.pone.0003694.s003

A. Zernecke, K. Bidzhekov, H. Noels, E. Shagdarsuren, L. Gan et al., Delivery of MicroRNA-126 by Apoptotic Bodies Induces CXCL12-Dependent Vascular Protection, Science Signaling, vol.2, issue.100, p.81, 2009.
DOI : 10.1126/scisignal.2000610

S. Gilad, E. Meiri, Y. Yogev, S. Benjamin, D. Lebanony et al., Serum MicroRNAs Are Promising Novel Biomarkers, PLoS ONE, vol.196, issue.2, p.3148, 2008.
DOI : 10.1371/journal.pone.0003148.t001

A. Caroli, M. Cardillo, R. Galea, and L. Biasucci, Potential therapeutic role of microRNAs in ischemic heart disease, Journal of Cardiology, vol.61, issue.5, pp.315-335, 2013.
DOI : 10.1016/j.jjcc.2013.01.012

R. Montgomery and E. Van-rooij, Therapeutic Advances in MicroRNA Targeting, Journal of Cardiovascular Pharmacology, vol.57, issue.1, pp.1-7, 2011.
DOI : 10.1097/FJC.0b013e3181f603d0

A. Grunweller, R. Hartmann, W. Marshall, and E. Olson, Locked nucleic acid oligonucleotides: the next generation of antisense agents? BioDrugs : clinical immunotherapeutics, biopharmaceuticals and gene therapy Toward microRNA-based therapeutics for heart disease: the sense in antisense. Circulation research, pp.235-43919, 2007.

S. Dangwal and T. Thum, microRNA therapeutics in cardiovascular disease models. Annual review of pharmacology and toxicology, pp.185-203, 2014.

M. Ebert, J. Neilson, and P. Sharp, MicroRNA sponges: competitive inhibitors of small RNAs in mammalian cells, Nature Methods, vol.18, issue.9, pp.721-727, 2007.
DOI : 10.1038/nmeth1079

P. Amaral, M. Clark, D. Gascoigne, M. Dinger, and J. Mattick, lncRNAdb: a reference database for long noncoding RNAs, Nucleic Acids Research, vol.104, issue.suppl_1, pp.146-51, 2011.
DOI : 10.1073/pnas.0702431104

C. Xie, J. Yuan, H. Li, M. Li, G. Zhao et al., NONCODEv4: exploring the world of long non-coding RNA genes, Nucleic Acids Research, vol.42, issue.D1, pp.98-103, 2014.
DOI : 10.1093/nar/gkt1057

H. Guo, N. Ingolia, J. Weissman, and D. Bartel, Mammalian microRNAs predominantly act to decrease target mRNA levels, Nature, vol.5, issue.7308, pp.835-840, 2010.
DOI : 10.1038/nature09267

URL : http://europepmc.org/articles/pmc2990499?pdf=render

L. Ma, V. Bajic, and Z. Zhang, On the classification of long non-coding RNAs, RNA Biology, vol.5, issue.6, pp.925-958, 2013.
DOI : 10.1126/scisignal.2000568

K. Pang, M. Frith, and J. Mattick, Rapid evolution of noncoding RNAs: lack of conservation does not mean lack of function, Trends in Genetics, vol.22, issue.1, pp.1-5, 2006.
DOI : 10.1016/j.tig.2005.10.003

A. Khalil, M. Guttman, M. Huarte, M. Garber, R. A. et al., Many human large intergenic noncoding RNAs associate with chromatin-modifying complexes and affect gene expression, Proceedings of the National Academy of Sciences, vol.106, issue.28, pp.11667-72, 2009.
DOI : 10.1101/gr.6036807

K. Cordes, N. Sheehy, M. White, E. Berry, S. Morton et al., miR-145 and miR-143 regulate smooth muscle cell fate and plasticity, Nature, vol.106, pp.705-715, 2009.
DOI : 10.1016/S0002-9440(10)64465-3

