, En effet, mieux comprendre la régulation et la fonction de gènes nouveaux, en particulier des gènes à ARN long non-codants, nécessitent de combiner différentes approches de génomique afin d'aboutir à des hypothèses solides qui seront ensuite à valider expérimentalement

S. Brenner, F. Jacob, and M. Meselson, An unstable intermediate carrying information from genes to ribosomes for protein synthesis, Nature, vol.190, pp.576-81, 1961.

S. Ohno, So much "junk" DNA in our genome, Brookhaven Symp Biol, vol.23, pp.366-70, 1972.

S. Brenner, Refuge of spandrels, Curr Biol, vol.8, p.669, 1998.

P. Drouin, A. Francoeur, J. Humbley, and A. Picton, Multiple Perspectives on Terminological Variation, 2017.

J. Brosius and S. J. Gould, On "genomenclature": a comprehensive (and respectful) taxonomy for pseudogenes and other "junk DNA, Proc Natl Acad Sci USA, vol.89, pp.10706-10716, 1992.

P. Kapranov, S. E. Cawley, J. Drenkow, S. Bekiranov, R. L. Strausberg et al., Largescale transcriptional activity in chromosomes 21 and 22, Science, vol.296, pp.916-925, 2002.

P. Bertone, V. Stolc, T. E. Royce, J. S. Rozowsky, A. E. Urban et al., Global identification of human transcribed sequences with genome tiling arrays, Science, vol.306, pp.2242-2248, 2004.

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, pp.1149-54, 2005.

, ENCODE Project Consortium. The ENCODE (ENCyclopedia Of DNA Elements) Project. Science, vol.306, pp.636-676, 2004.

P. Carninci, T. Kasukawa, S. Katayama, J. Gough, M. C. Frith et al., The transcriptional landscape of the mammalian genome, Science, vol.309, pp.1559-63, 2005.

A. T. Willingham and T. R. Gingeras, TUF love for "junk, DNA. Cell, vol.125, pp.1215-1235, 2006.

J. Ponjavic, C. P. Ponting, and G. Lunter, Functionality or transcriptional noise? Evidence for selection within long noncoding RNAs, Genome Res, vol.17, pp.556-65, 2007.

P. Kapranov, J. Cheng, S. Dike, D. A. Nix, R. Duttagupta et al., RNA Maps Reveal New RNA Classes and a Possible Function for Pervasive Transcription, Science, vol.316, pp.1484-1492, 2007.

J. Ponjavic and C. P. Ponting, The long and the short of RNA maps, Bioessays, vol.29, pp.1077-80, 2007.

. Encode-project-consortium, E. Birney, J. A. Stamatoyannopoulos, A. Dutta, R. Guigó et al., Identification and analysis of functional elements in 1% of the human genome by the ENCODE pilot project, Nature, vol.447, pp.799-816, 2007.

M. Kchouk, J. Gibrat, and M. Elloumi, Generations of Sequencing Technologies: From First to Next Generation, Biology and Medicine, p.9, 2017.

U. A. Ørom, T. Derrien, R. Guigo, and R. Shiekhattar, Long noncoding RNAs as enhancers of gene expression, Cold Spring Harb Symp Quant Biol, vol.75, pp.325-356, 2010.

U. A. Ørom, T. Derrien, M. Beringer, K. Gumireddy, A. Gardini et al., Long Noncoding RNAs with Enhancer-like Function in Human Cells, Cell, vol.143, pp.46-58, 2010.

T. Derrien, R. Johnson, G. Bussotti, A. Tanzer, S. Djebali et al., The GENCODE v7 catalog of human long noncoding RNAs: Analysis of their gene structure, evolution, and expression, Genome Research, vol.22, pp.1775-89, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01205054

J. Harrow, A. Frankish, J. M. Gonzalez, E. Tapanari, M. Diekhans et al., The reference human genome annotation for The ENCODE Project, Genome Research, vol.22, pp.1760-74, 2012.

J. M. Mudge and J. Harrow, Creating reference gene annotation for the mouse C57BL6/J genome assembly, Mamm Genome, vol.26, pp.366-78, 2015.

Y. Miao, S. Xu, L. Chen, G. Liang, Y. Pu et al., Trends of long noncoding RNA research from 2007 to 2016: a bibliometric analysis, Oncotarget, vol.8, pp.83114-83141, 2017.

K. Muret, C. Klopp, V. Wucher, D. Esquerré, F. Legeai et al., Long noncoding RNA repertoire in chicken liver and adipose tissue, Genetics Selection Evolution, vol.49, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01431420

S. Foissac, S. Djebali, K. Munyard, N. Villa-vialaneix, A. Rau et al., Livestock genome annotation: transcriptome and chromatin structure profiling in cattle, goat, chicken and pig, bioRxiv, p.316091, 2018.

S. Fang, L. Zhang, J. Guo, Y. Niu, Y. Wu et al., NONCODEV5: a comprehensive annotation database for long non-coding RNAs, Nucleic Acids Res, vol.46, pp.308-322, 2018.

A. Li, J. Zhang, Z. Zhou, L. Wang, Y. Liu et al., ALDB: A Domestic-Animal Long Noncoding RNA Database, PLOS ONE, vol.10, p.124003, 2015.

A. Listrat, B. Lebret, I. Louveau, T. Astruc, M. Bonnet et al., How Muscle Structure and Composition Influence Meat and Flesh Quality, ScientificWorldJournal, p.3182746, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01285897

J. D. Wood, M. Enser, A. V. Fisher, G. R. Nute, P. R. Sheard et al., Fat deposition, fatty acid composition and meat quality: A review, Meat Sci, vol.78, pp.343-58, 2008.

J. F. Hocquette, F. Gondret, E. Baéza, F. Médale, C. Jurie et al., Intramuscular fat content in meat-producing animals: development, genetic and nutritional control, and identification of putative markers, Animal, vol.4, pp.303-322, 2010.

C. Desert, E. Baéza, M. Aite, M. Boutin, L. Cam et al., Multi-tissue transcriptomic study reveals the main role of liver in the chicken adaptive response to a switch in dietary energy source through the transcriptional regulation of lipogenesis, BMC Genomics, vol.19, p.187, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01727161

K. Sato and T. Kamada, Regulation of bile acid, cholesterol, and fatty acid synthesis in chicken primary hepatocytes by different concentrations of T0901317, an agonist of liver X receptors

, Comp Biochem Physiol, Part A Mol Integr Physiol, vol.158, pp.201-207, 2011.

F. B. Hillgartner, L. M. Salati, and A. G. Goodridge, Physiological and molecular mechanisms involved in nutritional regulation of fatty acid synthesis, Physiol Rev, vol.75, pp.47-76, 1995.

D. H. Greene and D. P. Selivonchick, Lipid metabolism in fish, Prog Lipid Res, vol.26, pp.53-85, 1987.

W. G. Bergen and H. J. Mersmann, Comparative aspects of lipid metabolism: impact on contemporary research and use of animal models, J Nutr, vol.135, pp.2499-502, 2005.

J. Ye and R. A. Debose-boyd, Regulation of cholesterol and fatty acid synthesis, Cold Spring Harb Perspect Biol, vol.3, 2011.

E. Viturro, M. Koenning, A. Kroemer, G. Schlamberger, S. Wiedemann et al., Cholesterol synthesis in the lactating cow: Induced expression of candidate genes, J Steroid Biochem Mol Biol, vol.115, pp.62-69, 2009.

Y. Wang, Journal d'agriculture traditionnelle et de botanique appliquée, vol.28, pp.253-261, 1981.

A. Chevalier, Journal d'agriculture traditionnelle et de botanique appliquée, vol.19, pp.613-62, 1939.

W. Jiao and K. Schneeberger, The impact of third generation genomic technologies on plant genome assembly, Curr Opin Plant Biol, vol.36, pp.64-70, 2017.

S. Kumar and S. B. Hedges, A molecular timescale for vertebrate evolution, Nature, vol.392, pp.917-937, 1998.

S. B. Hedges, S. Kumar, and . Genomics, Vertebrate genomes compared, Science, vol.297, pp.1283-1288, 2002.

H. Hezroni, D. Koppstein, M. G. Schwartz, A. Avrutin, D. P. Bartel et al., Principles of Long Noncoding RNA Evolution Derived from Direct Comparison of Transcriptomes in 17 Species, Cell Reports, vol.11, pp.1110-1132, 2015.

