C. Yeh, M. Hsieh, Y. Hsieh, M. Chien, H. Chiou et al., Antimetastatic effects of norcantharidin on hepatocellular carcinoma by transcriptional inhibition of MMP-9 through modulation of NF-jB activity, PLoS ONE, vol.7, 2012.

L. Yan, N. Borregaard, L. Kjeldsen, and M. Moses, The High Molecular Weight Urinary Matrix Metalloproteinase (MMP) Activity Is a Complex of Gelatinase B/MMP-9 and Neutrophil Gelatinase-associated Lipocalin (NGAL): MODULATION OF MMP-9 ACTIVITY BY NGAL, Journal of Biological Chemistry, vol.276, issue.40, pp.37258-37265, 2001.
DOI : 10.1074/jbc.M106089200

J. Lee and D. Welch, Suppression of metastasis in human breast carcinoma MDA-MB-435 cells after transfection with the metastasis suppressor gene, KiSS-1, Cancer Res, vol.57, pp.2384-2387, 1997.

C. Takahashi, Z. Sheng, T. Horan, H. Kitayama, M. Maki et al., Regulation of matrix metalloproteinase-9 and inhibition of tumor invasion by the membrane-anchored glycoprotein RECK, Proceedings of the National Academy of Sciences, vol.95, issue.22, pp.13221-13226, 1998.
DOI : 10.1073/pnas.95.22.13221

J. Oh, R. Takahashi, S. Kondo, A. Mizoguchi, E. Adachi et al., The Membrane-Anchored MMP Inhibitor RECK Is a Key Regulator of Extracellular Matrix Integrity and Angiogenesis, Cell, vol.107, issue.6, pp.789-800, 2001.
DOI : 10.1016/S0092-8674(01)00597-9

S. Takagi, S. Simizu, and H. Osada, RECK Negatively Regulates Matrix Metalloproteinase-9 Transcription, Cancer Research, vol.69, issue.4, pp.1502-1508, 2009.
DOI : 10.1158/0008-5472.CAN-08-2635

P. Esteve, O. Robledo, E. Potworowski, and Y. St-pierre, Induced expression of MMP-9 in C6 glioma cells is inhibited by PDGF via a PI 3-kinase-dependent pathway, Biochemical and Biophysical Research Communications, vol.296, issue.4, pp.864-869, 2002.
DOI : 10.1016/S0006-291X(02)02008-9

A. Das, M. Fernandez-zapico, S. Cao, J. Yao, S. Fiorucci et al., Disruption of an SP2/KLF6 Repression Complex by SHP Is Required for Farnesoid X Receptor-induced Endothelial Cell Migration, Journal of Biological Chemistry, vol.281, issue.51, pp.39105-39113, 2006.
DOI : 10.1074/jbc.M607720200

A. Black, J. Black, and J. Azizkhan-clifford, Sp1 and kr??ppel-like factor family of transcription factors in cell growth regulation and cancer, Journal of Cellular Physiology, vol.21, issue.2, pp.143-160, 2001.
DOI : 10.1002/jcp.1111

F. Bouchard, S. Belanger, K. Biron-pain, and Y. St-pierre, EGR-1 activation by EGF inhibits MMP-9 expression and lymphoma growth, Blood, vol.116, issue.5, pp.759-766, 2009.
DOI : 10.1182/blood-2009-12-257030

URL : https://hal.archives-ouvertes.fr/pasteur-00819601

J. Ma, Z. Ren, Y. Ma, L. Xu, Y. Zhao et al., Targeted Knockdown of EGR-1 Inhibits IL-8 Production and IL-8-mediated Invasion of Prostate Cancer Cells through Suppressing EGR-1/NF-??B Synergy, Journal of Biological Chemistry, vol.284, issue.50, pp.34600-34606, 2009.
DOI : 10.1074/jbc.M109.016246

C. Waddington, The Epigenotype, International Journal of Epidemiology, vol.41, issue.1, pp.10-13, 2012.
DOI : 10.1093/ije/dyr184

A. Feinberg and B. Vogelstein, Hypomethylation distinguishes genes of some human cancers from their normal counterparts, Nature, vol.82, issue.5895, pp.89-92, 1983.
DOI : 10.1038/301089a0

C. Yoo and P. Jones, Epigenetic therapy of cancer: past, present and future, Nature Reviews Drug Discovery, vol.316, issue.1, pp.37-50, 1930.
DOI : 10.1038/nrd1930

M. Mayo, C. Denlinger, R. Broad, F. Yeung, E. Reilly et al., Ineffectiveness of Histone Deacetylase Inhibitors to Induce Apoptosis Involves the Transcriptional Activation of NF-??B through the Akt Pathway, Journal of Biological Chemistry, vol.278, issue.21, pp.18980-18989, 2003.
DOI : 10.1074/jbc.M211695200

Z. Ma, R. Shah, M. Chang, and E. Benveniste, Coordination of Cell Signaling, Chromatin Remodeling, Histone Modifications, and Regulator Recruitment in Human Matrix Metalloproteinase 9 Gene Transcription, Molecular and Cellular Biology, vol.24, issue.12, pp.5496-5509, 2004.
DOI : 10.1128/MCB.24.12.5496-5509.2004