X. Liu, Y. Cheng, S. Zhang, Y. Lin, Y. J. Zhang et al., A Necessary Role of miR-221 and miR-222 in Vascular Smooth Muscle Cell Proliferation and Neointimal Hyperplasia, Circulation Research, vol.104, issue.4, pp.476-87, 2009.
DOI : 10.1161/CIRCRESAHA.108.185363

J. Krutzfeldt, N. Rajewsky, R. Braich, R. K. Tuschl, T. Manoharan et al., Silencing of microRNAs in vivo with ???antagomirs???, Nature, vol.33, issue.7068, pp.685-689, 2005.
DOI : 10.1093/nar/gki025

K. Wang, F. Liu, L. Zhou, S. Ding, B. Long et al., miR-874 regulates myocardial necrosis by targeting caspase-8, Cell Death & Disease, vol.4, issue.7, p.709, 2013.
DOI : 10.1038/nm.2282

E. Small and E. Olson, Pervasive roles of microRNAs in cardiovascular biology, Nature, vol.327, issue.7330, pp.336-378, 2011.
DOI : 10.1126/science.1178178

T. Thum, P. Galuppo, C. Wolf, J. Fiedler, S. Kneitz et al., MicroRNAs in the Human Heart: A Clue to Fetal Gene Reprogramming in Heart Failure, Circulation, vol.116, issue.3, pp.258-67, 2007.
DOI : 10.1161/CIRCULATIONAHA.107.687947

T. Thum, C. Gross, J. Fiedler, T. Fischer, S. Kissler et al., MicroRNA-21 contributes to myocardial disease by stimulating MAP kinase signalling in fibroblasts, Nature, vol.100, issue.7224, pp.980-984, 2008.
DOI : 10.1101/SQB.1997.062.01.021

R. Montgomery, T. Hullinger, H. Semus, B. Dickinson, A. Seto et al., Therapeutic Inhibition of miR-208a Improves Cardiac Function and Survival During Heart Failure, Circulation, vol.124, issue.14, pp.1537-1547, 2011.
DOI : 10.1161/CIRCULATIONAHA.111.030932

H. Seok, J. Chen, M. Kataoka, Z. Huang, J. Ding et al., Loss of MicroRNA-155 Protects the Heart From Pathological Cardiac Hypertrophy, Circulation Research, vol.114, issue.10, pp.1585-95, 2014.
DOI : 10.1161/CIRCRESAHA.114.303784

J. Fiedler, V. Jazbutyte, B. Kirchmaier, S. Gupta, J. Lorenzen et al., MicroRNA-24 Regulates Vascularity After Myocardial Infarction, Circulation, vol.124, issue.6, pp.720-750, 2011.
DOI : 10.1161/CIRCULATIONAHA.111.039008

B. Lewis, C. Burge, and D. Bartel, Conserved Seed Pairing, Often Flanked by Adenosines, Indicates that Thousands of Human Genes are MicroRNA Targets, Cell, vol.120, issue.1, pp.15-20, 2005.
DOI : 10.1016/j.cell.2004.12.035

T. Hullinger, R. Montgomery, A. Seto, B. Dickinson, H. Semus et al., Inhibition of miR-15 Protects Against Cardiac Ischemic InjuryNovelty and Significance, Circulation Research, vol.110, issue.1, pp.71-81, 2012.
DOI : 10.1161/CIRCRESAHA.111.244442

B. Bernardo, X. Gao, C. Winbanks, E. Boey, Y. Tham et al., Therapeutic inhibition of the miR-34 family attenuates pathological cardiac remodeling and improves heart function, Proceedings of the National Academy of Sciences, vol.7, issue.3, pp.17615-17620, 2012.
DOI : 10.1371/journal.pone.0033844

E. Goretti, J. Zangrando, D. Wagner, and Y. Devaux, Unity is strength ??? A panel of 4 microRNAs decreases cardiomyocyte hypertrophy, International Journal of Cardiology, vol.182, pp.62-66, 2015.
DOI : 10.1016/j.ijcard.2014.12.065

R. Hinkel, D. Penzkofer, S. Zuhlke, A. Fischer, W. Husada et al., Inhibition of MicroRNA-92a Protects Against Ischemia/Reperfusion Injury in a Large-Animal Model, Circulation, vol.128, issue.10, pp.1066-1075, 2013.
DOI : 10.1161/CIRCULATIONAHA.113.001904