Q. Yin, Y. L. Zhang, Y. Xiang, J. Wu, Y. Carmichael et al., Long noncoding RNAs with snoRNA ends, Mol Cell, vol.48, pp.219-249, 2012.

L. Ma, V. B. Bajic, and Z. Zhang, On the classification of long non-coding RNAs, RNA Biol, vol.10, pp.925-958, 2013.

H. Bunch, Gene regulation of mammalian long non-coding RNA, Mol Genet Genomics, vol.293, pp.1-15, 2018.

C. Tordonato, D. Fiore, P. P. Nicassio, and F. , The role of non-coding RNAs in the regulation of stem cells and progenitors in the normal mammary gland and in breast tumors, Front Genet, vol.6, p.72, 2015.

B. S. Gloss and M. E. Dinger, The specificity of long noncoding RNA expression, Biochim Biophys Acta, vol.1859, pp.16-22, 2016.

I. Ulitsky, A. Shkumatava, C. H. Jan, H. Sive, and D. P. Bartel, Conserved Function of lincRNAs in Vertebrate Embryonic Development despite Rapid Sequence Evolution, Cell, vol.147, pp.1537-50, 2011.

A. Necsulea, M. Soumillon, M. Warnefors, A. Liechti, T. Daish et al., The evolution of lncRNA repertoires and expression patterns in tetrapods, Nature, vol.505, pp.635-675, 2014.

W. Haerty and C. P. Ponting, Unexpected selection to retain high GC content and splicing enhancers within exons of multiexonic lncRNA loci, RNA, vol.21, pp.320-352, 2015.

A. Nitsche, D. Rose, M. Fasold, K. Reiche, and P. F. Stadler, Comparison of splice sites reveals that long noncoding RNAs are evolutionarily well conserved, RNA, vol.21, pp.801-813, 2015.

D. Chernikova, D. Managadze, G. V. Glazko, W. Makalowski, and I. B. Rogozin, Conservation of the Exon-Intron Structure of Long Intergenic Non-Coding RNA Genes in Eutherian Mammals, Life

A. Zampetaki, A. Albrecht, and K. Steinhofel, Long Non-coding RNA Structure and Function: Is There a Link?, Front Physiol, vol.9, p.1201, 2018.

L. Fabris and G. A. Calin, Understanding the Genomic Ultraconservations: T-UCRs and Cancer, Int Rev Cell Mol Biol, vol.333, pp.159-72, 2017.

S. Xu, L. Zhou, M. Ponnusamy, L. Zhang, Y. Dong et al., A comprehensive review of circRNA: from purification and identification to disease marker potential, PeerJ, vol.6, p.5503, 2018.

R. Ashwal-fluss, M. Meyer, N. R. Pamudurti, A. Ivanov, O. Bartok et al., circRNA biogenesis competes with pre-mRNA splicing, Mol Cell, vol.56, pp.55-66, 2014.

N. R. Pamudurti, O. Bartok, M. Jens, R. Ashwal-fluss, C. Stottmeister et al., Translation of CircRNAs. Mol Cell, vol.66, pp.9-21, 2017.

A. Bagchi, Different roles of circular RNAs with protein coding potentials, Biochem Biophys Res Commun, vol.500, pp.907-916, 2018.

Y. Zhang, X. Zhang, T. Chen, J. Xiang, Q. Yin et al., Circular Intronic Long Noncoding RNAs, Molecular Cell, vol.51, pp.792-806, 2013.

J. L. Vandeberg, P. G. Johnston, D. W. Cooper, and E. S. Robinson, X-chromosome inactivation and evolution in marsupials and other mammals, Isozymes Curr Top Biol Med Res, vol.9, pp.201-219, 1983.

. Duret, The Xist RNA Gene Evolved in Eutherians by Pseudogenization of a ProteinCoding Gene, Science, vol.312, pp.1653-1658, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00427928

H. Hezroni, B. Perry, R. , M. Z. Housman, G. Lubelsky et al., A subset of conserved mammalian long non-coding RNAs are fossils of ancestral protein-coding genes

, Genome Biol, vol.18, p.162, 2017.

E. A. Elisaphenko, N. N. Kolesnikov, A. I. Shevchenko, I. B. Rogozin, T. B. Nesterova et al., A dual origin of the Xist gene from a protein-coding gene and a set of transposable elements, PLoS ONE, vol.3, p.2521, 2008.

L. Béguec, C. Wucher, V. Lagoutte, L. Cadieu, E. Botherel et al., Characterisation and functional predictions of canine long non-coding, RNAs. Sci Rep, vol.8, p.13444, 2018.

J. Schmitz and J. Brosius, Exonization of transposed elements: A challenge and opportunity for evolution, Biochimie, vol.93, pp.1928-1962, 2011.

D. Casane and P. Laurenti, Syllogomanie moléculaire : l'ADN non codant enrichit le jeu des possibles. médecine/sciences, vol.30, pp.1177-83, 2014.

R. Dong, X. Zhang, Y. Zhang, X. Ma, L. Chen et al., CircRNA-derived pseudogenes, Cell Res, vol.26, pp.747-50, 2016.

L. Kong, Y. Zhang, Z. Ye, X. Liu, S. Zhao et al., CPC: assess the proteincoding potential of transcripts using sequence features and support vector machine, Nucleic Acids Research, vol.35, pp.345-354, 2007.

Y. Kang, Y. Kong, L. Hou, M. Meng, Y. Wei et al., CPC2: a fast and accurate coding potential calculator based on sequence intrinsic features, Nucleic Acids Res, vol.45, pp.12-18, 2017.

M. F. Lin, I. Jungreis, and M. Kellis, PhyloCSF: a comparative genomics method to distinguish protein coding and non-coding regions, Bioinformatics, vol.27, pp.275-282, 2011.

L. Wang, H. J. Park, S. Dasari, S. Wang, J. Kocher et al., CPAT: Coding-Potential Assessment Tool using an alignment-free logistic regression model, Nucleic Acids Research, vol.41, pp.74-74, 2013.

L. Sun, H. Luo, D. Bu, G. Zhao, K. Yu et al., Utilizing sequence intrinsic composition to classify protein-coding and long non-coding transcripts, Nucleic Acids Research, vol.41, pp.166-166, 2013.

A. Li, J. Zhang, and Z. Zhou, PLEK: a tool for predicting long non-coding RNAs and messenger RNAs based on an improved k-mer scheme, BMC bioinformatics, vol.15, p.311, 2014.

V. Wucher, F. Legeai, B. Hedan, G. Rizk, L. Lagoutte et al., A tool for Long non-coding RNAs annotation and its application to the dog transcriptome, 2016.

L. Hu, Z. Xu, B. Hu, and Z. J. Lu, COME: a robust coding potential calculation tool for lncRNA identification and characterization based on multiple features, Nucleic Acids Res, vol.45, p.2, 2017.

C. Van-solingen, K. R. Scacalossi, and K. J. Moore, Long noncoding RNAs in lipid metabolism, Curr Opin Lipidol, vol.29, pp.224-256, 2018.

N. S. Benabdallah and W. A. Bickmore, Regulatory Domains and Their Mechanisms, Cold Spring Harb Symp Quant Biol, vol.80, pp.45-51, 2015.

K. Katsarou, A. Rao, M. Tsagris, and K. Kalantidis, Infectious long non-coding RNAs, Biochimie, vol.117, pp.37-47, 2015.

Y. Wang, T. Yang, Z. Zhang, M. Lu, W. Zhao et al., Long non-coding RNA TUG1 promotes migration and invasion by acting as a ceRNA of miR-335-5p in osteosarcoma cells, Cancer Sci, vol.108, pp.859-67, 2017.

E. Song, The Long and Short Non-coding RNAs in Cancer Biology, 2016.

R. Johnson, C. Teh, H. Jia, R. R. Vanisri, T. Pandey et al., Regulation of neural macroRNAs by the transcriptional repressor REST, RNA, vol.15, pp.85-96, 2009.

S. Lazorthes, C. Vallot, S. Briois, M. Aguirrebengoa, J. Thuret et al., A vlincRNA participates in senescence maintenance by relieving H2AZ-mediated repression at the INK4 locus, Nat Commun, vol.6, p.5971, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01465935

S. Povey, R. Lovering, E. Bruford, M. Wright, M. Lush et al., The HUGO Gene Nomenclature Committee (HGNC), Hum Genet, vol.109, pp.678-80, 2001.

A. V. Bartel, B. Bartel, D. P. Burge, C. B. Carrington, J. C. Chen et al., A uniform system for microRNA annotation, RNA, vol.9, pp.277-286, 2003.