M. Houde, G. De-bruyne, M. Bracke, M. Ingelman-sundberg, G. Skoglund et al., Differential regulation of gelatinase b and tissue-type plasminogen activator expression in human bowes melanoma cells, International Journal of Cancer, vol.62, issue.3, pp.395-400, 1993.
DOI : 10.1002/ijc.2910530309

J. Juarez, G. Clayman, M. Nakajima, K. Tanabe, H. Saya et al., Role and regulation of expression of 92-kDa type-IV collagenase (MMP-9) in 2 invasive squamous-cell-carcinoma cell lines of the oral cavity, International Journal of Cancer, vol.264, issue.1, pp.10-18, 1993.
DOI : 10.1002/ijc.2910550104

R. Hanemaaijer, P. Koolwijk, L. Le-clercq, W. De-vree, and V. Van-hinsbergh, Regulation of matrix metalloproteinase expression in human vein and microvascular endothelial cells. Effects of tumour necrosis factor ??, interleukin 1 and phorbol ester, Biochemical Journal, vol.296, issue.3, pp.803-809, 1993.
DOI : 10.1042/bj2960803

M. Fini, M. Girard, M. Matsubara, and J. Bartlett, Unique regulation of the matrix metalloproteinase, gelatinase B, Invest Ophthalmol Vis Sci, vol.36, pp.622-633, 1995.

Z. Ma, M. Chang, R. Shah, and E. Benveniste, Interferon-??-activated STAT-1?? suppresses MMP-9 gene transcription by sequestration of the coactivators CBP/p300, Journal of Leukocyte Biology, vol.78, issue.2, pp.515-523, 2005.
DOI : 10.1189/jlb.0205112

F. Kaneko, H. Saito, Y. Saito, K. Wakabayashi, N. Nakamoto et al., Downregulation of matrix-invasive potential of human liver cancer cells by type I interferon and a histone deacetylase inhibitor sodium butyrate, Int J Oncol, vol.24, pp.837-845, 2004.

S. Kuljaca, T. Liu, A. Tee, M. Haber, M. Norris et al., Enhancing the anti-angiogenic action of histone deacetylase inhibitors, Molecular Cancer, vol.6, issue.1, pp.68-78, 2007.
DOI : 10.1186/1476-4598-6-68

E. Mitmaker, N. Griff, R. Grogan, R. Sarkar, E. Kebebew et al., Modulation of matrix metalloproteinase activity in human thyroid cancer cell lines using demethylating agents and histone deacetylase inhibitors, Surgery, vol.149, issue.4, pp.504-511, 2011.
DOI : 10.1016/j.surg.2010.10.007

K. Lee, E. Choi, M. Kim, K. Kim, B. Jang et al., Inhibition of histone deacetylase activity down-regulates urokinase plasminogen activator and matrix metalloproteinase-9 expression in gastric cancer, Molecular and Cellular Biochemistry, vol.26, issue.4, pp.163-171, 2010.
DOI : 10.1007/s11010-010-0510-x

C. Estella, I. Herrer, S. Atkinson, A. Quinonero, S. Martinez et al., Inhibition of Histone Deacetylase Activity in Human Endometrial Stromal Cells Promotes Extracellular Matrix Remodelling and Limits Embryo Invasion, PLoS ONE, vol.94, issue.1, 2012.
DOI : 10.1371/journal.pone.0030508.s001

C. Reilly, M. Thomas, R. Gogal, . Jr, S. Olgun et al., The histone deacetylase inhibitor trichostatin A upregulates regulatory T cells and modulates autoimmunity in NZB/W F1 mice, Journal of Autoimmunity, vol.31, issue.2, pp.123-130, 2008.
DOI : 10.1016/j.jaut.2008.04.020

R. Vinodhkumar, Y. Song, V. Ravikumar, G. Ramakrishnan, and T. Devaki, Depsipeptide a histone deacetlyase inhibitor down regulates levels of matrix metalloproteinases 2 and 9 mRNA and protein expressions in lung cancer cells (A549), Chemico-Biological Interactions, vol.165, issue.3, 2007.
DOI : 10.1016/j.cbi.2006.12.012

M. Lai, C. Yang, T. Lin, F. Tsai, and W. Chen, Depsipeptide (FK228) inhibits growth of human prostate cancer cells, Urologic Oncology: Seminars and Original Investigations, vol.26, issue.2, pp.182-189, 2008.
DOI : 10.1016/j.urolonc.2007.01.020

D. Sinn, S. Kim, K. Chu, K. Jung, S. Lee et al., Valproic acid-mediated neuroprotection in intracerebral hemorrhage via histone deacetylase inhibition and transcriptional activation, Neurobiology of Disease, vol.26, issue.2, pp.464-472, 2007.
DOI : 10.1016/j.nbd.2007.02.006