S. Miyata, W. Minobe, M. Bristow, and L. Leinwand, Myosin Heavy Chain Isoform Expression in the Failing and Nonfailing Human Heart, Circulation Research, vol.86, issue.4, pp.386-90, 2000.
DOI : 10.1161/01.RES.86.4.386

E. Rooij, L. Sutherland, N. Liu, A. Williams, J. Mcanally et al., A signature pattern of stress-responsive microRNAs that can evoke cardiac hypertrophy and heart failure, Proceedings of the National Academy of Sciences, vol.26, issue.10, pp.18255-60, 2006.
DOI : 10.1128/MCB.26.10.3875-3888.2006

C. Sucharov, M. Bristow, and J. Port, miRNA expression in the failing human heart: Functional correlates, Journal of Molecular and Cellular Cardiology, vol.45, issue.2, pp.185-92, 2008.
DOI : 10.1016/j.yjmcc.2008.04.014

A. Tijsen, E. Creemers, P. Moerland, L. De-windt, A. Van-der-wal et al., MiR423-5p As a Circulating Biomarker for Heart Failure, Circulation Research, vol.106, issue.6, pp.1035-1044, 2010.
DOI : 10.1161/CIRCRESAHA.110.218297

F. Yue, Y. Cheng, A. Breschi, J. Vierstra, W. Wu et al., A comparative encyclopedia of DNA elements in the mouse genome, Nature, vol.46, issue.7527, pp.355-64, 2014.
DOI : 10.1038/ng.2965

C. Klattenhoff, J. Scheuermann, L. Surface, R. Bradley, P. Fields et al., Braveheart, a Long Noncoding RNA Required for Cardiovascular Lineage Commitment, Cell, vol.152, issue.3, pp.570-83, 2013.
DOI : 10.1016/j.cell.2013.01.003

L. Korostowski, N. Sedlak, and N. Engel, The Kcnq1ot1 Long Non-Coding RNA Affects Chromatin Conformation and Expression of Kcnq1, but Does Not Regulate Its Imprinting in the Developing Heart, PLoS Genetics, vol.8, issue.9, p.1002956, 2012.
DOI : 10.1371/journal.pgen.1002956.s009

N. Ishii, K. Ozaki, H. Sato, H. Mizuno, S. Saito et al., Identification of a novel non-coding RNA, MIAT, that confers risk of myocardial infarction, Journal of Human Genetics, vol.278, issue.12, pp.1087-99, 2006.
DOI : 10.1007/s10038-006-0070-9

J. 199-sheik-mohamed, P. Gaughwin, B. Lim, P. Robson, and L. Lipovich, Conserved long noncoding RNAs transcriptionally regulated by Oct4 and Nanog modulate pluripotency in mouse embryonic stem cells, RNA, vol.16, issue.2, pp.324-361, 2010.
DOI : 10.1261/rna.1441510

J. Aprea, S. Prenninger, M. Dori, T. Ghosh, L. Monasor et al., Transcriptome sequencing during mouse brain development identifies long non-coding RNAs functionally involved in neurogenic commitment, The EMBO Journal, vol.136, issue.24, pp.3145-60, 2013.
DOI : 10.1242/dev.022087

M. Vausort, D. Wagner, and Y. Devaux, Long noncoding RNAs in patients with acute myocardial infarction. Circulation research, pp.668-77, 2014.

O. Zolk, T. Solbach, T. Eschenhagen, A. Weidemann, and M. Fromm, Activation of negative regulators of the hypoxia-inducible factor (HIF) pathway in human end-stage heart failure. Biochemical and biophysical research communications, pp.315-320, 2008.