M. W. Wright and E. A. Bruford, Naming "junk": human non-protein coding RNA (ncRNA) gene nomenclature, Hum Genomics, vol.5, pp.90-98, 2011.

M. W. Wright, A short guide to long non-coding RNA gene nomenclature, Hum Genomics, vol.8, p.7, 2014.

J. S. Mattick and J. L. Rinn, Discovery and annotation of long noncoding RNAs, Nat Struct Mol Biol, vol.22, pp.5-7, 2015.

P. Volders, K. Verheggen, G. Menschaert, K. Vandepoele, L. Martens et al., An update on LNCipedia: a database for annotated human lncRNA sequences, Nucleic Acids Res, pp.174-80, 2015.

P. Volders, K. Helsens, X. Wang, B. Menten, L. Martens et al., LNCipedia: a database for annotated human lncRNA transcript sequences and structures, Nucleic Acids Res, pp.246-251, 2013.

Z. Lu, Q. C. Zhang, B. Lee, R. A. Flynn, M. A. Smith et al., RNA Duplex Map in Living Cells Reveals Higher-Order Transcriptome Structure. Cell, vol.165, pp.1267-79, 2016.

C. K. Kwok, Dawn of the in vivo RNA structurome and interactome, Biochem Soc Trans, vol.44, pp.1395-410, 2016.

G. Rothschild and U. Basu, Lingering Questions about Enhancer RNA and Enhancer Transcription-Coupled Genomic Instability, Trends Genet, vol.33, pp.143-54, 2017.

L. Costantino and D. Koshland, The Yin and Yang of R-loop biology, Curr Opin Cell Biol, vol.34, pp.39-45, 2015.

R. Boque-sastre, M. Soler, C. Oliveira-mateos, A. Portela, C. Moutinho et al., Headto-head antisense transcription and R-loop formation promotes transcriptional activation, Proc Natl Acad Sci U S A, vol.112, pp.5785-90, 2015.

S. W. Umlauf, M. M. Cox, and R. B. Inman, Triple-helical DNA Pairing Intermediates Formed by recA Protein, J Biol Chem, pp.16898-912, 1990.

T. De-lange, How Telomeres Solve the End-Protection Problem, Science, vol.326, p.948, 2009.

J. A. Brown, M. L. Valenstein, T. A. Yario, K. T. Tycowski, and J. A. Steitz, Formation of triple-helical structures by the 3'-end sequences of MALAT1 and MEN? noncoding RNAs, Proc Natl Acad Sci USA, vol.109, pp.19202-19209, 2012.

A. De-cian, J. Gros, A. Guedin, M. Haddi, S. Lyonnais et al., Nucleic Acids Symposium Series, vol.52, pp.7-8, 2008.

S. Burge, G. N. Parkinson, P. Hazel, A. K. Todd, and S. Neidle, Quadruplex DNA: sequence, topology and structure, Nucleic Acids Res, vol.34, pp.5402-5417, 2006.

K. Matsumura, Y. Kawasaki, M. Miyamoto, Y. Kamoshida, J. Nakamura et al., The novel G-quadruplex-containing long non-coding RNA GSEC antagonizes DHX36 and modulates colon cancer cell migration, Oncogene, vol.36, pp.1191-1200, 2017.

G. G. Jayaraj, S. Pandey, V. Scaria, and S. Maiti, Potential G-quadruplexes in the human long noncoding transcriptome, RNA Biol, vol.9, pp.81-87, 2012.

J. Kondo, W. Adachi, S. Umeda, T. Sunami, and A. Takénaka, Crystal structures of a DNA octaplex with I-motif of G-quartets and its splitting into two quadruplexes suggest a folding mechanism of eight tandem repeats, Nucleic Acids Res, vol.32, pp.2541-2550, 2004.

L. Su, L. Chen, M. Egli, J. M. Berger, and A. Rich, Minor groove RNA triplex in the crystal structure of a ribosomal frameshifting viral pseudoknot, Nat Struct Biol, vol.6, pp.285-92, 1999.

M. Tarköy, A. K. Phipps, P. Schultze, and J. Feigon, Solution structure of an intramolecular DNA triplex linked by hexakis(ethylene glycol) units: d(AGAGAGAA-(EG)6-TTCTCTCT-(EG)6-TCTCTCTT), Biochemistry, vol.37, pp.5810-5819, 1998.

W. J. Chung, B. Heddi, E. Schmitt, K. W. Lim, Y. Mechulam et al., Structure of a lefthanded DNA G-quadruplex, Proc Natl Acad Sci USA, vol.112, pp.2729-2762, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01140567

M. C. Chen, R. Tippana, N. A. Demeshkina, P. Murat, S. Balasubramanian et al., Structural basis of G-quadruplex unfolding by the DEAH/RHA helicase DHX36, Nature, vol.558, pp.465-474, 2018.

S. D. Auweter and F. C. Oberstrass, Allain FH-T. Sequence-specific binding of single-stranded RNA: is there a code for recognition?, Nucleic Acids Res, vol.34, pp.4943-59, 2006.

K. C. Wang and H. Y. Chang, Molecular Mechanisms of Long Noncoding RNAs, Molecular Cell, vol.43, pp.904-918, 2011.

J. Chen, R. Wang, K. Zhang, and L. Chen, Long non-coding RNAs in non-small cell lung cancer as biomarkers and therapeutic targets, J Cell Mol Med, vol.18, pp.2425-2461, 2014.

J. Sana, P. Faltejskova, M. Svoboda, and O. Slaby, Novel classes of non-coding RNAs and cancer, J Transl Med, vol.10, p.103, 2012.

A. Cipriano and M. Ballarino, The Ever-Evolving Concept of the Gene: The Use of RNA/Protein Experimental Techniques to Understand Genome Functions, Front Mol Biosci, vol.5, p.20, 2018.

A. H. Fox, Y. W. Lam, A. Leung, C. E. Lyon, J. Andersen et al., Paraspeckles: a novel nuclear domain, Curr Biol, vol.12, pp.13-25, 2002.

J. E. Wilusz, H. Sunwoo, and D. L. Spector, Long noncoding RNAs: functional surprises from the RNA world, Genes Dev, vol.23, pp.1494-504, 2009.

Y. Lin, B. F. Schmidt, M. P. Bruchez, and C. J. Mcmanus, Structural analyses of NEAT1 lncRNAs suggest long-range RNA interactions that may contribute to paraspeckle architecture, Nucleic Acids Res, vol.46, pp.3742-52, 2018.

C. M. Clemson, J. N. Hutchinson, S. A. Sara, A. W. Ensminger, A. H. Fox et al., An architectural role for a nuclear noncoding RNA: NEAT1 RNA is essential for the structure of paraspeckles, Mol Cell, vol.33, pp.717-743, 2009.

Q. Li, A. M. Spinelli, R. Wang, Y. Anfinogenova, H. A. Singer et al., Critical role of vimentin phosphorylation at Ser-56 by p21-activated kinase in vimentin cytoskeleton signaling, J Biol Chem, vol.281, pp.34716-34740, 2006.

P. Fernandes, D. Bitar, M. Jacobs, F. Barry, and G. , Long Non-Coding RNAs in Neuronal Aging, Noncoding RNA, vol.4, 2018.

D. Oliva-rico and L. A. Herrera, Regulated expression of the lncRNA TERRA and its impact on telomere biology, Mechanisms of Ageing and Development, vol.167, pp.16-23, 2017.

Y. Xu, Y. Suzuki, K. Ito, and M. Komiyama, Telomeric repeat-containing RNA structure in living cells, Proc Natl Acad Sci USA, vol.107, pp.14579-84, 2010.

M. Munschauer, C. T. Nguyen, K. Sirokman, C. R. Hartigan, L. Hogstrom et al., The NORAD lncRNA assembles a topoisomerase complex critical for genome stability, Nature, vol.561, pp.132-138, 2018.

K. Ng, D. Pullirsch, M. Leeb, and A. Wutz, Xist and the order of silencing, EMBO Rep, vol.8, pp.34-43, 2007.

F. P. Marchese, I. Raimondi, and M. Huarte, The multidimensional mechanisms of long noncoding RNA function, Genome Biol, vol.18, p.206, 2017.

S. Wang, H. Ke, H. Zhang, Y. Ma, L. Ao et al., LncRNA MIR100HG promotes cell proliferation in triple-negative breast cancer through triplex formation with p27 loci, Cell Death Dis, vol.9, p.805, 2018.