Z. Wang, Y. Leng, L. Tsai, P. Leeds, and D. Chuang, Valproic Acid Attenuates Blood???Brain Barrier Disruption in a Rat Model of Transient Focal Cerebral Ischemia: The Roles of HDAC and MMP-9 Inhibition, Journal of Cerebral Blood Flow & Metabolism, vol.41, issue.1, pp.52-57, 0195.
DOI : 10.1080/10409230290771546

A. Vinh, T. Gaspari, H. Liu, L. Dousha, R. Widdop et al., A Novel Histone Deacetylase Inhibitor Reduces Abdominal Aortic Aneurysm Formation in Angiotensin II-Infused Apolipoprotein E-Deficient Mice, Journal of Vascular Research, vol.45, issue.2, pp.143-152, 2008.
DOI : 10.1159/000110041

H. Liu, M. Voso, D. Gumiero, J. Duong, J. Mckendrick et al., The anti-leukemic effect of a novel histone deacetylase inhibitor MCT-1 and 5-aza-cytidine involves augmentation of Nur77 and inhibition of MMP-9 expression, Int J Oncol, vol.34, pp.573-579, 2009.

H. Liu, P. Mayes, P. Perlmutter, J. Mckendrick, and A. Dear, The anti-leukemic effect and molecular mechanisms of novel hydroxamate and benzamide histone deacetylase inhibitors with 5-aza-cytidine, Int J Oncol, vol.38, pp.1421-1425, 2011.

Z. Zhang, Z. Zhang, and H. Schluesener, MS-275, an histone deacetylase inhibitor, reduces the inflammatory reaction in rat experimental autoimmune neuritis, Neuroscience, vol.169, issue.1, pp.370-377, 2010.
DOI : 10.1016/j.neuroscience.2010.04.074

M. Hsu, H. Chang, and W. Hung, HER-2/neu Represses the Metastasis Suppressor RECK via ERK and Sp Transcription Factors to Promote Cell Invasion, Journal of Biological Chemistry, vol.281, issue.8, pp.4718-4725, 2006.
DOI : 10.1074/jbc.M510937200

D. Bosc, B. Goueli, and R. Janknecht, HER2/Neu-mediated activation of the ETS transcription factor ER81 and its target gene MMP-1, Oncogene, vol.20, issue.43, pp.6215-6224, 2001.
DOI : 10.1038/sj.onc.1204820

G. Konecny, M. Untch, J. Arboleda, C. Wilson, S. Kahlert et al., Her-2/neu and urokinase-type plasminogen activator and its inhibitor in breast cancer, Clin Cancer Res, vol.7, pp.2448-2457, 2001.

E. Laughner, P. Taghavi, K. Chiles, P. Mahon, and G. Semenza, HER2 (neu) Signaling Increases the Rate of Hypoxia-Inducible Factor 1?? (HIF-1??) Synthesis: Novel Mechanism for HIF-1-Mediated Vascular Endothelial Growth Factor Expression, Molecular and Cellular Biology, vol.21, issue.12, pp.3995-4004, 2001.
DOI : 10.1128/MCB.21.12.3995-4004.2001

L. Howe, K. Subbaramaiah, J. Patel, J. Masferrer, A. Deora et al., Celecoxib, a selective cyclooxygenase 2 inhibitor, protects against human epidermal growth factor receptor 2 (HER-2)/neu-induced breast cancer, Cancer Res, vol.62, pp.5405-5407, 2002.

L. Rocca, G. Pucci-minafra, I. Marrazzo, A. Taormina, P. Minafra et al., Zymographic detection and clinical correlations of MMP-2 and MMP-9 in breast cancer sera, British Journal of Cancer, vol.90, issue.7, pp.1414-1421, 2004.
DOI : 10.1038/sj.bjc.6601725

H. Jeon and Y. Lee, Inhibition of Histone Deacetylase Attenuates Hypoxia-Induced Migration and Invasion of Cancer Cells via the Restoration of RECK Expression, Molecular Cancer Therapeutics, vol.9, issue.5, pp.1361-1370, 2010.
DOI : 10.1158/1535-7163.MCT-09-0717

M. Wanczyk, K. Roszczenko, K. Marcinkiewicz, K. Bojarczuk, M. Kowara et al., HDACi - going through the mechanisms, Frontiers in Bioscience, vol.16, issue.1, pp.340-359, 2011.
DOI : 10.2741/3691

P. Marks, Discovery and development of SAHA as an anticancer agent, Oncogene, vol.265, issue.9, pp.1351-1356, 2007.
DOI : 10.1038/nrd1930

Y. Takada, A. Gillenwater, H. Ichikawa, and B. Aggarwal, Suberoylanilide Hydroxamic Acid Potentiates Apoptosis, Inhibits Invasion, and Abolishes Osteoclastogenesis by Suppressing Nuclear Factor-??B Activation, Journal of Biological Chemistry, vol.281, issue.9, pp.5612-5622, 2006.
DOI : 10.1074/jbc.M507213200

