M. Nunes, W. Dones, C. Morillo, J. Encina, and A. Ribeiro, Chagas Disease, Journal of the American College of Cardiology, vol.62, issue.9, pp.767-76, 2013.
DOI : 10.1016/j.jacc.2013.05.046

L. Abel, L. Rizzo, and B. Ianni, Chronic Chagas' Disease Cardiomyopathy Patients Display an Increased IFN-?? Response to Trypanosoma cruzi Infection, Journal of Autoimmunity, vol.17, issue.1, pp.99-107, 2001.
DOI : 10.1006/jaut.2001.0523

E. Cunha-neto, L. Nogueira, and P. Teixeira, Immunological and non-immunological effects of cytokines and chemokines in the pathogenesis of chronic Chagas disease cardiomyopathy, Mem??rias do Instituto Oswaldo Cruz, vol.43, issue.suppl 1, pp.252-260, 2009.
DOI : 10.4269/ajtmh.1990.43.498

L. Nogueira, R. Santos, and B. Ianni, Myocardial Chemokine Expression and Intensity of Myocarditis in Chagas Cardiomyopathy Are Controlled by Polymorphisms in CXCL9 and CXCL10, PLoS Neglected Tropical Diseases, vol.93, issue.1, p.1867, 2012.
DOI : 10.1371/journal.pntd.0001867.s012

L. Nogueira, R. Santos, and A. Fiorelli, 1-Type Response, Mediators of Inflammation, vol.159, issue.12, p.914326, 2014.
DOI : 10.1371/journal.pone.0083446

E. Cunha-neto and C. Chevillard, Chagas Disease Cardiomyopathy: Immunopathology and Genetics, Mediators of Inflammation, vol.148, issue.6, p.683230, 2014.
DOI : 10.1016/j.vetimm.2010.06.010

URL : https://hal.archives-ouvertes.fr/hal-01592694

L. Rodriguez, D. Carmona, F. Echeverría, L. González, C. Martin et al., IL18 Gene Variants Influence the Susceptibility to Chagas Disease, PLOS Neglected Tropical Diseases, vol.153, issue.3, p.4583, 2016.
DOI : 10.1371/journal.pntd.0004583.s002

L. Rodriguez, D. Carmona, F. González, C. Martin, and J. , Evaluation of VDR gene polymorphisms in Trypanosoma cruzi infection and chronic Chagasic cardiomyopathy, Scientific Reports, vol.38, issue.1, p.31263, 2016.
DOI : 10.1038/ng1706

F. Zicker, P. Smith, J. Netto, R. Oliveira, and E. Zicker, Physical Activity, Opportunity for Reinfection, and Sibling History of Heart Disease as Risk Factors for Chagas' Cardiopathy, The American Journal of Tropical Medicine and Hygiene, vol.43, issue.5, pp.498-505, 1990.
DOI : 10.4269/ajtmh.1990.43.498

E. Cunha-neto, V. Dzau, and P. Allen, Cardiac Gene Expression Profiling Provides Evidence for Cytokinopathy as a Molecular Mechanism in Chagas' Disease Cardiomyopathy, The American Journal of Pathology, vol.167, issue.2, pp.305-318, 2005.
DOI : 10.1016/S0002-9440(10)62976-8

J. Zhou, L. Yang, and T. Zhong, H19 lncRNA alters DNA methylation genome wide by regulating S-adenosylhomocysteine hydrolase, Nature Communications, vol.104, issue.1, p.10221, 2015.
DOI : 10.1016/j.ygeno.2014.08.020

URL : http://www.nature.com/articles/ncomms10221.pdf

L. Ferreira, A. Frade, and R. Santos, MicroRNAs miR-1, miR-133a, miR-133b, miR-208a and miR-208b are dysregulated in Chronic Chagas disease Cardiomyopathy, International Journal of Cardiology, vol.175, issue.3, pp.409-426, 2014.
DOI : 10.1016/j.ijcard.2014.05.019

URL : https://hal.archives-ouvertes.fr/hal-01592693

A. Frade, L. Laugier, and L. Ferreira, Myocardial Infarction???Associated Transcript, a Long Noncoding RNA, Is Overexpressed During Dilated Cardiomyopathy Due to Chronic Chagas Disease, Journal of Infectious Diseases, vol.91, issue.1, pp.161-166, 2016.
DOI : 10.1161/CIRCRESAHA.116.305510

