Q. A. Wang, C. Tao, R. K. Gupta, and P. E. Scherer, Tracking adipogenesis during white adipose tissue development, expansion and regeneration, Nat Med, vol.19, pp.1338-1344, 2013.
DOI : 10.1038/nm.3324

URL : http://europepmc.org/articles/pmc4075943?pdf=render

V. Pellegrinelli, S. Carobbio, and A. Vidal-puig, Adipose tissue plasticity: how fat depots respond differently to pathophysiological cues, Diabetologia, vol.59, pp.1075-1088, 2016.
DOI : 10.1007/s00125-016-3933-4

URL : https://link.springer.com/content/pdf/10.1007%2Fs00125-016-3933-4.pdf

S. Virtue and A. Vidal-puig, Adipose tissue expandability, lipotoxicity and the Metabolic Syndrome-an allostatic perspective, Biochim. Biophys. Acta, vol.1801, pp.338-349, 2010.
DOI : 10.1016/j.bbalip.2009.12.006

S. Nishimura, In vivo imaging in mice reveals local cell dynamics and inflammation in obese adipose tissue, J Clin Invest, vol.118, pp.710-721, 2008.

K. Sun, C. M. Kusminski, and P. E. Scherer, Adipose tissue remodeling and obesity, J Clin Invest, vol.121, pp.2094-2101, 2011.

L. B. Salans, J. L. Knittle, and J. Hirsch, The role of adipose cell size and adipose tissue insulin sensitivity in the carbohydrate intolerance of human obesity, J. Clin. Invest, vol.47, pp.153-165, 1968.

M. J. Lee, Y. Wu, and S. K. Fried, Adipose tissue remodeling in pathophysiology of obesity, Curr. Opin. Clin. Nutr. Metab Care, vol.13, pp.371-376, 2010.

F. Oger, Peroxisome proliferator-activated receptor gamma regulates genes involved in insulin/insulin-like growth factor signaling and lipid metabolism during adipogenesis through functionally distinct enhancer classes, J Biol Chem, vol.289, pp.708-722, 2014.

Q. Q. Tang and M. D. Lane, Adipogenesis: from stem cell to adipocyte, Annu Rev Biochem, vol.81, pp.715-736, 2012.
DOI : 10.1146/annurev-biochem-052110-115718

R. Siersbaek, Transcription factor cooperativity in early adipogenic hotspots and super-enhancers, Cell Rep, vol.7, pp.1443-1455, 2014.

J. Dubois-chevalier, A dynamic CTCF chromatin binding landscape promotes DNA hydroxymethylation and transcriptional induction of adipocyte differentiation, Nucleic Acids Res, vol.42, pp.10943-10959, 2014.
URL : https://hal.archives-ouvertes.fr/inserm-02159046

A. A. Serandour, Dynamic hydroxymethylation of DNA marks differentiation-driven enhancers, Nucl. Acid Res, vol.40, pp.8255-8265, 2012.

A. Fatica and I. Bozzoni, Long non-coding RNAs: new players in cell differentiation and development, Nat Rev Genet, vol.15, pp.7-21, 2014.
URL : https://hal.archives-ouvertes.fr/pasteur-01160208

J. L. Rinn and H. Y. Chang, Genome regulation by long noncoding RNAs, Annu Rev Biochem, vol.81, pp.145-166, 2012.
DOI : 10.1146/annurev-biochem-051410-092902

URL : http://europepmc.org/articles/pmc3858397?pdf=render

E. Hacisuleyman, Topological organization of multichromosomal regions by the long intergenic noncoding RNA Firre, Nat Struct Mol Biol, vol.21, pp.198-206, 2014.

C. M. Clemson, An architectural role for a nuclear noncoding RNA: NEAT1 RNA is essential for the structure of paraspeckles, Mol Cell, vol.33, pp.717-726, 2009.

M. Mele and J. L. Rinn, Cat's Cradling" the 3D Genome by the Act of LncRNA Transcription, Mol Cell, vol.62, pp.657-664, 2016.

D. R. Cooper, Long Non-Coding RNA NEAT1 Associates with SRp40 to Temporally Regulate PPARgamma2 Splicing during Adipogenesis in 3T3-L1 Cells, Genes (Basel), vol.5, pp.1050-1063, 2014.

, Scientific RepoRts |, vol.7

T. Xiao, Long Noncoding RNA ADINR Regulates Adipogenesis by Transcriptionally Activating C/EBPalpha. Stem Cell Reports, vol.5, pp.856-865, 2015.

J. Chen, The role and possible mechanism of lncRNA U90926 in modulating 3T3-L1 preadipocyte differentiation, Int J Obes (Lond), vol.41, pp.299-308, 2017.

T. S. Mikkelsen, Comparative epigenomic analysis of murine and human adipogenesis, Cell, vol.143, pp.156-169, 2010.
DOI : 10.1016/j.cell.2010.09.006

URL : https://doi.org/10.1016/j.cell.2010.09.006

L. L. Chen and R. N. Noncoding, Localization and Function, Trends Biochem Sci, vol.41, pp.761-772, 2016.

C. S. Bond and A. H. Fox, Paraspeckles: nuclear bodies built on long noncoding RNA, J Cell Biol, vol.186, pp.637-644, 2009.
DOI : 10.1083/jcb.200906113

URL : http://jcb.rupress.org/content/186/5/637.full.pdf

T. Iwasaki, W. W. Chin, and L. Ko, Identification and characterization of RRM-containing coactivator activator (CoAA) as TRBPinteracting protein, and its splice variant as a coactivator modulator (CoAM), J Biol Chem, vol.276, pp.33375-33383, 2001.

D. Auboeuf, CoAA, a Nuclear Receptor Coactivator Protein at the Interface of Transcriptional Coactivation and RNA Splicing, Molecular and Cellular Biology, vol.24, pp.442-453, 2004.

B. Lefebvre, Proteasomal degradation of retinoid X receptor alpha reprograms transcriptional activity of PPARgamma in obese mice and humans, J Clin Invest, vol.120, pp.1454-1468, 2010.
URL : https://hal.archives-ouvertes.fr/inserm-00472906

E. Dalmas, Irf5 deficiency in macrophages promotes beneficial adipose tissue expansion and insulin sensitivity during obesity, Nat Med, vol.21, pp.610-618, 2015.
DOI : 10.1038/nm.3829

C. C. Hon, An atlas of human long non-coding RNAs with accurate 5? ends, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01607778

J. Eeckhoute, F. Oger, B. Staels, and P. Lefebvre, Coordinated Regulation of PPARgamma Expression and Activity through Control of Chromatin Structure in Adipogenesis and Obesity, PPAR Res, vol.164140, 2012.

U. Perron, P. Provero, and I. Molineris, In silico prediction of lncRNA function using tissue specific and evolutionary conserved expression, BMC Bioinformatics, vol.18, 2017.

A. K. Haakonsson, M. Stahl-madsen, R. Nielsen, A. Sandelin, and S. Mandrup, Acute genome-wide effects of rosiglitazone on PPARgamma transcriptional networks in adipocytes, Mol Endocrinol, vol.27, pp.1536-1549, 2013.

C. Vernochet, C/EBPalpha and the corepressors CtBP1 and CtBP2 regulate repression of select visceral white adipose genes during induction of the brown phenotype in white adipocytes by peroxisome proliferator-activated receptor gamma agonists, Mol Cell Biol, vol.29, pp.4714-4728, 2009.

D. Auboeuf, A. Honig, S. M. Berget, and B. W. O'malley, Coordinate Regulation of Transcription and Splicing by Steroid Receptor Coregulators, Science, vol.298, pp.416-419, 2002.

M. Perani, The proto-oncoprotein SYT interacts with SYT-interacting protein/co-activator activator (SIP/CoAA), a human nuclear receptor co-activator with similarity to EWS and TLS/FUS family of proteins, J Biol Chem, vol.280, pp.42863-42876, 2005.

Y. Hasegawa, The matrix protein hnRNP U is required for chromosomal localization of Xist RNA, Dev Cell, vol.19, pp.469-476, 2010.

X. Li, L. H. Hoeppner, E. D. Jensen, R. Gopalakrishnan, and J. J. Westendorf, Co-activator activator (CoAA) prevents the transcriptional activity of Runt domain transcription factors, J Cell Biochem, vol.108, pp.378-387, 2009.

M. J. Herriges, Long noncoding RNAs are spatially correlated with transcription factors and regulate lung development, Genes Dev, vol.28, pp.1363-1379, 2014.
DOI : 10.1101/gad.238782.114

URL : http://genesdev.cshlp.org/content/28/12/1363.full.pdf

J. M. Engreitz, Local regulation of gene expression by lncRNA promoters, transcription and splicing, Nature, vol.539, pp.452-455, 2016.

A. H. Fox and A. I. Lamond, Paraspeckles. Cold Spring Harbor Perspectives in Biology, vol.2, 2010.

S. Y. Kim, miR-27a is a negative regulator of adipocyte differentiation via suppressing PPARgamma expression, Biochem Biophys Res Commun, vol.392, pp.323-328, 2010.