Q. Choo, P. Ho, Y. Tanaka, and H. Lin, Histone deacetylase inhibitors MS-275 and SAHA induced growth arrest and suppressed lipopolysaccharide-stimulated NF-??B p65 nuclear accumulation in human rheumatoid arthritis synovial fibroblastic E11 cells, Rheumatology, vol.49, issue.8, pp.1447-1460, 2010.
DOI : 10.1093/rheumatology/keq108

E. Greer and Y. Shi, Histone methylation: a dynamic mark in health, disease and inheritance, Nature Reviews Genetics, vol.45, issue.5, pp.343-357, 2012.
DOI : 10.1038/nrg3173

E. Ballestar and M. Esteller, The Epigenetic Breakdown of Cancer Cells: From DNA Methylation to Histone Modifications, Prog Mol Subcell Biol, vol.38, pp.169-181, 2005.
DOI : 10.1007/3-540-27310-7_7

R. Varier and H. Timmers, Histone lysine methylation and demethylation pathways in cancer, Biochimica et Biophysica Acta (BBA) - Reviews on Cancer, vol.1815, issue.1, pp.75-89, 2011.
DOI : 10.1016/j.bbcan.2010.10.002

M. Balemans, M. Huibers, N. Eikelenboom, A. Kuipers, R. Van-summeren et al., Reduced exploration, increased anxiety, and altered social behavior: Autistic-like features of euchromatin histone methyltransferase 1 heterozygous knockout mice, Behavioural Brain Research, vol.208, issue.1, pp.47-55, 2010.
DOI : 10.1016/j.bbr.2009.11.008

T. Kleefstra, W. Van-zelst-stams, W. Nillesen, V. Cormier-daire, G. Houge et al., Further clinical and molecular delineation of the 9q subtelomeric deletion syndrome supports a major contribution of EHMT1 haploinsufficiency to the core phenotype, Journal of Medical Genetics, vol.46, issue.9, pp.598-606062950, 2008.
DOI : 10.1136/jmg.2008.062950

N. Kurotaki, K. Imaizumi, N. Harada, M. Masuno, T. Kondoh et al., Haploinsufficiency of NSD1 causes Sotos syndrome, Nature Genetics, vol.30, issue.4, pp.365-36610, 1038.
DOI : 10.1038/ng863

E. Pollina and A. Brunet, Epigenetic regulation of aging stem cells, Oncogene, vol.15, issue.28, pp.3105-3126, 2011.
DOI : 10.1038/onc.2011.45

A. Cock-rada, S. Medjkane, N. Janski, N. Yousfi, M. Perichon et al., SMYD3 Promotes Cancer Invasion by Epigenetic Upregulation of the Metalloproteinase MMP-9, Cancer Research, vol.72, issue.3, pp.810-820, 2012.
DOI : 10.1158/0008-5472.CAN-11-1052

J. Law and S. Jacobsen, Establishing, maintaining and modifying DNA methylation patterns in plants and animals, Nature Reviews Genetics, vol.28, issue.3, pp.204-220, 2010.
DOI : 10.1038/nrg2719

F. Mohn, M. Weber, M. Rebhan, T. Roloff, J. Richter et al., Lineage-Specific Polycomb Targets and De Novo DNA Methylation Define Restriction and Potential of Neuronal Progenitors, Molecular Cell, vol.30, issue.6, pp.755-766, 2008.
DOI : 10.1016/j.molcel.2008.05.007

A. Meissner, T. Mikkelsen, H. Gu, M. Wernig, J. Hanna et al., Genome-scale DNA methylation maps of pluripotent and differentiated cells, Nature, vol.6, pp.766-770, 2008.
DOI : 10.1038/nature07107

Y. Sun, H. Kim, M. Parker, W. Stetler-stevenson, and N. Colburn, Lack of suppression of tumor cell phenotype by overexpression of TIMP-3 in mouse JB6 tumor cells identification of a transfectant with increased tumorigenicity and invasiveness, Anticancer Res, vol.16, pp.1-7, 1996.

E. Chicoine, P. Esteve, O. Robledo, C. Van-themsche, E. Potworowski et al., Evidence for the role of promoter methylation in the regulation of MMP-9 gene expression, Biochemical and Biophysical Research Communications, vol.297, issue.4, pp.765-772, 2002.
DOI : 10.1016/S0006-291X(02)02283-0

N. Sato, N. Maehara, G. Su, and M. Goggins, Effects of 5-Aza-2'-deoxycytidine on Matrix Metalloproteinase Expression and Pancreatic Cancer Cell Invasiveness, JNCI Journal of the National Cancer Institute, vol.95, issue.4, pp.327-330, 2003.
DOI : 10.1093/jnci/95.4.327

W. Gallagher, O. Bergin, M. Rafferty, Z. Kelly, I. Nolan et al., Multiple markers for melanoma progression regulated by DNA methylation: insights from transcriptomic studies, Carcinogenesis, vol.26, issue.11, pp.1856-1867, 2005.
DOI : 10.1093/carcin/bgi152

I. Rhee, K. Jair, R. Yen, C. Lengauer, J. Herman et al., CpG methylation is maintained in human cancer cells lacking DNMT1, Nature, vol.404, pp.1003-100710, 1038.