I. Martianov, A. Ramadass, S. Barros, A. Chow, N. Akoulitchev et al., Repression of the human dihydrofolate reductase gene by a non-coding interfering transcript, Nature, vol.445, pp.666-70, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00190957

P. Grote, L. Wittler, D. Hendrix, F. Koch, S. Währisch et al., The tissue-specific lncRNA Fendrr is an essential regulator of heart and body wall development in the mouse, Dev Cell, vol.24, pp.206-220, 2013.

P. Grote and B. G. Herrmann, The long non-coding RNA Fendrr links epigenetic control mechanisms to gene regulatory networks in mammalian embryogenesis, RNA Biol, vol.10, pp.1579-85, 2013.

T. Mondal, S. Subhash, R. Vaid, S. Enroth, S. Uday et al., MEG3 long noncoding RNA regulates the TGF-? pathway genes through formation of RNA-DNA triplex structures, Nat Commun, vol.6, p.7743, 2015.

A. Postepska-igielska, A. Giwojna, L. Gasri-plotnitsky, N. Schmitt, A. Dold et al., LncRNA Khps1 Regulates Expression of the Proto-oncogene SPHK1 via Triplex-Mediated Changes in Chromatin Structure, Mol Cell, vol.60, pp.626-662, 2015.

F. Aguilo, M. Zhou, and M. J. Walsh, Long noncoding RNA, polycomb, and the ghosts haunting INK4b-ARF-INK4a expression, Cancer Res, vol.71, pp.5365-5374, 2011.

N. Romero-barrios, M. F. Legascue, M. Benhamed, A. F. Crespi, and M. , Splicing regulation by long noncoding RNAs, Nucleic Acids Res, vol.46, pp.2169-84, 2018.

O. Villamizar, C. B. Chambers, J. M. Riberdy, D. A. Persons, and A. Wilber, Long noncoding RNA Saf and splicing factor 45 increase soluble Fas and resistance to apoptosis, Oncotarget, vol.7, pp.13810-13836, 2016.

M. Beltran, I. Puig, C. Peña, J. M. García, A. B. Alvarez et al., A natural antisense transcript regulates Zeb2/Sip1 gene expression during Snail1-induced epithelial-mesenchymal transition, Genes Dev, vol.22, pp.756-69, 2008.

V. Tripathi, J. D. Ellis, Z. Shen, D. Y. Song, Q. Pan et al., The Nuclear-Retained Noncoding RNA MALAT1 Regulates Alternative Splicing by Modulating SR Splicing Factor Phosphorylation, Molecular Cell, vol.39, pp.925-963, 2010.

M. Van-dijk, A. Visser, K. Buabeng, A. Poutsma, R. C. Van-der-schors et al., Mutations within the LINC-HELLP non-coding RNA differentially bind ribosomal and RNA splicing complexes and negatively affect trophoblast differentiation, Hum Mol Genet, vol.24, pp.5475-85, 2015.

J. Liz, A. Portela, M. Soler, A. Gómez, H. Ling et al., Regulation of primiRNA processing by a long noncoding RNA transcribed from an ultraconserved region, Mol Cell, vol.55, pp.138-185, 2014.

J. Guo, W. Fang, L. Sun, Y. Lu, L. Dou et al., Ultraconserved element uc.372 drives hepatic lipid accumulation by suppressing miR-195/miR4668 maturation, Nat Commun, vol.9, p.612, 2018.

J. Hu, Y. Qian, L. Peng, L. Ma, T. Qiu et al., Long Noncoding RNA EGFR-AS1 Promotes Cell Proliferation by Increasing EGFR mRNA Stability in Gastric Cancer, Cell Physiol Biochem, vol.49, pp.322-356, 2018.

M. A. Faghihi, F. Modarresi, A. M. Khalil, D. E. Wood, B. G. Sahagan et al., Expression of a noncoding RNA is elevated in Alzheimer's disease and drives rapid feedforward regulation of beta-secretase, Nat Med, vol.14, pp.723-753, 2008.

P. Johnsson, A. Ackley, L. Vidarsdottir, W. Lui, M. Corcoran et al., A pseudogene long-noncoding-RNA network regulates PTEN transcription and translation in human cells, Nat Struct Mol Biol, vol.20, pp.440-446, 2013.

C. Gong and L. E. Maquat, lncRNAs transactivate STAU1-mediated mRNA decay by duplexing with 3' UTRs via Alu elements, Nature, vol.470, pp.284-292, 2011.

J. Kim, J. H. Noh, S. Lee, R. Munk, A. Sharov et al., LncRNA OIP5-AS1/cyrano suppresses GAK expression to control mitosis, Oncotarget, vol.8, pp.49409-49429, 2017.

K. Katsushima, A. Natsume, F. Ohka, K. Shinjo, A. Hatanaka et al., Targeting the Notch-regulated non-coding RNA TUG1 for glioma treatment, Nat Commun, vol.7, 2016.

S. Li, X. Liu, H. Li, H. Pan, A. Acharya et al., Integrated analysis of long noncoding RNA-associated competing endogenous RNA network in periodontitis, Journal of Periodontal Research, vol.53, pp.495-505, 2018.

G. Li, K. Liu, and X. Du, Long Non-Coding RNA TUG1 Promotes Proliferation and Inhibits Apoptosis of Osteosarcoma Cells by Sponging miR-132-3p and Upregulating SOX4 Expression, Yonsei Med J, vol.59, pp.226-261, 2018.

J. Huang, X. , J. J. Pan, S. Zhuge, and X. , Inhibition of long non-coding RNA TUG1 on gastric cancer cell transference and invasion through regulating and controlling the expression of miR-144/c-Met axis, Asian Pac J Trop Med, vol.9, pp.508-520, 2016.

H. Cai, Y. Xue, P. Wang, Z. Wang, Z. Li et al., The long noncoding RNA TUG1 regulates blood-tumor barrier permeability by targeting miR-144, Oncotarget, vol.6, pp.19759-79, 2015.

J. Cao, X. Han, X. Qi, J. X. Li, and X. , TUG1 promotes osteosarcoma tumorigenesis by upregulating EZH2 expression via miR-144-3p, Int J Oncol, vol.51, pp.1115-1138, 2017.

J. Tan, K. Qiu, M. Li, and Y. Liang, Double-negative feedback loop between long non-coding RNA TUG1 and miR-145 promotes epithelial to mesenchymal transition and radioresistance in human bladder cancer cells, FEBS Lett, vol.589, issue.20, pp.3175-81, 2015.

H. Lei, Y. Gao, and X. Xu, LncRNA TUG1 influences papillary thyroid cancer cell proliferation, migration and EMT formation through targeting miR-145, Acta Biochim Biophys Sin (Shanghai), vol.49, pp.588-97, 2017.

K. Ren, Z. Li, Y. Li, W. Zhang, and X. Han, Long Noncoding RNA Taurine-Upregulated Gene, vol.1

, Promotes Cell Proliferation and Invasion in Gastric Cancer via Negatively Modulating miRNA-145-5p, Oncol Res, vol.25, pp.789-98, 2017.

Z. Wu, S. Zhao, C. Li, and C. Liu, LncRNA TUG1 serves an important role in hypoxia-induced myocardial cell injury by regulating the miR ; 145 ; 5p ; Binp3 axis, Mol Med Rep, vol.17, pp.2422-2452, 2018.

C. Li, Y. Gao, Y. Li, and D. Ding, TUG1 mediates methotrexate resistance in colorectal cancer via miR-186/CPEB2 axis, Biochem Biophys Res Commun, vol.491, pp.552-559, 2017.

C. Yu, L. Li, F. Xie, S. Guo, F. Liu et al., LncRNA TUG1 sponges miR-204-5p to promote osteoblast differentiation through upregulating Runx2 in aortic valve calcification, Cardiovasc Res, vol.114, pp.168-79, 2018.

J. Li, G. An, M. Zhang, and Q. Ma, Long non-coding RNA TUG1 acts as a miR-26a sponge in human glioma cells, Biochem Biophys Res Commun, vol.477, pp.743-751, 2016.

L. Liu, X. Chen, Y. Zhang, Y. Hu, X. Shen et al., Long non-coding RNA TUG1 promotes endometrial cancer development via inhibiting miR-299 and miR-34a-5p, Oncotarget, vol.8, pp.31386-94, 2017.