K. Yap, S. Li, A. Munoz-cabello, S. Raguz, L. Zeng et al., Molecular Interplay of the Noncoding RNA ANRIL and Methylated Histone H3 Lysine 27 by Polycomb CBX7 in Transcriptional Silencing of INK4a, Molecular Cell, vol.38, issue.5, pp.662-74, 2010.
DOI : 10.1016/j.molcel.2010.03.021

R. Mcpherson, A. Pertsemlidis, N. Kavaslar, A. Stewart, R. Roberts et al., A Common Allele on Chromosome 9 Associated with Coronary Heart Disease, Science, vol.106, issue.4, pp.1488-91, 2007.
DOI : 10.1038/ng1984

C. Consortium, P. Deloukas, S. Kanoni, C. Willenborg, M. Farrall et al., Large-scale association analysis identifies new risk loci for coronary artery disease, Nat Genet, vol.45, pp.25-33, 2013.

G. Bochenek, R. Hasler, E. Mokthari, N. Konig, I. Loos et al., The large non-coding RNA ANRIL, which is associated with atherosclerosis, periodontitis and several forms of cancer, regulates ADIPOR1, VAMP3 and C11ORF10, Human Molecular Genetics, vol.21, issue.22, 2013.
DOI : 10.1093/bioinformatics/bth457

N. Bokil, J. Baisden, D. Radford, and K. Summers, Molecular genetics of long QT syndrome, Molecular Genetics and Metabolism, vol.101, issue.1, pp.1-8, 2010.
DOI : 10.1016/j.ymgme.2010.05.011

Y. Liu, G. Li, H. Lu, W. Li, X. Li et al., Expression profiling and ontology analysis of long noncoding RNAs in post-ischemic heart and their implied roles in ischemia/reperfusion injury, Gene, vol.543, issue.1, pp.15-21, 2014.
DOI : 10.1016/j.gene.2014.04.016

K. Wang, B. Long, L. Zhou, F. Liu, Q. Zhou et al., CARL lncRNA inhibits anoxia-induced mitochondrial fission and apoptosis in cardiomyocytes by impairing miR-539-dependent PHB2 downregulation, Nature Communications, vol.69, p.3596, 2014.
DOI : 10.1158/0008-5472.CAN-08-2962

D. Li, G. Chen, Y. J. Fan, X. Gong, Y. Xu et al., Transcriptome Analysis Reveals Distinct Patterns of Long Noncoding RNAs in Heart and Plasma of Mice with Heart Failure, PLoS ONE, vol.28, issue.10, p.77938, 2013.
DOI : 10.1371/journal.pone.0077938.s006

K. Yang, K. Yamada, A. Patel, V. Topkara, I. George et al., Deep RNA Sequencing Reveals Dynamic Regulation of Myocardial Noncoding RNAs in Failing Human Heart and Remodeling With Mechanical Circulatory Support, Circulation, vol.129, issue.9, pp.1009-1021, 2014.
DOI : 10.1161/CIRCULATIONAHA.113.003863

X. Huang, T. Yuan, M. Tschannen, Z. Sun, H. Jacob et al., Characterization of human plasma-derived exosomal RNAs by deep sequencing, BMC Genomics, vol.14, issue.1, p.319, 2013.
DOI : 10.1093/bioinformatics/btm595

P. Han, W. Li, C. Lin, Y. J. Shang, C. Nurnberg et al., A long noncoding RNA protects the heart from pathological hypertrophy, Nature, vol.310, issue.7520, pp.102-108, 2014.
DOI : 10.1007/s11010-007-9683-3

R. Bell, X. Long, M. Lin, J. Bergmann, V. Nanda et al., Identification and initial functional characterization of a human vascular cellenriched long noncoding RNA. Arteriosclerosis, thrombosis, and vascular biology, pp.1249-59, 2014.

T. Thum and R. Kumarswamy, The smooth long noncoding RNA SENCR. Arteriosclerosis, thrombosis, and vascular biology, pp.1124-1129, 2014.