URL : https://hal.archives-ouvertes.fr/hal-01592691

I. Navarro, F. Ferreira, and H. Nakaya, MicroRNA Transcriptome Profiling in Heart of Trypanosoma cruzi-Infected Mice: Parasitological and Cardiological Outcomes, PLOS Neglected Tropical Diseases, vol.71, issue.6, p.3828, 2015.
DOI : 10.1371/journal.pntd.0003828.s004

URL : https://hal.archives-ouvertes.fr/hal-01207672

B. Duygu, E. Poels, C. Da, and P. Martins, Genetics and epigenetics of arrhythmia and heart failure, Frontiers in Genetics, vol.4, p.219, 2013.
DOI : 10.3389/fgene.2013.00219

J. Haas, K. Frese, and Y. Park, Alterations in cardiac DNA methylation in human dilated cardiomyopathy, EMBO Molecular Medicine, vol.90, issue.(web server iss, pp.413-442, 2013.
DOI : 10.1093/cvr/cvr003

B. Jo, I. Koh, and J. Bae, Methylome analysis reveals alterations in DNA methylation in the regulatory regions of left ventricle development genes in human dilated cardiomyopathy, Genomics, vol.108, issue.2, pp.84-92, 2016.
DOI : 10.1016/j.ygeno.2016.07.001

M. Davidson, Novel immortalized human ventricular cardiomyocyte cell line, designated AC16. US patent 7223599 Available at: http://google, 2005.

F. Dorri, L. Mendelowitz, C. Bravo, and H. , methylFlow: cell-specific methylation pattern reconstruction from high-throughput bisulfite-converted DNA sequencing, Bioinformatics, vol.32, issue.11, pp.1618-1642, 2016.
DOI : 10.1186/s13059-014-0419-x

URL : http://europepmc.org/articles/pmc4892417?pdf=render

F. Dotiwala, S. Mulik, and R. Polidoro, Killer lymphocytes use granulysin, perforin and granzymes to kill intracellular parasites, Nature Medicine, vol.116, issue.2, pp.210-216, 2016.
DOI : 10.1182/blood-2009-10-246116

URL : http://doc.rero.ch/record/258966/files/wal_klu.pdf

H. Seo, I. Lee, and J. Park, Role of Protein Tyrosine Phosphatase Non-Receptor Type 7 in the Regulation of TNF-?? Production in RAW 264.7 Macrophages, PLoS ONE, vol.430, issue.11, p.78776, 2013.
DOI : 10.1371/journal.pone.0078776.t002

D. Reis, E. Jones, and S. Tostes, Expression of Major Histocompatibility Complex Antigens and Adhesion Molecules in Hearts of Patients with Chronic Chagas' Disease, The American Journal of Tropical Medicine and Hygiene, vol.49, issue.2, pp.192-200, 1993.
DOI : 10.4269/ajtmh.1993.49.192

N. Deghaide, R. Dantas, and E. Donadi, HLA class I and II profiles of patients presenting with Chagas' disease, Digestive Diseases and Sciences, vol.43, issue.2, pp.246-52, 1998.
DOI : 10.1023/A:1018829600200

Q. Zou, J. J. Xiao, and Y. , T cell development involves TRAF3IP3-mediated ERK signaling in the Golgi, The Journal of Experimental Medicine, vol.159, issue.8, pp.1323-1359, 2015.
DOI : 10.1038/ni.2885

J. Cruz, A. Santos-miranda, and P. Sales-junior, Altered Cardiomyocyte Function and Trypanosoma cruzi Persistence in Chagas Disease, The American Journal of Tropical Medicine and Hygiene, vol.94, issue.5, pp.1028-1061, 2016.
DOI : 10.4269/ajtmh.15-0255

D. Roman-campos, P. Sales-júnior, and H. Duarte, Cardiomyocyte dysfunction during the chronic phase of Chagas disease, Mem??rias do Instituto Oswaldo Cruz, vol.98, issue.5273, pp.243-248, 2013.
DOI : 10.1161/01.RES.0000218989.52072.e7

J. Greenstein, R. Wu, S. Po, G. Tomaselli, and R. Winslow, Role of the Calcium-Independent Transient Outward Current Ito1 in Shaping Action Potential Morphology and Duration, Circulation Research, vol.87, issue.11, pp.1026-1059, 2000.
DOI : 10.1161/01.RES.87.11.1026