H. Cai, X. Liu, J. Zheng, Y. Xue, J. Ma et al., Long non-coding RNA taurine upregulated 1 enhances tumor-induced angiogenesis through inhibiting microRNA-299 in human glioblastoma, Oncogene, vol.36, pp.318-349, 2017.

F. Ma, S. Wang, Q. Cai, J. Zhou, D. Ding et al., Long non-coding RNA TUG1 promotes cell proliferation and metastasis by negatively regulating miR-300 in gallbladder carcinoma, Biomed Pharmacother, vol.88, pp.863-872, 2017.

R. Dong, G. Liu, B. Liu, G. Chen, K. Li et al., Targeting long non-coding RNA-TUG1 inhibits tumor growth and angiogenesis in hepatoblastoma, Cell Death Dis, vol.7, p.2278, 2016.

L. Duan, M. Ding, L. Hou, Y. Cui, C. Li et al., Long noncoding RNA TUG1 alleviates extracellular matrix accumulation via mediating microRNA-377 targeting of PPAR? in diabetic nephropathy, Biochem Biophys Res Commun, vol.484, pp.598-604, 2017.

M. Zhang, S. Huang, and D. Long, MiR-381 inhibits migration and invasion in human gastric carcinoma through downregulatedting SOX4, Oncol Lett, vol.14, pp.3760-3766, 2017.

L. Zhao, H. Sun, H. Kong, Z. Chen, B. Chen et al., The Lncrna-TUG1/EZH2 Axis Promotes Pancreatic Cancer Cell Proliferation, Migration and EMT Phenotype Formation Through Sponging Mir-382, Cell Physiol Biochem, vol.42, pp.2145-58, 2017.

G. Li, H. Song, L. Chen, W. Yang, K. Nan et al., TUG1 promotes lens epithelial cell apoptosis by regulating miR-421/caspase-3 axis in age-related cataract, Exp Cell Res, vol.356, pp.20-27, 2017.

Y. Lin, M. Wu, Y. Huang, C. Yeh, M. Cheng et al., Taurine upregulated gene 1 functions as a master regulator to coordinate glycolysis and metastasis in hepatocellular carcinoma, Hepatology, vol.67, pp.188-203, 2018.

X. Zhao and G. Ren, LncRNA Taurine-Upregulated Gene 1 Promotes Cell Proliferation by Inhibiting MicroRNA-9 in MCF-7 Cells, J Breast Cancer, vol.19, pp.349-57, 2016.

S. Chen, M. Wang, H. Yang, L. Mao, Q. He et al., LncRNA TUG1 sponges microRNA-9 to promote neurons apoptosis by up-regulated Bcl2l11 under ischemia, Biochem Biophys Res Commun, vol.485, pp.167-73, 2017.

C. Xie, Y. Cao, Y. Huang, Q. Shi, J. Guo et al., Long non-coding RNA TUG1 contributes to tumorigenesis of human osteosarcoma by sponging miR-9-5p and regulating POU2F1 expression, Tumour Biol, vol.37, pp.15031-15072, 2016.

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.495, pp.333-341, 2013.

K. Hu, L. Li, Y. Liao, and M. Liang, LncRNA Gm2044 highly expresses in spermatocyte and inhibits Utf1 translation by interacting with Utf1 mRNA, Genes Genomics, vol.40, pp.781-788, 2018.

J. Yoon, K. Abdelmohsen, S. Srikantan, X. Yang, J. L. Martindale et al., LincRNAp21 suppresses target mRNA translation, Mol Cell, vol.47, pp.648-55, 2012.

A. T. Willingham, A. P. Orth, S. Batalov, E. C. Peters, B. G. Wen et al., A strategy for probing the function of noncoding RNAs finds a repressor of NFAT, Science, vol.309, pp.1570-1573, 2005.

J. Chen, L. Liu, G. Wei, W. Wu, H. Luo et al., The long noncoding RNA ASNR regulates degradation of Bcl-2 mRNA through its interaction with AUF1, Sci Rep, vol.6, p.32189, 2016.

A. Lin, Q. Hu, C. Li, Z. Xing, G. Ma et al., The LINK-A lncRNA interacts with PtdIns(3,4,5)P3 to hyperactivate AKT and confer resistance to AKT inhibitors, Nat Cell Biol, vol.19, pp.238-51, 2017.

Z. Ji, R. Song, A. Regev, and K. Struhl, Many lncRNAs, 5'UTRs, and pseudogenes are translated and some are likely to express functional proteins, Elife, vol.4, p.8890, 2015.

D. M. Anderson, K. M. Anderson, C. Chang, C. A. Makarewich, B. R. Nelson et al., A micropeptide encoded by a putative long noncoding RNA regulates muscle performance, Cell, vol.160, pp.595-606, 2015.

B. R. Nelson, C. A. Makarewich, D. M. Anderson, B. R. Winders, C. D. Troupes et al., A peptide encoded by a transcript annotated as long noncoding RNA enhances SERCA activity in muscle, Science, vol.351, pp.271-276, 2016.

J. Ruiz-orera, X. Messeguer, J. A. Subirana, and M. M. Alba, Long non-coding RNAs as a source of new peptides, Elife, vol.3, p.3523, 2014.

J. Huang, M. Chen, D. Chen, X. Gao, S. Zhu et al., A Peptide Encoded by a Putative lncRNA HOXB-AS3 Suppresses Colon Cancer Growth, Mol Cell, vol.68, pp.171-184, 2017.

A. Matsumoto and K. I. Nakayama, Hidden Peptides Encoded by Putative Noncoding RNAs, Cell Struct Funct, vol.43, pp.75-83, 2018.

V. Boivin, G. Deschamps-francoeur, and M. S. Scott, Protein coding genes as hosts for noncoding RNA expression, Semin Cell Dev Biol, vol.75, pp.3-12, 2018.

M. K. Iyer, Y. S. Niknafs, R. Malik, U. Singhal, A. Sahu et al., The landscape of long noncoding RNAs in the human transcriptome, Nature Genetics, vol.47, pp.199-208, 2015.

A. Dobin, C. A. Davis, F. Schlesinger, J. Drenkow, C. Zaleski et al., STAR: ultrafast universal RNA-seq aligner, Bioinformatics, vol.29, pp.15-21, 2013.

C. Trapnell, B. A. Williams, G. Pertea, A. Mortazavi, G. Kwan et al., Transcript assembly and quantification by RNA-Seq reveals unannotated transcripts and isoform switching during cell differentiation, Nature Biotechnology, vol.28, pp.511-516, 2010.

Y. Liao, G. K. Smyth, and W. Shi, featureCounts: an efficient general purpose program for assigning sequence reads to genomic features, Bioinformatics, vol.30, pp.923-953, 2014.

A. Rau, M. Gallopin, G. Celeux, and F. Jaffrezic, Data-based filtering for replicated highthroughput transcriptome sequencing experiments, Bioinformatics, vol.29, pp.2146-52, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00927025

S. U. Schmitz, P. Grote, and B. G. Herrmann, Mechanisms of long noncoding RNA function in development and disease, Cellular and Molecular Life Sciences, vol.73, pp.2491-509, 2016.

H. Takahashi, S. Kato, M. Murata, and P. Carninci, CAGE (cap analysis of gene expression): a protocol for the detection of promoter and transcriptional networks, Methods Mol Biol, vol.786, pp.181-200, 2012.

J. E. Mank, Sex chromosome dosage compensation: definitely not for everyone, Trends Genet, vol.29, pp.677-83, 2013.

A. Frankish, B. Uszczynska, G. Ritchie, J. M. Gonzalez, D. Pervouchine et al., Comparison of GENCODE and RefSeq gene annotation and the impact of reference geneset on variant effect prediction, BMC Genomics, vol.16, 2015.

. Encode-project-consortium, A user's guide to the encyclopedia of DNA elements (ENCODE), PLoS Biol, vol.9, p.1001046, 2011.

R. Liu, A. E. Loraine, and J. A. Dickerson, Comparisons of computational methods for differential alternative splicing detection using RNA-seq in plant systems, BMC Bioinformatics, vol.15, p.364, 2014.

C. Trapnell, A. Roberts, L. Goff, G. Pertea, D. Kim et al., Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks, Nat Protoc, vol.7, pp.562-78, 2012.

S. Lin, Y. Lin, J. R. Nery, M. A. Urich, A. Breschi et al., Comparison of the transcriptional landscapes between human and mouse tissues, Proc Natl Acad Sci USA, vol.111, pp.17224-17233, 2014.