J. Couillard, M. Demers, G. Lavoie, and Y. St-pierre, The role of DNA hypomethylation in the control of stromelysin gene expression, Biochemical and Biophysical Research Communications, vol.342, issue.4, pp.1233-1239, 2006.
DOI : 10.1016/j.bbrc.2006.02.068

K. Chen, Y. Wang, C. Hu, W. Chang, Y. Liao et al., OxLDL up-regulates microRNA-29b, leading to epigenetic modifications of MMP-2/MMP-9 genes: a novel mechanism for cardiovascular diseases, The FASEB Journal, vol.25, issue.5, pp.1718-172810, 2011.
DOI : 10.1096/fj.10-174904

H. Roach, N. Yamada, K. Cheung, S. Tilley, N. Clarke et al., Association between the abnormal expression of matrix-degrading enzymes by human osteoarthritic chondrocytes and demethylation of specific CpG sites in the promoter regions, Arthritis & Rheumatism, vol.23, issue.10, pp.3110-3124, 2005.
DOI : 10.1002/art.21300

M. Da-silva, N. Yamada, N. Clarke, and H. Roach, Cellular and epigenetic features of a young healthy and a young osteoarthritic cartilage compared with aged control and OA cartilage, Journal of Orthopaedic Research, vol.13, issue.5, pp.593-601, 2009.
DOI : 10.1002/jor.20799

P. Esteve, H. Chin, A. Smallwood, G. Feehery, O. Gangisetty et al., Direct interaction between DNMT1 and G9a coordinates DNA and histone methylation during replication, Genes & Development, vol.20, issue.22, pp.3089-3103, 2006.
DOI : 10.1101/gad.1463706

J. Couillard, P. Esteve, S. Pradhan, and Y. St-pierre, 5-Aza-2???-deoxycytidine and interleukin-1 cooperate to regulate matrix metalloproteinase-3 gene expression, International Journal of Cancer, vol.35, issue.9, pp.2083-2092, 2011.
DOI : 10.1002/ijc.25865

URL : https://hal.archives-ouvertes.fr/pasteur-00723259

G. Lavoie and Y. St-pierre, Phosphorylation of human DNMT1: Implication of cyclin-dependent kinases, Biochemical and Biophysical Research Communications, vol.409, issue.2, pp.187-192, 2011.
DOI : 10.1016/j.bbrc.2011.04.115

URL : https://hal.archives-ouvertes.fr/pasteur-00723263

T. Eiseler, H. Doppler, I. Yan, S. Goodison, and P. Storz, Protein kinase D1 regulates matrix metalloproteinase expression and inhibits breast cancer cell invasion, Breast Cancer Research, vol.283, issue.1, pp.10-1186, 2009.
DOI : 10.1074/jbc.M800264200

P. Storz, H. Doppler, and A. Toker, Protein Kinase D Mediates Mitochondrion-to-Nucleus Signaling and Detoxification from Mitochondrial Reactive Oxygen Species, Molecular and Cellular Biology, vol.25, issue.19, pp.8520-8530, 2005.
DOI : 10.1128/MCB.25.19.8520-8530.2005

M. Biswas, C. Du, C. Zhang, J. Straubhaar, L. Languino et al., Protein Kinase D1 Inhibits Cell Proliferation through Matrix Metalloproteinase-2 and Matrix Metalloproteinase-9 Secretion in Prostate Cancer, Cancer Research, vol.70, issue.5, pp.2095-2104, 2010.
DOI : 10.1158/0008-5472.CAN-09-4155

C. Ha, W. Wang, B. Jhun, C. Wong, A. Hausser et al., Protein Kinase D-dependent Phosphorylation and Nuclear Export of Histone Deacetylase 5 Mediates Vascular Endothelial Growth Factor-induced Gene Expression and Angiogenesis, Journal of Biological Chemistry, vol.283, issue.21, pp.14590-14599, 2008.
DOI : 10.1074/jbc.M800264200

R. Sasahara, S. Brochado, C. Takahashi, J. Oh, S. Maria-engler et al., Transcriptional control of the RECK metastasis/angiogenesis suppressor gene, Cancer Detection and Prevention, vol.26, issue.6, pp.435-443, 2002.
DOI : 10.1016/S0361-090X(02)00123-X

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

A. Mcdermott, H. Heneghan, N. Miller, and M. Kerin, The Therapeutic Potential of MicroRNAs: Disease Modulators and Drug Targets, Pharmaceutical Research, vol.65, issue.14, pp.3016-3029, 2011.
DOI : 10.1007/s11095-011-0550-2

J. Stanczyk, D. Pedrioli, F. Brentano, O. Sanchez-pernaute, C. Kolling et al., Altered expression of MicroRNA in synovial fibroblasts and synovial tissue in rheumatoid arthritis, Arthritis & Rheumatism, vol.179, issue.Suppl, pp.1001-1009, 2008.
DOI : 10.1002/art.23386