G. Shiratsuchi, K. Takaoka, T. Ashikawa, H. Hamada, and D. Kitagawa, RBM14 prevents assembly of centriolar protein complexes and maintains mitotic spindle integrity, EMBO J, vol.34, pp.97-114, 2015.
DOI : 10.15252/embj.201488979

URL : http://emboj.embopress.org/content/34/1/97.full.pdf

J. Wang, RNA-binding protein PSPC1 promotes the differentiation-dependent nuclear export of adipocyte RNAs, J Clin Invest, vol.127, pp.987-1004, 2017.

M. Matsui and D. R. Corey, Non-coding RNAs as drug targets, Nat Rev Drug Discov, vol.16, pp.167-179, 2017.
DOI : 10.1038/nrd.2016.117

URL : http://europepmc.org/articles/pmc5831170?pdf=render

J. Ding, TNF-alpha and IL-1beta inhibit RUNX2 and collagen expression but increase alkaline phosphatase activity and mineralization in human mesenchymal stem cells, Life Sci, vol.84, pp.499-504, 2009.
DOI : 10.1016/j.lfs.2009.01.013

L. Guevel and R. , Inactivation of the Nuclear Orphan Receptor COUP-TFII by Small Chemicals, ACS Chem Biol, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01497231

W. Zhao, Genome-wide expression profiling revealed peripheral effects of cannabinoid receptor 1 inverse agonists in improving insulin sensitivity and metabolic parameters, Mol Pharmacol, vol.78, pp.350-359, 2010.

U. Mudunuri, A. Che, M. Yi, and R. M. Stephens, bioDBnet: the biological database network, Bioinformatics, vol.25, pp.555-556, 2009.
DOI : 10.1093/bioinformatics/btn654

URL : https://academic.oup.com/bioinformatics/article-pdf/25/4/555/16892063/btn654.pdf

A. Subramanian, Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles, Proc. Natl. Acad. Sci. USA, vol.102, pp.15545-15550, 2005.
DOI : 10.1073/pnas.0506580102

URL : http://www.pnas.org/content/102/43/15545.full.pdf

H. C. Roh, Simultaneous Transcriptional and Epigenomic Profiling from Specific Cell Types within Heterogeneous Tissues In Vivo, Cell Rep, vol.18, pp.1048-1061, 2017.

K. A. Lo, Analysis of in vitro insulin-resistance models and their physiological relevance to in vivo diet-induced adipose insulin resistance, Cell Rep, vol.5, pp.259-270, 2013.

L. Kong, CPC: assess the protein-coding potential of transcripts using sequence features and support vector machine, Nucleic Acids Res, vol.35, pp.345-349, 2007.

L. Wang, CPAT: Coding-Potential Assessment Tool using an alignment-free logistic regression model, Nucleic Acids Res, vol.41, 2013.
DOI : 10.1093/nar/gkt006

URL : https://academic.oup.com/nar/article-pdf/41/6/e74/25339876/gkt006.pdf

S. Caron, Farnesoid X receptor inhibits the transcriptional activity of carbohydrate response element binding protein in human hepatocytes, Mol. Cell Biol, vol.33, pp.2202-2211, 2013.
URL : https://hal.archives-ouvertes.fr/inserm-00806064

K. Verreman, The coactivator activator CoAA regulates PEA3 group member transcriptional activity, Biochem J, vol.439, pp.469-477, 2011.
DOI : 10.1042/bj20110728

URL : https://hal.archives-ouvertes.fr/hal-00633602

C. Depoix, M. H. Delmotte, P. Formstecher, and P. Lefebvre, Control of retinoic acid receptor heterodimerization by ligand-induced structural transitions. a novel mechanism of action for retinoid antagonists, J. Biol. Chem, vol.276, pp.9452-9459, 2001.

S. Hauser, Degradation of the peroxisome proliferator-activated receptor gamma is linked to ligand-dependent activation, J. Biol. Chem, vol.275, pp.18527-18533, 2000.

M. Fan, Suppression of mitochondrial respiration through recruitment of p160 myb binding protein to PGC-1alpha: modulation by p38 MAPK, Genes Dev, vol.18, pp.278-289, 2004.

P. Sacchetti, R. Carpentier, P. Segard, C. Olive-cren, and P. Lefebvre, Multiple signaling pathways regulate the transcriptional activity of the orphan nuclear receptor NURR1, Nucleic Acids Res, vol.34, pp.5515-5527, 2006.

R. M. Kuhn, The UCSC genome browser database: update, Nucleic Acids Res, vol.35, pp.668-673, 2007.