A. Gabory, H. Jammes, and L. Dandolo, The H19 locus: Role of an imprinted non-coding RNA in growth and development, BioEssays, vol.136, issue.6, pp.473-480, 2010.
DOI : 10.1002/bies.200900170

T. Yoshimizu, A. Miroglio, M. A. Ripoche, A. Gabory, M. Vernucci et al., The H19 locus acts in vivo as a tumor suppressor, Proceedings of the National Academy of Sciences, vol.105, issue.34, pp.12417-12422, 2008.
DOI : 10.1073/pnas.0801540105

I. Matouk, E. Raveh, P. Ohana, R. A. Lail, E. Gershtain et al., The Increasing Complexity of the Oncofetal H19 Gene Locus: Functional Dissection and Therapeutic Intervention, International Journal of Molecular Sciences, vol.14, issue.2, pp.4298-4316, 2013.
DOI : 10.3390/ijms14024298

M. Luo, Z. Li, W. Wang, Y. Zeng, Z. Liu et al., Long non-coding RNA H19 increases bladder cancer metastasis by associating with EZH2 and inhibiting E-cadherin expression, Cancer Letters, vol.333, issue.2, pp.213-221, 2013.
DOI : 10.1016/j.canlet.2013.01.033

L. Yan, J. Zhou, Y. Gao, S. Ghazal, L. Lu et al., Regulation of tumor cell migration and invasion by the H19/let-7 axis is antagonized by metformin-induced DNA methylation, Oncogene, vol.11, issue.23, 2014.
DOI : 10.1038/onc.2014.236

C. Ma, K. Nong, H. Zhu, W. Wang, X. Huang et al., H19 promotes pancreatic cancer metastasis by derepressing let-7???s suppression on its target HMGA2-mediated EMT, Tumor Biology, vol.174, issue.9, pp.9163-9169, 2014.
DOI : 10.1007/s13277-014-2185-5

P. Monnier, C. Martinet, J. Pontis, I. Stancheva, S. Ait-si-ali et al., H19 lncRNA controls gene expression of the Imprinted Gene Network by recruiting MBD1, Proceedings of the National Academy of Sciences, vol.110, issue.51, pp.20693-20698, 2014.
DOI : 10.1073/pnas.1310201110

X. Cai and B. R. Cullen, The imprinted H19 noncoding RNA is a primary microRNA precursor, RNA, vol.13, issue.3, pp.313-316, 2007.
DOI : 10.1261/rna.351707

A. Keniry, D. Oxley, P. Monnier, M. Kyba, L. Dandolo et al., The H19 lincRNA is a developmental reservoir of miR-675 that suppresses growth and Igf1r, Nature Cell Biology, vol.419, issue.7, pp.659-665, 2012.
DOI : 10.1093/emboj/18.14.4060

A. Venkatraman, X. C. He, J. L. Thorvaldsen, R. Sugimura, J. M. Perry et al., Maternal imprinting at the H19???Igf2 locus maintains adult haematopoietic stem cell quiescence, 2013) Maternal imprinting at the H19-Igf2 locus maintains adult haematopoietic stem cell quiescence, pp.345-349
DOI : 10.1038/nbt.1621

B. K. Dey, K. Pfeifer, and A. Dutta, The H19 long noncoding RNA gives rise to microRNAs miR-675-3p and miR-675-5p to promote skeletal muscle differentiation and regeneration, Genes & Development, vol.28, issue.5, pp.491-501, 2014.
DOI : 10.1101/gad.234419.113

A. N. Kallen, X. B. Zhou, J. Xu, C. Qiao, J. Ma et al., The Imprinted H19 LncRNA Antagonizes Let-7 MicroRNAs, Molecular Cell, vol.52, issue.1, pp.101-112, 2013.
DOI : 10.1016/j.molcel.2013.08.027

J. Wang, X. Liu, H. Wu, P. Ni, Z. Gu et al., CREB up-regulates long non-coding RNA, HULC expression through interaction with microRNA-372 in liver cancer, Nucleic Acids Research, vol.38, issue.16, pp.5366-5383, 2010.
DOI : 10.1093/nar/gkq285

J. M. Franco-zorrilla, A. Valli, M. Todesco, I. Mateos, M. I. Puga et al., Target mimicry provides a new mechanism for regulation of microRNA activity, Nature Genetics, vol.17, issue.8, pp.1033-1037, 2007.
DOI : 10.1038/ng2079

D. Cazalla, T. Yario, and J. A. Steitz, Down-Regulation of a Host MicroRNA by a Herpesvirus saimiri Noncoding RNA, Science, vol.328, issue.5985, pp.1563-1566, 2010.
DOI : 10.1126/science.1187197

L. Poliseno, L. Salmena, J. Zhang, B. Carver, W. J. Haveman et al., A coding-independent function of gene and pseudogene mRNAs regulates tumour biology, Nature, vol.33, issue.7301, pp.1033-1038, 2010.
DOI : 10.1038/nature09144