X. Li, G. Gibson, J. Kim, J. Kroin, S. Xu et al., MicroRNA-146a is linked to pain-related pathophysiology of osteoarthritis, Gene, vol.480, issue.1-2, pp.34-41, 2011.
DOI : 10.1016/j.gene.2011.03.003

K. Yamasaki, T. Nakasa, S. Miyaki, M. Ishikawa, M. Deie et al., Expression of MicroRNA-146a in osteoarthritis cartilage, Arthritis & Rheumatism, vol.13, issue.4, pp.1035-1041, 2009.
DOI : 10.1002/art.24404

K. Taganov, M. Boldin, K. Chang, and D. Baltimore, NF-??B-dependent induction of microRNA miR-146, an inhibitor targeted to signaling proteins of innate immune responses, Proceedings of the National Academy of Sciences, vol.103, issue.33, pp.12481-12486, 2006.
DOI : 10.1073/pnas.0605298103

N. Akhtar, Z. Rasheed, S. Ramamurthy, A. Anbazhagan, F. Voss et al., MicroRNA-27b regulates the expression of matrix metalloproteinase 13 in human osteoarthritis chondrocytes, Arthritis & Rheumatism, vol.456, issue.Suppl, pp.1361-1371, 2010.
DOI : 10.1002/art.27329

Z. Liang, Y. Li, K. Huang, N. Wagar, and H. Shim, Regulation of miR-19 to Breast Cancer Chemoresistance Through Targeting PTEN, Pharmaceutical Research, vol.286, issue.1, pp.3091-3100, 2011.
DOI : 10.1007/s11095-011-0570-y

G. Tardif, D. Hum, J. Pelletier, N. Duval, and J. Martel-pelletier, Regulation of the IGFBP-5 and MMP-13 genes by the microRNAs miR-140 and miR-27a in human osteoarthritic chondrocytes, BMC Musculoskeletal Disorders, vol.43, issue.1, pp.148-158, 2009.
DOI : 10.1002/1529-0131(200009)43:9<2100::AID-ANR22>3.0.CO;2-#

A. Ucar, V. Vafaizadeh, H. Jarry, J. Fiedler, P. Klemmt et al., miR-212 and miR-132 are required for epithelial stromal interactions necessary for mouse mammary gland development, Nature Genetics, vol.126, issue.12, pp.1101-1108, 2010.
DOI : 10.1016/j.bbrc.2006.06.025

K. Mayor-lynn, T. Toloubeydokhti, A. Cruz, and N. Chegini, Expression Profile of MicroRNAs and mRNAs in Human Placentas From Pregnancies Complicated by Preeclampsia and Preterm Labor, Reproductive Sciences, vol.22, issue.2, pp.46-56, 2011.
DOI : 10.1677/JOE-09-0136

W. Yan, W. Zhang, L. Sun, Y. Liu, G. You et al., Identification of MMP-9 specific microRNA expression profile as potential targets of anti-invasion therapy in glioblastoma multiforme, Brain Research, vol.1411, pp.108-115, 2011.
DOI : 10.1016/j.brainres.2011.07.002

P. Liu and M. Wilson, miR-520c and miR-373 upregulate MMP9 expression by targeting mTOR and SIRT1, and activate the Ras/Raf/MEK/Erk signaling pathway and NF-??B factor in human fibrosarcoma cells, Journal of Cellular Physiology, vol.28, issue.2, pp.867-876
DOI : 10.1002/jcp.22993

G. Gabriely, T. Wurdinger, S. Kesari, C. Esau, J. Burchard et al., MicroRNA 21 Promotes Glioma Invasion by Targeting Matrix Metalloproteinase Regulators, Molecular and Cellular Biology, vol.28, issue.17, pp.5369-5380, 2008.
DOI : 10.1128/MCB.00479-08

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2519720

L. Song, Q. Huang, K. Chen, L. Liu, C. Lin et al., miR-218 inhibits the invasive ability of glioma cells by direct downregulation of IKK-??, Biochemical and Biophysical Research Communications, vol.402, issue.1, pp.135-140, 2010.
DOI : 10.1016/j.bbrc.2010.10.003

P. Lopez-serra and M. Esteller, DNA methylation-associated silencing of tumor-suppressor microRNAs in cancer, Oncogene, vol.25, issue.13, 2012.
DOI : 10.1073/pnas.0508889103

J. Stanczyk, C. Ospelt, E. Karouzakis, A. Filer, K. Raza et al., Altered expression of microRNA-203 in rheumatoid arthritis synovial fibroblasts and its role in fibroblast activation, Arthritis & Rheumatism, vol.327, issue.Suppl 2, pp.373-381, 2011.
DOI : 10.1002/art.30115

S. Roy, S. Khanna, S. Hussain, S. Biswas, A. Azad et al., MicroRNA expression in response to murine myocardial infarction: miR-21 regulates fibroblast metalloprotease-2 via phosphatase and tensin homologue, Cardiovascular Research, vol.82, issue.1, pp.21-29, 2009.
DOI : 10.1093/cvr/cvp015

H. Liu, Y. Cao, W. Ye, and Y. Sun, Effect of microRNA- 206 on cytoskeleton remodelling by downregulating Cdc42 in MDA-MB-231 cells, Tumori, vol.96, pp.751-755, 2010.