W. Guo, H. Li, B. London, and J. Nerbonne, Functional Consequences of Elimination of Ito, f and Ito, s : Early Afterdepolarizations, Atrioventricular Block, and Ventricular Arrhythmias in Mice Lacking Kv1.4 and Expressing a Dominant-Negative Kv4 ?? Subunit, Circulation Research, vol.87, issue.1, pp.73-82, 2000.
DOI : 10.1161/01.RES.87.1.73

L. Bouter, S. , E. Harchi, A. Marionneau, and C. , Long-Term Amiodarone Administration Remodels Expression of Ion Channel Transcripts in the Mouse Heart, Circulation, vol.110, issue.19, pp.3028-3063, 2004.
DOI : 10.1161/01.CIR.0000147187.78162.AC

K. Rhodes, K. Carroll, and M. Sung, KChIPs and Kv4 ?? Subunits as Integral Components of A-Type Potassium Channels in Mammalian Brain, Journal of Neuroscience, vol.24, issue.36, pp.7903-7918, 2004.
DOI : 10.1523/JNEUROSCI.0776-04.2004

Y. Tang, P. Liang, and J. Zhou, Current through Endoplasmic Reticulum (ER) Retention and Promoting Closed-state Inactivation of Kv4 Channels, Journal of Biological Chemistry, vol.77, issue.21, pp.14727-14768, 2013.
DOI : 10.1001/archneur.64.2.185

S. Birlea, K. Gowan, P. Fain, and R. Spritz, Genome-Wide Association Study of Generalized Vitiligo in an Isolated European Founder Population Identifies SMOC2, in Close Proximity to IDDM8, Journal of Investigative Dermatology, vol.130, issue.3, pp.798-803, 2010.
DOI : 10.1038/jid.2009.347

C. Gerarduzzi, R. Kumar, and P. Trivedi, Silencing SMOC2 ameliorates kidney fibrosis by inhibiting fibroblast to myofibroblast transformation, JCI Insight, vol.2, issue.8, 2017.
DOI : 10.1172/jci.insight.90299DS1

URL : http://europepmc.org/articles/pmc5396522?pdf=render

L. Ng, I. Squire, and D. Jones, Proenkephalin, Renal Dysfunction, and??Prognosis in Patients With Acute??Heart??Failure, Journal of the American College of Cardiology, vol.69, issue.1, pp.56-69, 2017.
DOI : 10.1016/j.jacc.2016.10.038

X. Liang, R. Liu, C. Chen, J. F. Li, and T. , Opioid system modulates the immune function: a review, Transl Perioper Pain Med, vol.1, pp.5-13, 2016.

A. Karaji, D. Reiss, A. Matifas, B. Kieffer, and C. Gavériaux-ruff, Influence of Endogenous Opioid Systems on T Lymphocytes as Assessed by the Knockout of Mu, Delta and Kappa Opioid Receptors, Journal of Neuroimmune Pharmacology, vol.13, issue.4, pp.608-624, 2011.
DOI : 10.1016/S0376-8716(97)00059-8

P. Majumder and J. Boss, DNA methylation dysregulates and silences the HLA-DQ locus by altering chromatin architecture, Genes & Immunity, vol.132, issue.4, pp.291-300, 2011.
DOI : 10.3324/haematol.2008.000752

H. Ko, S. Chen, H. Chen, H. Hao, and F. Liu, Cell type-selective expression of the zinc finger-containing gene Nolz-1/Zfp503 in the developing mouse striatum, Neuroscience Letters, vol.548, pp.44-53, 2013.
DOI : 10.1016/j.neulet.2013.05.020

B. Majello, D. Luca, P. Suske, G. Lania, and L. , Differential transcriptional regulation of c-myc promoter through the same DNA binding sites targeted by Sp1-like proteins, Oncogene, vol.10, pp.1841-1849, 1995.

S. Safe, P. Imanirad, S. Sreevalsan, V. Nair, and I. Jutooru, Transcription factor Sp1, also known as specificity protein 1 as a therapeutic target, Expert Opinion on Therapeutic Targets, vol.28, issue.6, pp.759-69, 2014.
DOI : 10.1159/000324278