Y. Tay, L. Kats, L. Salmena, D. Weiss, S. M. Tan et al., Coding-Independent Regulation of the Tumor Suppressor PTEN by Competing Endogenous mRNAs, Cell, vol.147, issue.2, pp.344-357, 2011.
DOI : 10.1016/j.cell.2011.09.029

M. Cesana, D. Cacchiarelli, I. Legnini, T. Santini, O. Sthandier et al., A Long Noncoding RNA Controls Muscle Differentiation by Functioning as a Competing Endogenous RNA, Cell, vol.147, issue.2, pp.358-369, 2011.
DOI : 10.1016/j.cell.2011.09.028

S. Memczak, M. Jens, A. Elefsinioti, F. Torti, J. Krueger et al., Circular RNAs are a large class of animal RNAs with regulatory potency, Nature, vol.40, issue.7441, pp.333-338, 2013.
DOI : 10.1038/nature11928

T. B. Hansen, T. I. Jensen, B. H. Clausen, J. B. Bramsen, B. Finsen et al., Natural RNA circles function as efficient microRNA sponges, Nature, vol.175, issue.7441, pp.384-388, 2013.
DOI : 10.1038/nature11993

H. Zhu, N. Shyh-chang, A. V. Segre, G. Shinoda, S. P. Shah et al., The Lin28/let-7 Axis Regulates Glucose Metabolism, Cell, vol.147, issue.1, pp.81-94, 2011.
DOI : 10.1016/j.cell.2011.08.033

R. J. Frost and E. N. Olson, Control of glucose homeostasis and insulin sensitivity by the Let-7 family of microRNAs, Proceedings of the National Academy of Sciences, vol.108, issue.52, pp.21075-21080, 2011.
DOI : 10.1073/pnas.1118922109

N. Shyh-chang and G. Q. Daley, Lin28: Primal Regulator of Growth and Metabolism in Stem Cells, Cell Stem Cell, vol.12, issue.4, pp.395-406, 2013.
DOI : 10.1016/j.stem.2013.03.005

J. E. Thornton and R. I. Gregory, How does Lin28 let-7 control development and disease?, Trends in Cell Biology, vol.22, issue.9, pp.474-482, 2012.
DOI : 10.1016/j.tcb.2012.06.001

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3432650

Y. Huang, A mirror of two faces: Lin28 as a master regulator of both miRNA and mRNA, Wiley Interdisciplinary Reviews: RNA, vol.136, issue.4, pp.483-494, 2012.
DOI : 10.1002/wrna.1112

P. Briata, W. J. Lin, M. Giovarelli, M. Pasero, C. F. Chou et al., PI3K/AKT signaling determines a dynamic switch between distinct KSRP functions favoring skeletal myogenesis, Cell Death and Differentiation, vol.11, issue.3, pp.478-487, 2012.
DOI : 10.1038/nsmb.1874

URL : https://hal.archives-ouvertes.fr/hal-00675790

M. Trabucchi, P. Briata, M. Garcia-mayoral, A. D. Haase, W. Filipowicz et al., The RNA-binding protein KSRP promotes the biogenesis of a subset of microRNAs, Nature, vol.5, issue.5, pp.1010-1014, 2009.
DOI : 10.1038/nature08025

P. Briata, S. V. Forcales, M. Ponassi, G. Corte, C. Chen et al., p38-Dependent Phosphorylation of the mRNA Decay-Promoting Factor KSRP Controls the Stability of Select Myogenic Transcripts, Molecular Cell, vol.20, issue.6, pp.891-903, 2005.
DOI : 10.1016/j.molcel.2005.10.021

P. Onyango and A. P. Feinberg, A nucleolar protein, H19 opposite tumor suppressor (HOTS), is a tumor growth inhibitor encoded by a human imprinted H19 antisense transcript, Proceedings of the National Academy of Sciences, vol.108, issue.40, pp.16759-16764, 2011.
DOI : 10.1073/pnas.1110904108

J. C. Bruning, M. D. Michael, J. N. Winnay, T. Hayashi, D. Horsch et al., A Muscle-Specific Insulin Receptor Knockout Exhibits Features of the Metabolic Syndrome of NIDDM without Altering Glucose Tolerance, Molecular Cell, vol.2, issue.5, pp.559-569, 1998.
DOI : 10.1016/S1097-2765(00)80155-0

J. R. Mead, S. A. Irvine, and D. P. Ramji, Lipoprotein lipase: structure, function, regulation, and role in disease, Journal of Molecular Medicine, vol.80, issue.12, pp.753-769, 2002.
DOI : 10.1007/s00109-002-0384-9

K. Morino, K. F. Petersen, S. Sono, C. S. Choi, V. T. Samuel et al., Regulation of Mitochondrial Biogenesis by Lipoprotein Lipase in Muscle of Insulin-Resistant Offspring of Parents With Type 2 Diabetes, Diabetes, vol.61, issue.4, pp.61-877, 2012.
DOI : 10.2337/db11-1391

K. Morino, K. F. Petersen, S. Dufour, D. Befroy, J. Frattini et al., Reduced mitochondrial density and increased IRS-1 serine phosphorylation in muscle of insulin-resistant offspring of type 2 diabetic parents, Journal of Clinical Investigation, vol.115, issue.12, pp.3587-3593, 2005.
DOI : 10.1172/JCI25151

D. E. Befroy, K. F. Petersen, S. Dufour, G. F. Mason, R. A. De-graaf et al., Impaired Mitochondrial Substrate Oxidation in Muscle of Insulin-Resistant Offspring of Type 2 Diabetic Patients, Diabetes, vol.56, issue.5, pp.1376-1381, 2007.
DOI : 10.2337/db06-0783

K. F. Petersen, S. Dufour, D. Befroy, R. Garcia, and G. I. Shulman, Impaired Mitochondrial Activity in the Insulin-Resistant Offspring of Patients with Type 2 Diabetes, New England Journal of Medicine, vol.350, issue.7, pp.664-671, 2004.
DOI : 10.1056/NEJMoa031314

K. F. Petersen, S. Dufour, K. Morino, P. S. Yoo, G. W. Cline et al., Reversal of muscle insulin resistance by weight reduction in young, lean, insulin-resistant offspring of parents with type 2 diabetes, Proceedings of the National Academy of Sciences, vol.109, issue.21, pp.8236-8240, 2012.
DOI : 10.1073/pnas.1205675109

J. H. Warram, B. C. Martin, A. S. Krolewski, J. S. Soeldner, and C. R. Kahn, Slow Glucose Removal Rate and Hyperinsulinemia Precede the Development of Type II Diabetes in the Offspring of Diabetic Parents, Annals of Internal Medicine, vol.113, issue.12, pp.909-915, 1990.
DOI : 10.7326/0003-4819-113-12-909

J. E. Ayala, V. T. Samuel, G. J. Morton, S. Obici, C. M. Croniger et al., Standard operating procedures for describing and performing metabolic tests of glucose homeostasis in mice, Disease Models & Mechanisms, vol.3, issue.9-10, pp.525-534, 2010.
DOI : 10.1242/dmm.006239

L. Milligan, E. Antoine, C. Bisbal, M. Weber, C. Brunel et al., H19 gene expression is up-regulated exclusively by stabilization of the RNA during muscle cell differentiation, Oncogene, vol.19, issue.50, pp.5810-5816, 2000.
DOI : 10.1038/sj.onc.1203965

S. Fransson, A. Uv, H. Eriksson, M. K. Andersson, Y. Wettergren et al., ) p37delta is a new isoform of PI3K p110delta that increases cell proliferation and is overexpressed in tumors, Oncogene, pp.31-3277, 2012.

K. Kim, K. H. Kim, H. Y. Kim, H. K. Cho, N. Sakamoto et al., Curcumin inhibits hepatitis C virus replication via suppressing the Akt-SREBP-1 pathway, FEBS Letters, vol.44, issue.4, pp.707-712, 2010.
DOI : 10.1016/j.febslet.2009.12.019

A. C. Boaglio, A. E. Zucchetti, E. J. Sanchez-pozzi, J. M. Pellegrino, J. E. Ochoa et al., Phosphoinositide 3-kinase/protein kinase B signaling pathway is involved in estradiol 17??-d-glucuronide-induced cholestasis: Complementarity with classical protein kinase c, Hepatology, vol.38, issue.Suppl 1, pp.1465-1476, 2010.
DOI : 10.1002/hep.23846

G. Nicastro, M. F. Garcia-mayoral, D. Hollingworth, G. Kelly, S. R. Martin et al., Noncanonical G recognition mediates KSRP regulation of let-7 biogenesis, Nature Structural & Molecular Biology, vol.19, issue.12, pp.1282-1286, 2012.
DOI : 10.1038/nsmb.2427

G. Michlewski and J. F. Caceres, Antagonistic role of hnRNP A1 and KSRP in the regulation of let-7a biogenesis, Nature Structural & Molecular Biology, vol.623, issue.8, pp.1011-1018, 2010.
DOI : 10.1083/jcb.138.2.225

A. Gabory, M. A. Ripoche, L. Digarcher, A. Watrin, F. Ziyyat et al., H19 acts as a trans regulator of the imprinted gene network controlling growth in mice, Development, vol.136, issue.20, pp.3413-3421, 2009.
DOI : 10.1242/dev.036061

M. Borensztein, P. Monnier, F. Court, Y. Louault, M. A. Ripoche et al., Myod and H19-Igf2 locus interactions are required for diaphragm formation in the mouse, Development, vol.140, issue.6, pp.1231-1239, 2013.
DOI : 10.1242/dev.084665