K. Michalik, X. You, Y. Manavski, A. Doddaballapur, M. Zornig et al., Long Noncoding RNA MALAT1 Regulates Endothelial Cell Function and Vessel GrowthNovelty and Significance, Circulation Research, vol.114, issue.9, pp.1389-97, 2014.
DOI : 10.1161/CIRCRESAHA.114.303265

D. Kelly, G. Cockerill, L. Ng, M. Thompson, S. Khan et al., Plasma matrix metalloproteinase-9 and left ventricular remodelling after acute myocardial infarction in man: a prospective cohort study, European Heart Journal, vol.28, issue.6, pp.711-719, 2007.
DOI : 10.1093/eurheartj/ehm003

J. Virag and R. Lust, Coronary Artery Ligation and Intramyocardial Injection in a Murine Model of Infarction, Journal of Visualized Experiments, issue.52, 2011.
DOI : 10.3791/2581

G. Segall, Assessment of myocardial viability by positron emission tomography, Nuclear Medicine Communications, vol.23, issue.4, pp.323-353, 2002.
DOI : 10.1097/00006231-200204000-00005

R. Slart, J. Bax, D. Van-veldhuisen, E. Van-der-wall, R. Dierckx et al., Imaging techniques in nuclear cardiology for the assessment of myocardial viability. The international journal of cardiovascular imaging, pp.63-80, 2006.

M. Phelps, E. Hoffman, N. Mullani, and M. Ter-pogossian, Application of annihilation coincidence detection to transaxial reconstruction tomography, J Nucl Med, vol.16, pp.210-234, 1975.

M. Bousquenaud, F. Maskali, S. Poussier, P. Marie, H. Boutley et al., Acipimox-enhanced (18)F-fluorodeoxyglucose positron emission tomography for characterizing and predicting early remodeling in the rat infarct model. The international journal of cardiovascular imaging, pp.1407-1415, 2012.

M. Cerqueira, N. Weissman, V. Dilsizian, A. Jacobs, S. Kaul et al., Standardized Myocardial Segmentation and Nomenclature for Tomographic Imaging of the Heart, Journal of Cardiovascular Magnetic Resonance, vol.4, issue.2, pp.539-581, 2002.
DOI : 10.1081/JCMR-120003946

P. Haaf, B. Drexler, T. Reichlin, R. Twerenbold, M. Reiter et al., High-Sensitivity Cardiac Troponin in the Distinction of Acute Myocardial Infarction From Acute Cardiac Noncoronary Artery Disease, Circulation, vol.126, issue.1, pp.31-40, 2012.
DOI : 10.1161/CIRCULATIONAHA.112.100867

K. Salic, D. Windt, and L. , MicroRNAs as biomarkers for myocardial infarction. Current atherosclerosis reports, pp.193-200, 2012.

S. Talwar, I. Squire, P. Downie, A. Mccullough, M. Campton et al., Profile of plasma N-terminal proBNP following acute myocardial infarction. Correlation with left ventricular systolic dysfunction, European Heart Journal, vol.21, issue.18, pp.1514-1535, 2000.
DOI : 10.1053/euhj.1999.2045

D. Sayed, C. Hong, I. Chen, J. Lypowy, and M. Abdellatif, MicroRNAs play an essential role in the development of cardiac hypertrophy. Circulation research, pp.416-440, 2007.

Y. Alessandra, P. Devanna, F. Limana, S. Straino, D. Carlo et al., Circulating microRNAs are new and sensitive biomarkers of myocardial infarction, European Heart Journal, vol.118, issue.22, pp.2765-73, 2010.
DOI : 10.1371/journal.pone.0002213

C. Widera, S. Gupta, J. Lorenzen, C. Bang, J. Bauersachs et al., Diagnostic and prognostic impact of six circulating microRNAs in acute coronary syndrome, Journal of Molecular and Cellular Cardiology, vol.51, issue.5, pp.872-875, 2011.
DOI : 10.1016/j.yjmcc.2011.07.011

O. Gidlof, J. Smith, K. Miyazu, P. Gilje, A. Spencer et al., Circulating cardio-enriched microRNAs are associated with long-term prognosis following myocardial infarction, BMC Cardiovascular Disorders, vol.51, issue.5, p.12, 2013.
DOI : 10.1016/j.yjmcc.2011.07.011

K. Thygesen, J. Alpert, H. White, A. Jaffe, F. Apple et al., Universal Definition of Myocardial Infarction, Circulation, vol.116, issue.22, pp.2634-53, 2007.
DOI : 10.1161/CIRCULATIONAHA.107.187397

S. Matsumoto, Y. Sakata, D. Nakatani, S. Suna, H. Mizuno et al., A subset of circulating microRNAs are predictive for cardiac death after discharge for acute myocardial infarction. Biochemical and biophysical research communications, pp.280-284, 2012.

S. Sadiq and D. Agranoff, Pooling serum samples may lead to loss of potential biomarkers in SELDI-ToF MS proteomic profiling., Proteome Science, vol.6, issue.1, p.16, 2008.
DOI : 10.1186/1477-5956-6-16

N. Zidar, E. Bostjancic, D. Glavac, and D. Stajer, MicroRNAs, innate immunity and ventricular rupture in human myocardial infarction. Disease markers, pp.259-65, 2011.

Y. Tang, Y. Wang, K. Park, Q. Hu, J. Teoh et al., MicroRNA-150 protects the mouse heart from ischaemic injury by regulating cell death, Cardiovascular Research, vol.45, issue.3, pp.387-97, 2015.
DOI : 10.1093/abbs/gmt067

A. Hsu, S. Chen, Y. Chang, H. Chen, and P. Chu, Systemic Approach to Identify Serum microRNAs as Potential Biomarkers for Acute Myocardial Infarction, BioMed Research International, vol.62, issue.3, p.418628, 2014.
DOI : 10.1097/FJC.0b013e31829b82e6

Y. Zhang, D. Liu, X. Chen, J. Li, L. Li et al., Secreted Monocytic miR-150 Enhances Targeted Endothelial Cell Migration, Molecular Cell, vol.39, issue.1, pp.133-144, 2010.
DOI : 10.1016/j.molcel.2010.06.010

S. Poussier, F. Maskali, N. Tran, C. Person, P. Maureira et al., ECG-triggered (18)F-fluorodeoxyglucose positron emission tomography imaging of the rat heart is dramatically enhanced by acipimox. European journal of nuclear medicine and molecular imaging, pp.1745-1750, 2010.

S. Motiwala, J. Szymonifka, A. Belcher, R. Weiner, A. Baggish et al., Serial measurement of galectin-3 in patients with chronic heart failure: results from the ProBNP Outpatient Tailored Chronic Heart Failure Therapy (PROTECT) study, European Journal of Heart Failure, vol.14, issue.10, pp.1157-1163, 2013.
DOI : 10.1161/CIRCHEARTFAILURE.112.000146

S. Allender, P. Scarborough, V. Peto, M. Rayner, J. Leal et al., European Cardiovascular Disease Statistics, 2008.

E. Lander, L. Linton, B. Birren, C. Nusbaum, M. Zody et al., Initial sequencing and analysis of the human genome, Nature, vol.6, issue.6822, pp.860-921, 2001.
DOI : 10.1089/cmb.1999.6.91

J. Venter, M. Adams, E. Myers, P. Li, R. Mural et al., The Sequence of the Human Genome, Science, vol.6, issue.5507, pp.1304-1351, 2001.
DOI : 10.1093/nar/28.1.33

URL : https://hal.archives-ouvertes.fr/hal-00465088

J. Cheng, P. Kapranov, J. Drenkow, S. Dike, S. Brubaker et al., Transcriptional Maps of 10 Human Chromosomes at 5-Nucleotide Resolution, Science, vol.308, issue.5725, pp.1149-1154, 2005.
DOI : 10.1126/science.1108625

E. Consortium, I. Dunham, A. Kundaje, S. Aldred, P. Collins et al., An integrated encyclopedia of DNA elements in the human genome, Nature, vol.463, issue.7414, pp.57-74, 2012.
DOI : 10.1038/nature08795

S. Djebali, C. Davis, A. Merkel, A. Dobin, T. Lassmann et al., Landscape of transcription in human cells, Nature, vol.474, issue.7414, pp.101-108, 2012.
DOI : 10.1038/nature10006

