A. Siepel, G. Bejerano, J. S. Pedersen, A. S. Hinrichs, M. Hou et al., Evolutionarily conserved elements in vertebrate, insect, worm, and yeast genomes, Genome Res, vol.15, pp.1034-50, 2005.

L. A. Hindorff, P. Sethupathy, H. A. Junkins, E. M. Ramos, J. P. Mehta et al., Potential etiologic and functional implications of genome-wide association loci for human diseases and traits, Proc Natl Acad Sci, vol.106, pp.9362-9369, 2009.

M. T. Maurano, R. Humbert, E. Rynes, R. E. Thurman, E. Haugen et al., Systematic localization of common disease-associated variation in regulatory DNA, Science, vol.337, pp.1190-95, 2012.

, The ENCODE Project, Consortium. An integrated encyclopedia of DNA elements in the human genome, Nature, vol.489, pp.57-74, 2012.

F. Yue, Y. Cheng, A. Breschi, J. Vierstra, W. Wu et al., A comparative encyclopedia of DNA elements in the mouse genome, Nature, vol.515, pp.355-64, 2014.

M. B. Gerstein, J. Rozowsky, K. K. Yan, D. Wang, C. Cheng et al., Comparative analysis of the transcriptome across distant species, Nature, vol.512, pp.445-453, 2014.

B. Vernot, A. B. Stergachis, M. T. Maurano, J. Vierstra, S. Neph et al., Personal and population genomics of human regulatory variation

, Genome Res, vol.22, pp.1689-97, 2012.

A. Kundaje, W. Meuleman, J. Ernst, M. Bilenky, Y. A. Heravi-moussavi et al., Integrative analysis of 111 reference human epigenomes, Nature, vol.518, pp.317-347, 2015.

A. B. Stergachis, S. Neph, R. Sandstrom, E. Haugen, A. P. Reynolds et al., Conservation of trans-acting circuitry during mammalian regulatory evolution, Nature, vol.515, pp.365-70, 2014.

Y. Cheng, Z. Ma, B. H. Kim, W. Wu, P. Cayting et al., Principles of regulatory information conservation between mouse and human, Nature, vol.515, pp.371-376, 2014.

D. Schmidt, M. D. Wilson, B. Ballester, P. C. Schwalie, G. D. Brown et al., Five-vertebrate ChIP-seq reveals the evolutionary dynamics of transcription factor binding, Science, vol.328, pp.1036-1076, 2010.
URL : https://hal.archives-ouvertes.fr/hal-01615156

F. The and . Consortium, Functional Annotation of Animal Genomes (FAANG): a coordinated international action to accelerate genome to phenome, 2019.

L. Andersson, A. L. Archibald, C. D. Bottema, R. Brauning, S. C. Burgess et al., Coordinated international action to accelerate genome-to-phenome with FAANG, the Functional Annotation of Animal Genomes project, Genome Biol, vol.16, p.57, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01194102

C. K. Tuggle, E. Giuffra, S. N. White, L. Clarke, H. Zhou et al., GO-FAANG meeting: a gathering on functional annotation of animal genomes, Anim Genet, vol.47, pp.528-561, 2016.

C. Kern, Y. Wang, J. Chitwood, I. Korf, M. Delany et al.,

, Genome-wide identification of tissue-specific long non-coding RNA in three farm animal species, BMC Genomics, vol.19, p.684, 2018.

E. Giuffra, C. K. Tuggle, and . Consortium, Functional annotation of animal genomes (FAANG): current achievements and roadmap, Ann Rev Anim Biosci, vol.7, pp.65-88, 2019.

P. Harrison, J. Fan, D. Richardson, L. Clarke, D. Zerbino et al., establishing metadata standards, validation and best practices for the farmed and companion animal community, Anim Genet, vol.49, pp.520-526, 2018.

F. The and . Consortium, The FAANG Data Coordination Center, 2019.

, FR-AgENCODE: a FAANG pilot project for the annotation of livestock genomes, The FR-AgENCODE group, 2019.

J. D. Buenrostro, P. G. Giresi, L. C. Zaba, H. Y. Chang, and W. J. Greenleaf, Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position, Nat Methods, vol.10, pp.1213-1221, 2013.