M. Griffith, J. R. Walker, N. C. Spies, B. J. Ainscough, and O. L. Griffith, Informatics for RNA Sequencing: A Web Resource for Analysis on the Cloud, PLoS Comput Biol, vol.11, p.1004393, 2015.

M. Tasnim, S. Ma, E. Yang, T. Jiang, and W. Li, Accurate inference of isoforms from multiple sample RNA-Seq data, BMC Genomics, vol.16, issue.2, p.15, 2015.

T. Griebel, B. Zacher, P. Ribeca, E. Raineri, V. Lacroix et al., Modelling and simulating generic RNA-Seq experiments with the flux simulator, Nucleic Acids Res, vol.40, pp.10073-83, 2012.
URL : https://hal.archives-ouvertes.fr/hal-00744755

B. Li and C. N. Dewey, RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome, BMC Bioinformatics, vol.12, p.323, 2011.

N. L. Barbosa-morais, M. Irimia, Q. Pan, H. Y. Xiong, S. Gueroussov et al., The evolutionary landscape of alternative splicing in vertebrate species, Science, vol.338, pp.1587-93, 2012.

J. Merkin, C. Russell, P. Chen, and C. B. Burge, Evolutionary dynamics of gene and isoform regulation in Mammalian tissues, Science, vol.338, pp.1593-1602, 2012.

D. Brawand, M. Soumillon, A. Necsulea, P. Julien, G. Csárdi et al., The evolution of gene expression levels in mammalian organs, Nature, vol.478, pp.343-351, 2011.

R. M. Kuhn, D. Haussler, and W. J. Kent, The UCSC genome browser and associated tools, Brief Bioinform, vol.14, pp.144-61, 2013.

M. Lizio, R. Deviatiiarov, H. Nagai, L. Galan, E. Arner et al., Systematic analysis of transcription start sites in avian development, PLoS Biol, vol.15, p.2002887, 2017.

T. Zhang, X. Zhang, K. Han, G. Zhang, J. Wang et al., Analysis of long noncoding RNA and mRNA using RNA sequencing during the differentiation of intramuscular preadipocytes in chicken, PLoS ONE, vol.12, p.172389, 2017.

Y. Wang, H. Xu, M. Wang, N. O. Otecko, L. Ye et al., Annotating long intergenic non-coding RNAs under artificial selection during chicken domestication, BMC Evol Biol, vol.17, p.192, 2017.

M. Calderon-dominguez, J. F. Mir, R. Fucho, M. Weber, D. Serra et al., Fatty acid metabolism and the basis of brown adipose tissue function, Adipocyte, vol.5, pp.98-118, 2016.

N. Dakovi?, M. Térézol, F. Pitel, V. Maillard, S. Elis et al., The loss of adipokine genes in the chicken genome and implications for insulin metabolism, Mol Biol Evol, vol.31, pp.2637-2683, 2014.

A. T. Ali, W. E. Hochfeld, R. Myburgh, and M. S. Pepper, Adipocyte and adipogenesis, Eur J Cell Biol, vol.92, pp.229-265, 2013.

M. Li, X. Sun, H. Cai, Y. Sun, M. Plath et al., Long non-coding RNA ADNCR suppresses adipogenic differentiation by targeting miR-204, Biochim Biophys Acta, vol.1859, pp.871-82, 2016.

S. Gesta, Y. Tseng, and C. R. Kahn, Developmental origin of fat: tracking obesity to its source, Cell, vol.131, pp.242-56, 2007.

R. Oelkrug, E. T. Polymeropoulos, and M. Jastroch, Brown adipose tissue: physiological function and evolutionary significance, Biochem Syst Environ Physiol, vol.185, pp.587-606, 2015.

N. V. Mezentseva, J. S. Kumaratilake, and S. A. Newman, The brown adipocyte differentiation pathway in birds: an evolutionary road not taken, BMC Biol, vol.6, p.17, 2008.

L. K. Townsend and D. C. Wright, Looking on the "brite" side exercise-induced browning of white adipose tissue, Pflugers Arch, 2018.

M. Sun and W. L. Kraus, From discovery to function: the expanding roles of long noncoding RNAs in physiology and disease, Endocr Rev, vol.36, pp.25-64, 2015.

I. Letunic and P. Bork, Interactive tree of life (iTOL) v3: an online tool for the display and annotation of phylogenetic and other trees, Nucleic Acids Res, vol.44, pp.242-245, 2016.

L. Fagerberg, B. M. Hallström, P. Oksvold, C. Kampf, D. Djureinovic et al., Analysis of the human tissue-specific expression by genome-wide integration of transcriptomics and antibody-based proteomics, Mol Cell Proteomics, vol.13, pp.397-406, 2014.

L. Sun, L. A. Goff, C. Trapnell, R. Alexander, K. A. Lo et al., Long noncoding RNAs regulate adipogenesis, Proc Natl Acad Sci USA, vol.110, pp.3387-92, 2013.

T. S. Mikkelsen, Z. Xu, X. Zhang, L. Wang, J. M. Gimble et al., Comparative epigenomic analysis of murine and human adipogenesis, Cell, vol.143, pp.156-69, 2010.

K. D. Macisaac, K. A. Lo, W. Gordon, S. Motola, T. Mazor et al., A quantitative model of transcriptional regulation reveals the influence of binding location on expression, PLoS Comput Biol, vol.6, p.1000773, 2010.

S. Xu, P. Chen, and L. Sun, Regulatory networks of non-coding RNAs in brown/beige adipogenesis, Biosci Rep, vol.35, 2015.

J. R. Alvarez-dominguez, Z. Bai, D. Xu, B. Yuan, K. A. Lo et al., De Novo Reconstruction of Adipose Tissue Transcriptomes Reveals Long Non-coding RNA Regulators of Brown Adipocyte Development, Cell Metab, vol.21, pp.764-76, 2015.

Z. Chen, Progress and prospects of long noncoding RNAs in lipid homeostasis, Molecular Metabolism, vol.5, pp.164-70, 2016.

A. Divoux, K. Karastergiou, H. Xie, W. Guo, R. J. Perera et al., Identification of a novel lncRNA in gluteal adipose tissue and evidence for its positive effect on preadipocyte differentiation, Obesity (Silver Spring), vol.22, pp.1781-1786, 2014.

D. R. Cooper, G. Carter, P. Li, R. Patel, J. E. Watson et al., Long Non-Coding RNA NEAT1 Associates with SRp40 to Temporally Regulate PPAR?2 Splicing during Adipogenesis in 3T3-L1 Cells, Genes (Basel), vol.5, pp.1050-63, 2014.

S. Wei, M. Du, Z. Jiang, G. J. Hausman, L. Zhang et al., Long noncoding RNAs in regulating adipogenesis: new RNAs shed lights on obesity, Cell Mol Life Sci, vol.73, pp.2079-87, 2016.

F. Yi, F. Yang, X. Liu, H. Chen, J. T. Jiang et al., RNA-seq identified a super-long intergenic transcript functioning in adipogenesis, RNA Biol, vol.10, pp.991-1001, 2013.

H. Yoshioka and Y. Yoshiko, The Roles of Long Non-Protein-Coding RNAs in OsteoAdipogenic Lineage Commitment, Int J Mol Sci, vol.18, 2017.

T. Sun, M. Fu, A. L. Bookout, S. A. Kliewer, and D. J. Mangelsdorf, MicroRNA let-7 Regulates 3T3-L1 Adipogenesis, Mol Endocrinol, vol.23, pp.925-956, 2009.

T. Xiao, L. Liu, H. Li, Y. Sun, H. Luo et al., Long Noncoding RNA ADINR Regulates Adipogenesis by Transcriptionally Activating C/EBP?. Stem Cell Reports, vol.5, pp.856-65, 2015.

R. Cai, Y. Sun, N. Qimuge, G. Wang, Y. Wang et al., Adiponectin AS lncRNA inhibits adipogenesis by transferring from nucleus to cytoplasm and attenuating Adiponectin mRNA translation, Biochim Biophys Acta, vol.1863, pp.420-452, 2018.

J. Chen, X. Cui, C. Shi, L. Chen, L. Yang et al., Differential lncRNA expression profiles in brown and white adipose tissues, Mol Genet Genomics, vol.290, pp.699-707, 2015.

H. Gao, A. Kerr, H. Jiao, C. Hon, M. Rydén et al., Long Non-Coding RNAs Associated with Metabolic Traits in Human White Adipose Tissue, EBioMedicine, vol.30, pp.248-60, 2018.