URL : https://hal.archives-ouvertes.fr/hal-01216755

H. Guo, N. Ingolia, J. Weissman, and D. Bartel, Mammalian microRNAs predominantly act to decrease target mRNA levels, Nature, vol.5, issue.7308, pp.835-840, 2010.
DOI : 10.1038/nature09267

C. Wahlestedt, Targeting long non-coding RNA to therapeutically upregulate gene expression, Nature Reviews Drug Discovery, vol.28, issue.6, pp.433-446, 2013.
DOI : 10.1038/onc.2008.373

P. Carninci, T. Kasukawa, S. Katayama, J. Gough, M. Frith et al., The transcriptional landscape of the mammalian genome, Science, vol.309, pp.1559-1563, 2005.

P. Kapranov, J. Cheng, S. Dike, D. Nix, R. Duttagupta et al., RNA Maps Reveal New RNA Classes and a Possible Function for Pervasive Transcription, Science, vol.4, issue.6, pp.1484-1488, 2007.
DOI : 10.1016/0959-437X(94)90066-3

C. Klattenhoff, J. Scheuermann, L. Surface, R. Bradley, P. Fields et al., Braveheart, a Long Noncoding RNA Required for Cardiovascular Lineage Commitment, Cell, vol.152, issue.3, pp.570-583, 2013.
DOI : 10.1016/j.cell.2013.01.003

P. Grote, L. Wittler, D. Hendrix, F. Koch, S. Wahrisch et al., The Tissue-Specific lncRNA Fendrr Is an Essential Regulator of Heart and Body Wall Development in the Mouse, Developmental Cell, vol.24, issue.2, pp.206-214, 2013.
DOI : 10.1016/j.devcel.2012.12.012

N. Ishii, K. Ozaki, H. Sato, H. Mizuno, S. Saito et al., Identification of a novel non-coding RNA, MIAT, that confers risk of myocardial infarction, Journal of Human Genetics, vol.278, issue.12, pp.1087-1099, 2006.
DOI : 10.1007/s10038-006-0070-9

D. Li, G. Chen, Y. J. Fan, X. Gong, Y. Xu et al., Transcriptome Analysis Reveals Distinct Patterns of Long Noncoding RNAs in Heart and Plasma of Mice with Heart Failure, PLoS ONE, vol.28, issue.10, p.77938, 2013.
DOI : 10.1371/journal.pone.0077938.s006

K. Yang, K. Yamada, A. Patel, V. Topkara, I. George et al., Deep RNA Sequencing Reveals Dynamic Regulation of Myocardial Noncoding RNAs in Failing Human Heart and Remodeling With Mechanical Circulatory Support, Circulation, vol.129, issue.9, pp.1009-1021, 2014.
DOI : 10.1161/CIRCULATIONAHA.113.003863

S. Poussier, F. Maskali, N. Tran, C. Person, P. Maureira et al., ECG-triggered 18F-fluorodeoxyglucose positron emission tomography imaging of the rat heart is dramatically enhanced by acipimox, European Journal of Nuclear Medicine and Molecular Imaging, vol.267, issue.6 Pt 1, pp.1745-1750, 2010.
DOI : 10.1161/CIRCULATIONAHA.106.645184

M. Cerqueira, N. Weissman, V. Dilsizian, A. Jacobs, S. Kaul et al., Standardized Myocardial Segmentation and Nomenclature for Tomographic Imaging of the Heart: A Statement for Healthcare Professionals From the Cardiac Imaging Committee of the Council on Clinical Cardiology of the American Heart Association, Circulation, vol.105, issue.4, pp.539-542, 2002.
DOI : 10.1161/hc0402.102975

G. Germano, H. Kiat, P. Kavanagh, M. Moriel, M. Mazzanti et al., Automatic quantification of ejection fraction from gated myocardial perfusion SPECT, J Nucl Med, vol.36, pp.2138-2147, 1995.