E. Lieberman-aiden, V. Berkum, N. L. Williams, L. Imakaev, M. Ragoczy et al., Comprehensive mapping of long-range interactions reveals folding principles of the human genome, Science, vol.326, pp.289-93, 2009.

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.

A. Dobin and T. R. Gingeras, Mapping RNA-seq reads with STAR, Curr Protocol Bioinform, vol.51, pp.11-15, 2015.

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.

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

A. Breschi, S. Djebali, J. Gillis, D. D. Pervouchine, A. Dobin et al., Gene-specific patterns of expression variation across organs and species, Genome Biol, vol.17, p.151, 2016.

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, vol.111, pp.17224-17233, 2014.

P. H. Sudmant, M. S. Alexis, and C. B. Burge, Meta-analysis of RNA-seq expression data across species, tissues and studies, Genome Biol, vol.16, p.287

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.

M. Melé, P. G. Ferreira, F. Reverter, D. S. Deluca, J. Monlong et al., The human transcriptome across tissues and individuals, Science, vol.348, pp.660-665, 2015.

W. Gerner, T. Käser, and A. Saalmüller, Porcine T lymphocytes and NK cells -an update, Dev Comp Immunol, vol.33, pp.310-330, 2009.

E. Guzman, J. Hope, G. Taylor, A. L. Smith, C. Cubillos-zapata et al., Bovine ? ? T cells are a major regulatory T cell subset, J Immunol, vol.193, pp.208-230, 2014.

M. Kapushesky, T. Adamusiak, T. Burdett, A. Culhane, A. Farne et al., Gene Expression Atlas update -a value-added database of microarray and sequencing-based functional genomics experiments, Nucleic Acids Res, vol.40, pp.1077-81, 2011.

R. Petryszak, T. Burdett, B. Fiorelli, N. A. Fonseca, M. Gonzalez-porta et al., Expression Atlas update -a database of gene and transcript expression from microarray-and sequencing-based functional genomics experiments, Nucleic Acids Res, vol.42, pp.926-958, 2014.

S. Djebali, C. A. Davis, A. Merkel, A. Dobin, T. Lassmann et al., Landscape of transcription in human cells, Nature, vol.489, p.101, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01216755

V. Wucher, F. Legeai, B. Hédan, G. Rizk, L. Lagoutte et al., FEELnc: a tool for long non-coding RNA annotation and its application to the dog transcriptome, Nucleic Acids Res, vol.45, p.57, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01532061

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 Res, vol.22, pp.1775-89, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01205054

K. Muret, C. Klopp, V. Wucher, D. Esquerré, F. Legeai et al., Long noncoding RNA repertoire in chicken liver and adipose tissue, Genet Sel Evol, vol.49, p.6, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01431420

J. Lagarde, B. Uszczynska-ratajczak, J. Santoyo-lopez, J. M. Gonzalez, E. Tapanari et al., Extension of human lncRNA transcripts by RACE coupled with long-read high-throughput sequencing

, Nat Commun, vol.7, p.12339, 2016.

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 Rep, vol.11, pp.1110-1132, 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.242-247, 2016.

D. Villar, C. Berthelot, S. Aldridge, T. F. Rayner, M. Lukk et al., Enhancer evolution across 20 mammalian species, Cell, vol.160, pp.554-66, 2015.

J. F. Degner, A. A. Pai, R. Pique-regi, J. B. Veyrieras, D. J. Gaffney et al., DNase I sensitivity QTLs are a major determinant of human expression variation, Nature, vol.482, p.390, 2012.

K. Qu, L. C. Zaba, P. G. Giresi, R. Li, M. Longmire et al., Individuality and variation of personal regulomes in primary human T cells, Cell Syst, vol.1, pp.51-61, 2015.

J. P. Scott-browne, I. F. López-moyado, S. Trifari, V. Wong, L. Chavez et al., Dynamic changes in chromatin accessibility occur in CD8+ T cells responding to viral infection, Immunity, vol.45, pp.1327-1367, 2016.