G. Liu, X. Zheng, Y. Xu, J. Lu, J. Chen et al., Long non-coding RNAs expression profile in HepG2 cells reveals the potential role of long non-coding RNAs in the cholesterol metabolism, Chin Med J, vol.128, pp.91-98, 2015.

X. Zhao, S. Li, G. Wang, Y. Q. Lin, and J. D. , A long noncoding RNA transcriptional regulatory circuit drives thermogenic adipocyte differentiation, Mol Cell, vol.55, pp.372-82, 2014.

S. Li, L. Mi, L. Yu, Q. Yu, T. Liu et al., Zbtb7b engages the long noncoding RNA Blnc1 to drive brown and beige fat development and thermogenesis, Proc Natl Acad Sci, vol.114, pp.7111-7131, 2017.

X. Zhao, S. Li, J. L. Delproposto, T. Liu, M. L. Porsche et al., The long noncoding RNA Blnc1 orchestrates homeostatic adipose tissue remodeling to preserve metabolic health. Mol Metab, 2018.

L. Mi, X. Zhao, S. Li, G. Yang, and J. D. Lin, Conserved function of the long noncoding RNA Blnc1 in brown adipocyte differentiation, Mol Metab, vol.6, pp.101-111, 2017.

M. Li, Z. Xie, P. Wang, J. Li, W. Liu et al., The long noncoding RNA GAS5 negatively regulates the adipogenic differentiation of MSCs by modulating the miR-18a/CTGF axis as a ceRNA, Cell Death Dis, vol.9, p.554, 2018.

L. You, Y. Zhou, X. Cui, X. Wang, Y. Sun et al., GM13133 is a negative regulator in mouse white adipocytes differentiation and drives the characteristics of brown adipocytes, J Cell Physiol, vol.233, pp.313-337, 2018.

W. Liu, C. Ma, B. Yang, C. Yin, B. Zhang et al., LncRNA Gm15290 sponges miR-27b to promote PPAR?-induced fat deposition and contribute to body weight gain in mice, Biochem Biophys Res Commun, vol.493, pp.1168-75, 2017.

Y. Huang, Y. Zheng, J. C. Li, X. Jia, L. Li et al., Long Non-coding RNA H19 Inhibits Adipocyte Differentiation of Bone Marrow Mesenchymal Stem Cells through Epigenetic Modulation of Histone Deacetylases, Sci Rep, vol.6, p.28897, 2016.

Y. Wang, W. Liu, Y. Liu, J. Cui, Z. Zhao et al., Long noncoding RNA H19 mediates LCoR to impact the osteogenic and adipogenic differentiation of mBMSCs in mice through sponging miR-188, J Cell Physiol, vol.233, pp.7435-7481, 2018.

E. Schmidt, I. Dhaouadi, I. Gaziano, M. Oliverio, P. Klemm et al., LincRNA H19 protects from dietary obesity by constraining expression of monoallelic genes in brown fat, Nat Commun, vol.9, p.3622, 2018.

N. Li, S. Hébert, J. Song, C. L. Kleinman, and S. Richard, Transcriptome profiling in preadipocytes identifies long noncoding RNAs as Sam68 targets, Oncotarget, vol.8, pp.81994-2005, 2017.

N. Nuermaimaiti, J. Liu, X. Liang, Y. Jiao, D. Zhang et al., Effect of lncRNA HOXA11-AS1 on adipocyte differentiation in human adipose-derived stem cells, Biochem Biophys Res Commun, vol.495, pp.1878-84, 2018.

X. Zhu, Y. Yan, C. D. Ai, C. Lu, X. Xu et al., Long non-coding RNA HoxA-AS3 interacts with EZH2 to regulate lineage commitment of mesenchymal stem cells, Oncotarget, vol.7, pp.63561-70, 2016.

X. Zhang, C. Xue, J. Lin, J. F. Ferguson, A. Weiner et al., Interrogation of nonconserved human adipose lincRNAs identifies a regulatory role of linc-ADAL in adipocyte metabolism, Sci Transl Med, vol.10, 2018.

Z. Bai, X. Chai, M. J. Yoon, H. Kim, K. A. Lo et al., Dynamic transcriptome changes during adipose tissue energy expenditure reveal critical roles for long noncoding RNA regulators, PLoS Biol, vol.15, p.2002176, 2017.

Y. Xiong, F. Yue, Z. Jia, Y. Gao, J. W. Hu et al., A novel brown adipocyte-enriched long non-coding RNA that is required for brown adipocyte differentiation and sufficient to drive thermogenic gene program in white adipocytes, Biochim Biophys Acta Mol Cell Biol Lipids, vol.1863, pp.409-428, 2018.

Z. Li, J. C. Chen, S. Zheng, Y. Huang, Y. Jia et al., Long non-coding RNA MEG3 inhibits adipogenesis and promotes osteogenesis of human adipose-derived mesenchymal stem cells via miR-140-5p, Mol Cell Biochem, vol.433, pp.51-60, 2017.

Y. Huang, C. Jin, Y. Zheng, X. Li, S. Zhang et al., Knockdown of lncRNA MIR31HG inhibits adipocyte differentiation of human adipose-derived stem cells via histone modification of FABP4, Sci Rep, vol.7, p.8080, 2017.

R. Gernapudi, B. Wolfson, Y. Zhang, Y. Yao, P. Yang et al., MicroRNA 140 Promotes Expression of Long Noncoding RNA NEAT1 in Adipogenesis, Mol Cell Biol, vol.36, pp.30-38, 2016.

F. F. Firmin, F. Oger, C. Gheeraert, J. Dubois-chevalier, A. Vercoutter-edouart et al., The RBM14/CoAA-interacting, long intergenic non-coding RNA Paral1 regulates adipogenesis and coactivates the nuclear receptor PPAR?, Sci Rep, vol.7, p.14087, 2017.

W. Pang, L. Lin, Y. Xiong, N. Wei, Y. Wang et al., Knockdown of PU.1 AS lncRNA inhibits adipogenesis through enhancing PU.1 mRNA translation, J Cell Biochem, vol.114, pp.2500-2512, 2013.

Y. Liu, Y. Ji, M. Li, M. Wang, X. Yi et al., Integrated analysis of long noncoding RNA and mRNA expression profile in children with obesity by microarray analysis, Sci Rep, vol.8, p.8750, 2018.

Q. Wang, Q. Yang, G. Chen, Z. Du, M. Ren et al., LncRNA expression profiling of BMSCs in osteonecrosis of the femoral head associated with increased adipogenic and decreased osteogenic differentiation, Sci Rep, vol.8, p.9127, 2018.

S. Liu, L. Sheng, H. Miao, T. L. Saunders, O. A. Macdougald et al., SRA gene knockout protects against diet-induced obesity and improves glucose tolerance, J Biol Chem, vol.289, pp.13000-13009, 2014.

B. Xu, I. Gerin, H. Miao, D. Vu-phan, C. N. Johnson et al., Multiple roles for the noncoding RNA SRA in regulation of adipogenesis and insulin sensitivity, PLoS ONE, vol.5, p.14199, 2010.

J. Wang, L. Hua, J. Chen, J. Zhang, X. Bai et al., Identification and characterization of long non-coding RNAs in subcutaneous adipose tissue from castrated and intact full-sib pair Huainan male pigs, BMC Genomics, vol.18, p.542, 2017.

Y. Liu, Y. Wang, X. He, S. Zhang, K. Wang et al.,

C. Ebp, -? feedback loop modulates the adipogenic differentiation process in human adipose tissue-derived mesenchymal stem cells, Stem Cell Research, vol.32, pp.35-42, 2018.

J. Chen, Y. Liu, S. Lu, L. Yin, C. Zong et al., The role and possible mechanism of lncRNA U90926 in modulating 3T3-L1 preadipocyte differentiation, Int J Obes, vol.41, pp.299-308, 2017.

V. L. Battula, Y. Chen, G. Cabreira-m-da, V. Ruvolo, Z. Wang et al., Connective tissue growth factor regulates adipocyte differentiation of mesenchymal stromal cells and facilitates leukemia bone marrow engraftment, Blood, vol.122, pp.357-66, 2013.

H. Cao, S. Zhang, S. Shan, C. Sun, Y. Li et al., Ligand-dependent corepressor (LCoR) represses the transcription factor C/EBP? during early adipocyte differentiation, J Biol Chem, vol.292, pp.18973-87, 2017.