G. Smyth, J. Michaud, and H. Scott, Use of within-array replicate spots for assessing differential expression in microarray experiments, Bioinformatics, vol.8, issue.6, pp.2067-2075, 2005.
DOI : 10.1089/106652701753307520

W. Huber, A. Von-heydebreck, H. Sültmann, A. Poustka, and M. Vingron, Variance stabilization applied to microarray data calibration and to the quantification of differential expression, Bioinformatics, vol.18, issue.Suppl 1, pp.96-104, 2002.
DOI : 10.1093/bioinformatics/18.suppl_1.S96

K. Coombes, ClassDiscovery: Classes and methods for " class discovery " with microarrays or proteomics, 2009.

V. Tusher, R. Tibshirani, and G. Chu, Significance analysis of microarrays applied to the ionizing radiation response, Proceedings of the National Academy of Sciences, vol.57, issue.2, pp.5116-5121, 2001.
DOI : 10.2307/3579583

M. Eisen, P. Spellman, P. Brown, and D. Botstein, Cluster analysis and display of genome-wide expression patterns, Proceedings of the National Academy of Sciences, vol.24, issue.2, pp.14863-14868, 1998.
DOI : 10.1016/0092-8674(81)90326-3

W. Da-huang, B. Sherman, Q. Tan, J. Collins, W. Alvord et al., The DAVID Gene Functional Classification Tool: a novel biological module-centric algorithm to functionally analyze large gene lists, Genome Biology, vol.8, issue.9, p.183, 2007.
DOI : 10.1186/gb-2007-8-9-r183

P. Amaral, M. Clark, D. Gascoigne, M. Dinger, and J. Mattick, lncRNAdb: a reference database for long noncoding RNAs, Nucleic Acids Research, vol.104, issue.suppl_1, pp.146-151, 2011.
DOI : 10.1073/pnas.0702431104

K. Pruitt, T. Tatusova, and D. Maglott, NCBI Reference Sequence (RefSeq): a curated non-redundant sequence database of genomes, transcripts and proteins, Nucleic Acids Research, vol.33, issue.Database issue, pp.501-504, 2005.
DOI : 10.1093/nar/gki025

P. Flicek, M. Amode, D. Barrell, K. Beal, S. Brent et al., Ensembl 2011, Nucleic Acids Research, vol.15, issue.7, pp.800-806, 2011.
DOI : 10.1101/gr.3577405

C. Camacho, G. Coulouris, V. Avagyan, N. Ma, J. Papadopoulos et al., BLAST+: architecture and applications, BMC Bioinformatics, vol.10, issue.1, p.421, 2009.
DOI : 10.1186/1471-2105-10-421

P. Langfelder and S. Horvath, WGCNA: an R package for weighted correlation network analysis, BMC Bioinformatics, vol.9, issue.1, p.559, 2008.
DOI : 10.1186/1471-2105-9-559

S. Michelhaugh, L. Lipovich, J. Blythe, H. Jia, G. Kapatos et al., Mining Affymetrix microarray data for long non-coding RNAs: altered expression in the nucleus accumbens of heroin abusers, Journal of Neurochemistry, vol.38, issue.3, pp.459-466, 2011.
DOI : 10.1016/j.biocel.2006.05.004

S. Motiwala, J. Szymonifka, A. Belcher, R. Weiner, A. Baggish et al., Serial measurement of galectin-3 in patients with chronic heart failure: results from the ProBNP Outpatient Tailored Chronic Heart Failure Therapy (PROTECT) study, European Journal of Heart Failure, vol.14, issue.10, pp.1157-1163, 2013.
DOI : 10.1161/CIRCHEARTFAILURE.112.000146

A. Van-der-velde, L. Gullestad, T. Ueland, P. Aukrust, Y. Guo et al., Prognostic Value of Changes in Galectin-3 Levels Over Time in Patients With Heart Failure: Data From CORONA and COACH, Circulation: Heart Failure, vol.6, issue.2, pp.219-226, 2013.
DOI : 10.1161/CIRCHEARTFAILURE.112.000129

R. Weir, C. Petrie, C. Murphy, S. Clements, T. Steedman et al., Galectin-3 and Cardiac Function in Survivors of Acute Myocardial Infarction, Circulation: Heart Failure, vol.6, issue.3, pp.492-498, 2013.
DOI : 10.1161/CIRCHEARTFAILURE.112.000146