R. E. Thurman, E. Rynes, R. Humbert, J. Vierstra, M. T. Maurano et al., The accessible chromatin landscape of the human genome, Nature, vol.489, p.75, 2012.

S. Rao, M. Huntley, N. Durand, E. Stamenova, I. Bochkov et al., A 3D map of the human genome at kilobase resolution reveals principles of chromatin looping, Cell, vol.159, pp.1665-80, 2014.

N. Servant, B. R. Lajoie, E. P. Nora, L. Giorgetti, C. J. Chen et al., HiTC: exploration of high-throughput 'C' experiments, Bioinformatics, vol.28, pp.2843-2847, 2012.

N. C. Durand, M. S. Shamim, I. Machol, S. S. Rao, M. H. Huntley et al., Juicer provides a one-click system for analyzing loop-resolution Hi-C experiments, Cell Syst, vol.3, pp.95-103, 2016.

J. R. Dixon, S. Selvaraj, F. Yue, A. Kim, Y. Li et al., Topological domains in mammalian genomes identified by analysis of chromatin interactions, Nature, vol.485, pp.376-80, 2012.

Y. Gong, C. Lazaris, T. Sakellaropoulos, A. Lozano, P. Kambadur et al., Stratification of TAD boundaries reveals preferential insulation of super-enhancers by strong boundaries, Nat Commun, vol.9, p.542, 2018.

E. Crane, Q. Bian, R. P. Mccord, B. R. Lajoie, B. S. Wheeler et al., Condensin-driven remodelling of X chromosome topology during dosage compensation, Nature, vol.523, p.240, 2015.

S. Sofueva, E. Yaffe, W. C. Chan, D. Georgopoulou, M. V. Rudan et al., Cohesin-mediated interactions organize chromosomal domain architecture, EMBO J, vol.32, pp.3119-3148, 2013.

M. V. Rudan, C. Barrington, S. Henderson, C. Ernst, D. T. Odom et al., Comparative Hi-C reveals that CTCF underlies evolution of chromosomal domain architecture, Cell Rep, vol.10, pp.1297-309, 2015.

D. Filippova, R. Patro, G. Duggal, and C. Kingsford, Identification of alternative topological domains in chromatin, Algoritm Mol Bio, vol.9, p.14, 2014.

J. R. Dixon, D. U. Gorkin, and B. Ren, Chromatin domains: the unit of chromosome organization, Mol Cell, vol.62, pp.668-80, 2016.

D. G. Lupiáñez, K. Kraft, V. Heinrich, P. Krawitz, F. Brancati et al., Disruptions of topological chromatin domains cause pathogenic rewiring of gene-enhancer interactions, Cell, vol.161, pp.1012-1037, 2015.

Y. Yang, Y. Zhang, B. Ren, J. R. Dixon, and J. Ma, Comparing 3D genome organization in multiple species using Phylo-HMRF, Cell Syst, 2019.

V. Fishman, N. Battulin, M. Nuriddinov, A. Maslova, A. Zlotina et al., 3D organization of chicken genome demonstrates evolutionary conservation of topologically associated domains and highlights unique architecture of erythrocytes' chromatin, Nucleic Acids Res, vol.47, pp.648-65, 2018.

N. Harmston, E. Ing-simmons, G. Tan, M. Perry, M. Merkenschlager et al., Topologically associating domains are ancient features that coincide with Metazoan clusters of extreme noncoding conservation, Nat Commun, vol.8, p.441, 2017.

J. R. Dixon, I. Jung, S. Selvaraj, Y. Shen, J. E. Antosiewicz-bourget et al., Chromatin architecture reorganization during stem cell differentiation, Nature, vol.518, p.331, 2015.

M. D. Doynova, J. F. Markworth, D. Cameron-smith, M. H. Vickers, O. Sullivan et al., Linkages between changes in the 3D organization of the genome and transcription during myotube differentiation in vitro, Skelet Muscle, vol.7, p.5, 2017.

A. D. Schmitt, M. Hu, I. Jung, Z. Xu, Y. Qiu et al., A compendium of chromatin contact maps reveals spatially active regions in the human genome, Cell Rep, vol.17, pp.2042-59, 2016.