E. Giovannetti, N. Funel, G. Peters, D. Chiaro, M. Erozenci et al., MicroRNA-21 in Pancreatic Cancer: Correlation with Clinical Outcome and Pharmacologic Aspects Underlying Its Role in the Modulation of Gemcitabine Activity, Cancer Research, vol.70, issue.11, pp.4528-4538, 2010.
DOI : 10.1158/0008-5472.CAN-09-4467

J. Chan, A. Krichevsky, and K. Kosik, MicroRNA-21 Is an Antiapoptotic Factor in Human Glioblastoma Cells, Cancer Research, vol.65, issue.14, pp.6029-6033, 2005.
DOI : 10.1158/0008-5472.CAN-05-0137

S. Reis, J. Pontes-junior, A. Antunesnes, D. Oglio, M. Dip et al., miR-21 may acts as an oncomir by targeting RECK, a matrix metalloproteinase regulator, in prostate cancer, BMC Urology, vol.107, issue.1, pp.14-24, 2012.
DOI : 10.1016/S0092-8674(01)00597-9

S. Volinia, G. Calin, C. Liu, S. Ambs, A. Cimmino et al., A microRNA expression signature of human solid tumors defines cancer gene targets, Proceedings of the National Academy of Sciences, vol.103, issue.7, pp.2257-2261, 2006.
DOI : 10.1073/pnas.0510565103

L. Silverman and G. Mufti, Methylation inhibitor therapy in the treatment of myelodysplastic syndrome, Nature Clinical Practice Oncology, vol.23, pp.12-23, 2005.
DOI : 10.1038/ncponc0347

J. Issa, H. Kantarjian, and P. Kirkpatrick, Fresh from the pipeline: Azacitidine, Nature Reviews Drug Discovery, vol.42, issue.4, pp.275-276, 2005.
DOI : 10.1182/blood-2003-03-0687

P. Fenaux, G. Mufti, E. Hellstrom-lindberg, V. Santini, C. Finelli et al., Efficacy of azacitidine compared with that of conventional care regimens in the treatment of higher-risk myelodysplastic syndromes: a randomised, open-label, phase III study, The Lancet Oncology, vol.10, issue.3, pp.223-232, 2009.
DOI : 10.1016/S1470-2045(09)70003-8

P. Garg, D. Sarma, S. Jeppsson, N. Patel, A. Gewirtz et al., Matrix Metalloproteinase-9 Functions as a Tumor Suppressor in Colitis-Associated Cancer, Cancer Research, vol.70, issue.2, pp.792-801, 2010.
DOI : 10.1158/0008-5472.CAN-09-3166

P. Garg, A. Ravi, N. Patel, J. Roman, A. Gewirtz et al., Matrix Metalloproteinase-9 Regulates MUC-2 Expression Through Its Effect on Goblet Cell Differentiation, Gastroenterology, vol.132, issue.5, pp.1877-1889, 2007.
DOI : 10.1053/j.gastro.2007.02.048

C. Overall and R. Dean, Degradomics: Systems biology of the protease web. Pleiotropic roles of MMPs in cancer, Cancer and Metastasis Reviews, vol.383, issue.1, pp.69-75, 2006.
DOI : 10.1007/s10555-006-7890-0

M. Fanjul-fernandez, A. Folgueras, S. Cabrera, and C. Lopez-otin, Matrix metalloproteinases: Evolution, gene regulation and functional analysis in mouse models, Biochimica et Biophysica Acta (BBA) - Molecular Cell Research, vol.1803, issue.1, 2010.
DOI : 10.1016/j.bbamcr.2009.07.004

N. Emenaker and M. Basson, Short Chain Fatty Acids Inhibit Human (SW1116) Colon Cancer Cell Invasion by Reducing Urokinase Plasminogen Activator Activity and Stimulating TIMP-1 and TIMP-2 Activities, Rather Than via MMP Modulation, Journal of Surgical Research, vol.76, issue.1, pp.41-465279, 1998.
DOI : 10.1006/jsre.1998.5279

H. Zeng and M. Briske-anderson, Prolonged butyrate treatment inhibits the migration and invasion potential of HT1080 tumor cells, J Nutr, vol.1352, pp.291-295, 2005.

J. Lee, M. Maa, H. Yu, J. Wang, C. Yen et al., Butyrate regulates the expression of c-Src and focal adhesion kinase and inhibits cell invasion of human colon cancer cells, Molecular Carcinogenesis, vol.273, issue.4, pp.207-214, 2005.
DOI : 10.1002/mc.20117

J. Rodriguez-salvador, C. Armas-pineda, M. Perezpena-diazconti, F. Chico-ponce-de-leon, G. Sosa-sainz et al., Effect of sodium butyrate on pro-matrix metalloproteinase-9 and -2 differential secretion in pediatric tumors and cell lines, J Exp Clin Cancer Res, vol.24, pp.463-473, 2005.

Y. Chen, Y. Tsai, and S. Tseng, Valproic acid affected the survival and invasiveness of human glioma cells through diverse mechanisms, Journal of Neuro-Oncology, vol.107, issue.1, 2012.
DOI : 10.1007/s11060-012-0871-y

D. Klisovic, M. Klisovic, D. Effron, S. Liu, G. Marcucci et al., Depsipeptide inhibits migration of primary and metastatic uveal melanoma cell lines in vitro: a potential strategy for uveal melanoma, Melanoma Research, vol.15, issue.3, pp.147-153, 2005.
DOI : 10.1097/00008390-200506000-00002

Y. Sun, G. Hegamyer, H. Kim, K. Sithanandam, H. Li et al., Molecular cloning of mouse tissue inhibitor of metalloproteinases-3 and its promoter. Specific lack of expression in neoplastic JB6 cells may reflect altered gene methylation, J Biol Chem, vol.270, pp.19312-19319, 1995.

F. Xu, C. T. Li, M. Sanchez-sweatman, O. Khokha, R. Gorelik et al., Inhibition of VLA-4 and up-regulation of TIMP-1 expression in B16BL6 melanoma cells transfected with MHC class I genes, Clinical and Experimental Metastasis, vol.16, issue.4, pp.358-370, 1998.
DOI : 10.1023/A:1006569631330

E. Cameron, K. Bachman, S. Myohanen, J. Herman, and S. Baylin, Synergy of demethylation and histone deacetylase inhibition in the re-expression of genes silenced in cancer, Nature Genetics, vol.21, issue.1, pp.103-107105047, 1038.
DOI : 10.1038/5047

W. Pennie, G. Hegamyer, M. Young, and N. Colburn, Specific methylation events contribute to the transcriptional repression of the mouse tissue inhibitor of metalloproteinases-3 gene in neoplastic cells, Cell Growth Differ, vol.10, pp.279-286, 1999.

X. Huang, A. Orucevic, M. Li, and E. Gorelik, Nitric oxide (NO), methylation and TIMP-1 expression in BL6 melanoma cells transfected with MHC class I genes, Clinical and Experimental Metastasis, vol.18, issue.4, pp.329-335, 2000.
DOI : 10.1023/A:1010867618014

T. Ivanova, S. Vinokurova, A. Petrenko, E. Eshilev, N. Solovyova et al., Frequent hypermethylation of 5? flanking region ofTIMP-2 gene in cervical cancer, International Journal of Cancer, vol.9, issue.6, pp.882-886, 2004.
DOI : 10.1002/ijc.11652

K. Bachman, J. Herman, P. Corn, A. Merlo, J. Costello et al., Methylation-associated silencing of the tissue inhibitor of metalloproteinase-3 gene suggest a suppressor role in kidney, brain, and other human cancers, Cancer Res, vol.59, pp.798-802, 1999.

J. Macdougall, M. Bani, Y. Lin, R. Muschel, and R. Kerbel, ???Proteolytic switching???: opposite patterns of regulation of gelatinase B and its inhibitor TIMP-1 during human melanoma progression and consequences of gelatinase B overexpression, British Journal of Cancer, vol.80, issue.3-4, 1999.
DOI : 10.1038/sj.bjc.6690385

J. Gagnon, S. Shaker, M. Primeau, A. Hurtubise, and R. Momparler, Interaction of 5-aza-2???-deoxycytidine and depsipeptide on antineoplastic activity and activation of 14-3-3??, E-cadherin and tissue inhibitor of metalloproteinase 3 expression in human breast carcinoma cells, Anti-Cancer Drugs, vol.14, issue.3, pp.193-202, 2003.
DOI : 10.1097/00001813-200303000-00002

B. Yuan, A. Jefferson, N. Popescu, and S. Reynolds, Aberrant Gene Expression in Human Non Small Cell Lung Carcinoma Cells Exposed to Demethylating Agent 5-Aza-2'-Deoxycytidine, Neoplasia, vol.6, issue.4, pp.412-419, 2004.
DOI : 10.1593/neo.03490

H. Feng, A. Cheung, W. Xue, Y. Wang, X. Wang et al., Down-regulation and promoter methylation of tissue inhibitor of metalloproteinase 3 in choriocarcinoma, Gynecologic Oncology, vol.94, issue.2, pp.375-382, 2004.
DOI : 10.1016/j.ygyno.2004.04.019

O. Galm, H. Suzuki, Y. Akiyama, M. Esteller, M. Brock et al., Inactivation of the tissue inhibitor of metalloproteinases-2 gene by promoter hypermethylation in lymphoid malignancies, Oncogene, vol.95, issue.30, pp.4799-4805, 2005.
DOI : 10.1038/sj.onc.1208599

S. Pulukuri, S. Patibandla, J. Patel, N. Estes, and J. Rao, Epigenetic inactivation of the tissue inhibitor of metalloproteinase-2 (TIMP-2) gene in human prostate tumors, Oncogene, vol.28, issue.36, pp.5229-5237, 2007.
DOI : 10.1038/sj.onc.1210329