X. Hui, M. Zhang, P. Gu, K. Li, Y. Gao et al., Adipocyte SIRT1 controls systemic insulin sensitivity by modulating macrophages in adipose tissue, EMBO Rep, vol.18, pp.645-57, 2017.

F. Wang and Q. Tong, Transcription factor PU.1 is expressed in white adipose and inhibits adipocyte differentiation, American journal of physiology Cell physiology, vol.295, 2008.

Y. Zhang, X. Li, S. Qian, L. Guo, H. Huang et al., Down-Regulation of Type I Runx2 Mediated by Dexamethasone Is Required for 3T3-L1 Adipogenesis, Molecular endocrinology, vol.26, pp.798-808, 2012.

Y. Chen, F. Siegel, S. Kipschull, B. Haas, H. Fröhlich et al., miR-155 regulates differentiation of brown and beige adipocytes via a bistable circuit, Nat Commun, vol.4, p.1769, 2013.

L. M. Szafron, A. Balcerak, E. A. Grzybowska, B. Pienkowska-grela, A. Felisiak-golabek et al., The Novel Gene CRNDE Encodes a Nuclear Peptide (CRNDEP) Which Is Overexpressed in Highly Proliferating Tissues, PLoS ONE, vol.10, p.127475, 2015.

H. Liu, H. Li, J. L. Li, G. Hu, S. Ning et al., Long Noncoding RNA GAS5 Suppresses 3T3-L1 Cells Adipogenesis Through miR-21a-5p/PTEN Signal Pathway, DNA Cell Biol, 2018.

Z. Xu, S. Yu, C. Hsu, J. Eguchi, and E. D. Rosen, The orphan nuclear receptor chicken ovalbumin upstream promoter-transcription factor II is a critical regulator of adipogenesis, Proc Natl Acad Sci USA, vol.105, pp.2421-2427, 2008.

B. B. Brandão, B. A. Guerra, and M. A. Mori, Shortcuts to a functional adipose tissue: The role of small non-coding RNAs, Redox Biol, vol.12, pp.82-102, 2017.

M. Sauvageau, L. A. Goff, S. Lodato, B. Bonev, A. F. Groff et al., Multiple knockout mouse models reveal lincRNAs are required for life and brain development, Elife, vol.2, p.1749, 2013.

C. Cooper, D. Vincett, Y. Yan, M. K. Hamedani, Y. Myal et al., Steroid Receptor RNA Activator bi-faceted genetic system: Heads or Tails?, Biochimie, vol.93, pp.1973-80, 2011.

F. Bost, M. Aouadi, L. Caron, and B. Binétruy, The role of MAPKs in adipocyte differentiation and obesity, Biochimie, vol.87, pp.51-57, 2005.
URL : https://hal.archives-ouvertes.fr/inserm-00000011

S. Liu, R. Xu, I. Gerin, W. P. Cawthorn, O. A. Macdougald et al., SRA regulates adipogenesis by modulating p38/JNK phosphorylation and stimulating insulin receptor gene expression and downstream signaling, PLoS ONE, vol.9, p.95416, 2014.

R. F. , Essai d'estimation directe des différents éléments de la carcasse du poulet en vue d'apprécier son rendement en viande, Ann Zootech, vol.13, pp.355-66, 1964.

B. Leclercq, J. C. Blum, and J. P. Boyer, Selecting broilers for low or high abdominal fat: Initial observations, British Poultry Science, vol.21, pp.107-120, 1980.

B. Leclercq and J. Simon, Selecting broilers for low or high abdominal fat: observations on the hens during the breeding period, Annales de zootechnie, pp.161-170, 1982.
URL : https://hal.archives-ouvertes.fr/hal-00888141

H. Li, B. Handsaker, A. Wysoker, T. Fennell, J. Ruan et al., The Sequence Alignment/Map format and SAMtools, Bioinformatics, vol.25, pp.2078-2087, 2009.

S. Lê, J. Josse, and F. Husson, FactoMineR: An R package for multivariate analysis, Journal of Statistical Software, vol.25, pp.1-18, 2008.

M. D. Robinson, D. J. Mccarthy, and G. K. Smyth, edgeR: a Bioconductor package for differential expression analysis of digital gene expression data, Bioinformatics, vol.26, pp.139-179, 2010.

Y. Benjamini and Y. Hochberg, Controlling the False Discovery Rate: A Practical and Powerful Approach to Multiple Testing, Journal of the Royal Statistical Society, vol.57, pp.289-300, 1995.

D. W. Huang, B. T. Sherman, and R. A. Lempicki, Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources, Nat Protoc, vol.4, pp.44-57, 2009.

R. J. Kinsella, A. Kahari, S. Haider, J. Zamora, G. Proctor et al., Ensembl BioMarts: a hub for data retrieval across taxonomic space, Database, pp.30-030, 2011.

K. B. Wicher and E. Fries, Haptoglobin, a hemoglobin-binding plasma protein, is present in bony fish and mammals but not in frog and chicken, Proc Natl Acad Sci USA, vol.103, pp.4168-73, 2006.

P. Mamczur, A. Gamian, J. Kolodziej, P. Dziegiel, and D. Rakus, Nuclear localization of aldolase A correlates with cell proliferation, Biochim Biophys Acta, vol.1833, pp.2812-2834, 2013.

L. A. Cogburn, D. N. Smarsh, X. Wang, N. Trakooljul, W. Carré et al., Transcriptional profiling of liver in riboflavin-deficient chicken embryos explains impaired lipid utilization, energy depletion, massive hemorrhaging, and delayed feathering, BMC Genomics, vol.19, p.177, 2018.

T. Grahn, Y. Zhang, M. Lee, A. G. Sommer, G. Mostoslavsky et al., FSP27 and PLIN1 interaction promotes the formation of large lipid droplets in human adipocytes, Biochem Biophys Res Commun, vol.432, pp.296-301, 2013.

A. Khan, W. M. Khan, M. Ayub, M. Humayun, and M. Haroon, Ferritin Is a Marker of Inflammation rather than Iron Deficiency in Overweight and Obese People, J Obes, 2016.

S. Tartare-deckert, C. Chavey, M. N. Monthouel, N. Gautier, and E. Van-obberghen, The matricellular protein SPARC/osteonectin as a newly identified factor up-regulated in obesity, J Biol Chem, vol.276, pp.22231-22238, 2001.

W. H. Moolenaar and A. Perrakis, Insights into autotaxin: how to produce and present a lipid mediator, Nat Rev Mol Cell Biol, vol.12, pp.674-683, 2011.

H. Matsuda, M. Sato, M. Yakushiji, M. Koshiguchi, S. Hirai et al., Regulation of rat hepatic ?-amino-?-carboxymuconate-?-semialdehyde decarboxylase, a key enzyme in the tryptophan-NAD pathway, by dietary cholesterol and sterol regulatory element-binding protein-2, Eur J Nutr, vol.53, pp.469-77, 2014.

H. Thorvaldsdottir, J. T. Robinson, and J. P. Mesirov, Integrative Genomics Viewer (IGV): highperformance genomics data visualization and exploration, Briefings in Bioinformatics, vol.14, pp.178-92, 2013.

S. Parekh, C. Ziegenhain, B. Vieth, W. Enard, and I. Hellmann, The impact of amplification on differential expression analyses by RNA-seq, Sci Rep, vol.6, p.25533, 2016.

V. Marx, How to deduplicate PCR, Nat Methods, vol.14, pp.473-479, 2017.

M. Ebbert, M. E. Wadsworth, L. A. Staley, K. L. Hoyt, B. Pickett et al., Evaluating the necessity of PCR duplicate removal from next-generation sequencing data and a comparison of approaches, BMC Bioinformatics, vol.17, p.239, 2016.

W. Mclaren, L. Gil, S. E. Hunt, H. S. Riat, G. Ritchie et al., The Ensembl Variant Effect Predictor, Genome Biology, vol.17, p.122, 2016.

P. Cingolani, A. Platts, L. L. Wang, M. Coon, T. Nguyen et al., A program for annotating and predicting the effects of single nucleotide polymorphisms, SnpEff. Fly (Austin), vol.6, pp.80-92, 2012.

P. C. Ng and S. Henikoff, SIFT: predicting amino acid changes that affect protein function, Nucleic Acids Res, vol.31, pp.3812-3816, 2003.

A. Albrecht and U. A. Ørom, Bidirectional expression of long ncRNA/protein-coding gene pairs in cancer, Brief Funct Genomics, vol.15, pp.167-73, 2016.