S. Djebali, V. Wucher, S. Foissac, C. Hitte, E. Corre et al., Bioinformatics pipeline for transcriptome sequencing analysis, U Ørom, Enhancer RNAs, vol.1468, pp.201-219, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01439357

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, Nat Biotechnol, vol.28, pp.511-516, 2010.

A. Roberts, H. Pimentel, C. Trapnell, and L. Pachter, Identification of novel transcripts in annotated genomes using RNA-Seq, Bioinformatics, vol.27, pp.2325-2334, 2011.

F. Rohart, B. Gautier, and A. Singh, Lê Cao KA. mixOmics: an R package for omics feature selection and multiple data integration, PLoS Comput Biol, vol.13, p.5752, 2017.

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

M. D. Robinson and A. Oshlack, A scaling normalization method for differential expression analysis of RNA-seq data, Genome Biol, vol.11, p.25, 2010.

D. J. Mccarthy, Y. Chen, and G. K. Smyth, Differential expression analysis of multifactor RNA-Seq experiments with respect to biological variation, Nucleic Acids Res, vol.40, pp.4288-97, 2012.

Y. Benjamini and Y. Hochberg, Controlling the false discovery rate: a practical and powerful approach to multiple testing, J R Stat Soc Ser B (Methodol), vol.57, pp.289-300

S. Falcon and R. Gentleman, Using GOstats to test gene lists for GO term association, Bioinformatics, vol.23, pp.257-265, 2006.

L. Breiman, Random forests, Mach Learn, vol.45, pp.5-32, 2001.

B. Langmead and S. L. Salzberg, Fast gapped-read alignment with Bowtie 2, Nat Methods, vol.9, pp.357-366, 2012.

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.

J. Feng, T. Liu, B. Qin, Y. Zhang, and X. S. Liu, Identifying ChIP-seq enrichment using MACS, Nat Protocol, vol.7, pp.1728-1768, 2012.

A. R. Quinlan, BEDTools: the Swiss-army tool for genome feature analysis, Curr Protocol Bioinform, vol.47, pp.11-13, 2014.

K. V. Ballman, D. E. Grill, A. L. Oberg, and T. M. Therneau, Faster cyclic loess: normalizing RNA arrays via linear models, Bioinformatics, vol.20, pp.2778-86, 2004.

A. T. Lun and G. K. Smyth, csaw: a Bioconductor package for differential binding analysis of ChIP-seq data using sliding windows, Nucleic Acids Res, vol.44, p.45, 2015.

C. E. Grant, T. L. Bailey, and W. S. Noble, FIMO: scanning for occurrences of a given motif, Bioinformatics, vol.27, pp.1017-1025, 2011.

A. Mathelier, O. Fornes, D. J. Arenillas, C. Cy, G. Denay et al., JASPAR 2016: a major expansion and update of the open-access database of transcription factor binding profiles, Nucleic Acids Res, vol.44, pp.110-115, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01281181

N. Servant, N. Varoquaux, B. R. Lajoie, E. Viara, C. J. Chen et al., HiC-Pro: an optimized and flexible pipeline for Hi-C data processing
URL : https://hal.archives-ouvertes.fr/hal-01246671

, Genome Biol, vol.16, p.259, 2015.

M. Imakaev, G. Fudenberg, R. P. Mccord, N. Naumova, A. Goloborodko et al., Iterative correction of Hi-C data reveals hallmarks of chromosome organization, Nat Methods, vol.9, pp.999-1003, 2012.

N. C. Durand, J. T. Robinson, M. S. Shamim, I. Machol, J. P. Mesirov et al., Juicebox provides a visualization system for Hi-C contact maps with unlimited zoom, Cell Syst, vol.3, pp.99-101, 2016.

T. Barrett, K. Clark, R. Gevorgyan, V. Gorelenkov, E. Gribov et al., BioProject and BioSample databases at NCBI: facilitating capture and organization of metadata, Nucleic Acids Res, vol.40, pp.57-63, 2012.

. Embl-ebi and . Biosamples, , 2019.

, Publisher's Note

, Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations