M. J. Evans and M. H. Kaufman, Establishment in culture of pluripotential cells from mouse embryos, Nature, vol.292, pp.154-156, 1981.

J. A. Thomson, J. Itskovitz-eldor, S. S. Shapiro, M. A. Waknitz, and J. J. Swiergiel, Embryonic stem cell lines derived from human blastocysts, Science, vol.282, pp.1145-1147, 1998.

K. Takahashi and S. Yamanaka, Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors, Cell, vol.126, pp.663-676, 2006.

S. J. Dunn, G. Martello, B. Yordanov, S. Emmott, and A. G. Smith, Defining an essential transcription factor program for naïve pluripotency, Science, vol.344, pp.1156-1160, 2014.

H. W. Chen, I. Aksoy, F. Gonnot, P. Osteil, M. Aubry et al., Reinforcement of STAT3 activity reprogrammes human embryonic stem cells to naive-like pluripotency, Nature Communications, vol.6, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01997120

J. Nichols and A. Smith, Naive and primed pluripotent states, Cell Stem Cell, vol.5, issue.6, pp.487-92, 2009.

F. Gandolfi, G. Pennarossa, S. Maffei, and T. Brevini, Why is it so difficult to derive pluripotent stem cells in domestic ungulates?, Reprod Domest Anim, vol.47, pp.11-18, 2012.

J. C. Litten-brown, A. M. Corson, and L. Clarke, Porcine models for the metabolic syndrome, digestive and bone disorders: a general overview, Animal, vol.4, pp.899-920, 2010.

H. Matsunari, H. Nagashima, M. Watanabe, K. Umeyama, and K. Nakano, Blastocyst complementation generates exogenic pancreas in vivo in apancreatic cloned pigs, Proc Natl Acad Sci U S A, vol.110, pp.4557-4562, 2013.

L. F. Malaver-ortega, H. Sumer, J. Liu, and P. J. Verma, The state of the art for pluripotent stem cells derivation in domestic ungulates, Theriogenology, vol.78, pp.1749-1762, 2012.

A. A. Pollen, T. J. Nowakowski, J. Shuga, X. Wang, A. A. Leyrat et al., Low-coverage single-cell mRNA sequencing reveals cellular heterogeneity and activated signaling pathways in developing cerebral cortex, Nat Biotechnol, vol.32, issue.10, pp.1053-1061, 2014.

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.25, p.57, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01194102

P. D. Hsu, E. S. Lander, and F. Zhang, Development and applications of CRISPR-Cas9 for genome engineering, Cell, vol.157, issue.6, pp.1262-78, 2014.

A. Congras, H. Barasc, K. Canale-tabet, F. Plisson-petit, C. Delcros et al., Non integrative strategy decreases chromosome instability and improve reprogramming of porcine induced pluripotent stem cells

O. Gafni, L. Weinberger, A. A. Mansour, Y. S. Manor, E. Chomsky et al., Derivation of novel human ground state naive pluripotent stem cells, Nature, vol.504, pp.282-286, 2013.

T. W. Theunissen, B. E. Powell, H. Wang, M. Mitalipova, D. A. Faddah et al., Systematic identification of culture conditions for induction and maintenance of naive human pluripotency, Cell Stem Cell, vol.15, pp.471-487, 2014.

E. Apostolou, Genome-wide Chromatin Interactions of the Nanog Locus in Pluripotency, Differentiation, and Reprogramming, Cell stem cell, vol.12, pp.699-712, 2013.

F. Ay, Three-dimensional modeling of the P. falciparum genome during the erythrocytic cycle reveals a strong connection between genome architecture and gene expression, Genome research, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01101873

C. Berthelot, The rainbow trout genome provides novel insights into evolution after whole-genome duplication in vertebrates, Nature Communications, vol.5, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01193812

E. Birney, The making of ENCODE: lessons for big-data projects, Nature, vol.489, pp.49-51, 2012.

J. N. Burton, Chromosome-scale scaffolding of de novo genome assemblies based on chromatin interactions, Nature biotechnology, 2013.

G. Bussotti, BlastR-fast and accurate database searches for non-coding RNAs, Nucleic acids research, vol.39, pp.6886-6895, 2011.
URL : https://hal.archives-ouvertes.fr/hal-02644812

S. Campo, Identification of a novel microRNA (miRNA) from rice that targets an alternatively spliced transcript of the Nramp6 (Natural resistance-associated macrophage protein 6) gene involved in pathogen resistance, New Phytologist, vol.199, pp.212-227, 2013.
URL : https://hal.archives-ouvertes.fr/hal-02650361

M. Cros, RNAspace. org: An integrated environment for the prediction, annotation, and analysis of ncRNA, RNA, vol.17, pp.1947-1956, 2011.
URL : https://hal.archives-ouvertes.fr/hal-02819286

T. Derrien, The GENCODE v7 catalog of human long noncoding RNAs: analysis of their gene structure, evolution, and expression, Genome research, vol.22, pp.1775-1789, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01205054

J. R. Dixon, Topological domains in mammalian genomes identified by analysis of chromatin interactions, Nature, vol.485, pp.376-380, 2012.

Y. Dong, Sequencing and automated whole-genome optical mapping of the genome of a domestic goat (Capra hircus), Nature biotechnology, vol.31, pp.135-141, 2013.
URL : https://hal.archives-ouvertes.fr/hal-02651460

C. G. Elsik, L. Ross, K. C. Tellam, and . Worley, The genome sequence of taurine cattle: a window to ruminant biology and evolution, ENCODE Project Consortium, vol.324, pp.57-74, 2009.
URL : https://hal.archives-ouvertes.fr/hal-02667039

C. W. Ernst and J. P. Steibel, Molecular advances in QTL discovery and application in pig breeding, Trends in Genetics, vol.29, pp.215-224, 2013.

M. Y. Gerner, Histo-cytometry: a method for highly multiplex quantitative tissue imaging analysis applied to dendritic cell subset microanatomy in lymph nodes, Immunity, vol.37, pp.364-376, 2012.

. Groenen and A. M. Martien, Analyses of pig genomes provide insight into porcine demography and evolution, Nature, vol.491, pp.393-398, 2012.
URL : https://hal.archives-ouvertes.fr/cea-00880676

L. W. Hillier, Sequence and comparative analysis of the chicken genome provide unique perspectives on vertebrate evolution, Nature, vol.432, pp.695-716, 2004.
URL : https://hal.archives-ouvertes.fr/hal-02675974

F. Jin, A high-resolution map of the three-dimensional chromatin interactome in human cells, Nature, vol.503, pp.290-294, 2013.

N. Kaplan and J. Dekker, High-throughput genome scaffolding from in vivo DNA interaction frequency, Nature biotechnology, 2013.

D. Karolchik, The UCSC Genome Browser database: 2014 update, Nucleic Acids Res, vol.42, 2013.

S. G. Landt, ChIP-seq guidelines and practices of the ENCODE and modENCODE consortia, Genome research, vol.22, pp.1813-1831, 2012.

. Lieberman-aiden and . Erez, Comprehensive mapping of long-range interactions reveals folding principles of the human genome, science, vol.326, pp.289-293, 2009.

T. Meuwissen and . He, Fine mapping of a quantitative trait locus for twinning rate using combined linkage and linkage disequilibrium mapping, Genetics, vol.161, pp.373-379, 2002.

G. Morota, Kernel-based variance component estimation and whole-genome prediction of precorrected phenotypes and progeny tests for dairy cow health traits, Frontiers in genetics, vol.5, 2014.

C. Seliger, A rapid high-precision flow cytometry based technique for total white blood cell counting in chickens, Veterinary immunology and immunopathology, vol.145, pp.86-99, 2012.

T. Sexton, Three-Dimensional Folding and Functional Organization Principles of the Drosophila Genome, Cell, vol.148, pp.458-472, 2012.

C. Sheridan, . Berkum, and L. Nynke, Hi-C: a method to study the three-dimensional architecture of genomes, Journal of visualized experiments: JoVE, vol.32, p.39, 2010.

-. Hi, ;. Pro, and . Zhang, En effet les matrices d'interaction associent les lectures d'un segment, 2012.

H. Acloque, M. S. Adams, K. Fishwick, M. Bronner-fraser, and M. A. Nieto, Epithelial-mesenchymal transitions: the importance of changing cell state in development and disease, J Clin Invest, vol.119, pp.1438-1449, 2009.

H. Acloque, D. G. Wilkinson, and M. A. Nieto, In situ hybridization analysis of chick embryos in whole-mount and tissue sections, Methods Cell Biol, vol.87, pp.169-185, 2008.

R. Alberio, N. Croxall, and C. Allegrucci, Pig epiblast stem cells depend on activin/nodal signaling for pluripotency and selfrenewal, Stem Cells Dev, vol.19, issue.10, pp.1627-1663, 2010.

M. Al-hajj, M. S. Wicha, A. Benito-hernandez, S. J. Morrison, C. et al., Prospective identification of tumorigenic breast cancer cells, Proc Natl Acad Sci U S A, vol.100, pp.3983-3988, 2003.

G. Ambartsumyan and A. T. Clark, Aneuploidy and early human embryo development, Hum Mol Genet, vol.17, issue.R1, p.10, 2008.

, American Zoologist, vol.2, p.432

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

J. B. Arnes, J. S. Brunet, I. Stefansson, L. R. Bégin, N. Wong et al., Placental cadherin and the basal epithelial phenotype of BRCA1-related breast cancer, Clin Cancer Res, vol.11, pp.4003-4011, 2005.

K. I. Aston, V. Punj, L. Liu, and D. T. Carrell, Genome-wide sperm deoxyribonucleic acid methylation is altered in some men with abnormal chromatin packaging or poor in vitro fertilization embryogenesis, Fertil Steril, vol.97, pp.285-292, 2012.

Q. Bai, J. M. Ramirez, F. Becker, V. Pantesco, T. Lavabre-bertrand et al., Temporal analysis of genome alterations induced by single-cell passaging in human embryonic stem cells, Stem Cells, vol.24, pp.653-662, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01840168

S. Bao, F. Tang, X. Li, K. Hayashi, A. Gillich et al., Epigenetic reversion of post-implantation epiblast to pluripotent embryonic stem cells, Nature, vol.461, pp.1292-1295, 2009.

H. Barasc, N. Mary, R. Letron, A. Calgaro, A. M. Dudez et al., Y-autosome translocation interferes with meiotic sex inactivation and expression of autosomal genes: a case study in the pig, Sex Dev, vol.6, pp.143-150, 2012.
URL : https://hal.archives-ouvertes.fr/hal-02651727

A. Barrallo-gimeno and M. A. Nieto, The Snail genes as inducers of cell movement and survival: implications in development and cancer, Development, vol.132, pp.3151-3161, 2005.

B. Beck, G. Lapouge, S. Rorive, B. Drogat, K. Desaedelaere et al., Different levels of Twist1 regulate skin tumor initiation, stemness, and progression, Cell Stem Cell, vol.16, pp.67-79, 2015.

H. Bildsoe, D. A. Loebel, V. J. Jones, A. C. Hor, A. W. Braithwaite et al., The mesenchymal architecture of the cranial mesoderm of mouse embryos is disrupted by the loss of Twist1 function, Dev Biol, vol.374, pp.295-307, 2013.

I. Bittmann, D. Mihica, R. Plesker, and J. Denner, Expression of porcine endogenous retroviruses (PERV) in different organs of a pig, Virology, vol.433, pp.329-336, 2012.

M. J. Blanco, G. Moreno-bueno, D. Sarrio, A. Locascio, A. Cano et al., Correlation of Snail expression with histological grade and lymph node status in breast carcinomas, Oncogene, vol.21, pp.3241-3246, 2002.

L. A. Blomberg and B. P. Telugu, Twenty years of embryonic stem cell research in farm animals, Reprod Domest Anim, vol.47, pp.80-85, 2012.

C. C. Boissonnas, H. E. Abdalaoui, V. Haelewyn, P. Fauque, J. M. Dupont et al., Specific epigenetic alterations of IGF2-H19 locus in spermatozoa from infertile men, Eur J Hum Genet, vol.18, pp.73-80, 2010.
URL : https://hal.archives-ouvertes.fr/hal-02668707

C. C. Boissonnas, P. Jouannet, J. , and H. , Epigenetic disorders and male subfertility, Fertil Steril, vol.99, pp.624-631, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01001417

T. Brabletz, EMT and MET in metastasis: where are the cancer stem cells, Cancer Cell, vol.22, pp.699-701, 2012.

A. Bradley, M. Evans, M. H. Kaufman, and E. Robertson, Formation of germ-line chimaeras from embryo-derived teratocarcinoma cell lines, Nature, vol.309, pp.255-261, 1984.

T. A. Brevini, S. Antonini, F. Cillo, M. Crestan, and F. Gandolfi, Porcine embryonic stem cells: Facts, challenges and hopes, Theriogenology, vol.68, pp.206-219, 2007.

T. A. Brevini, S. Antonini, G. Pennarossa, and F. Gandolfi, Recent progress in embryonic stem cell research and its application in domestic species, Reprod Domest Anim, vol.43, pp.193-202, 2008.

T. A. Brevini, V. Tosetti, M. Crestan, S. Antonini, and F. Gandolfi, Derivation and characterization of pluripotent cell lines from pig embryos of different origins, Theriogenology, vol.67, issue.1, pp.54-63, 2007.

R. L. Brinster, Studies on the development of mouse embryos in vitro. IV. Interaction of energy sources, J Reprod Fertil, vol.10, pp.227-267, 1965.

I. G. Brons, L. E. Smithers, M. W. Trotter, P. Rugg-gunn, B. Sun et al., Derivation of pluripotent epiblast stem cells from mammalian embryos, Nature, vol.448, issue.7150, pp.191-196, 2007.

I. G. Brons, L. E. Smithers, M. W. Trotter, P. Rugg-gunn, B. Sun et al., Derivation of pluripotent epiblast stem cells from mammalian embryos, Nature, vol.448, pp.191-195, 2007.

M. Buehr, S. Meek, K. Blair, J. Yang, J. Ure et al., Capture of authentic embryonic stem cells from rat blastocysts, Cell, vol.135, issue.7, pp.1287-98, 2008.

S. Cao, J. Han, J. Wu, Q. Li, S. Liu et al., Specific gene-regulation networks during the pre-implantation development of the pig embryo as revealed by deep sequencing, BMC Genomics, vol.15, issue.1, p.4, 2014.

R. S. Carsience, M. E. Clark, A. M. Verrinder-gibbins, and R. J. Etches, Germline chimeric chickens from dispersed donor blastodermal cells and compromised recipient embryos, Development, vol.117, pp.669-75, 1993.

E. A. Carver, R. Jiang, Y. Lan, K. F. Oram, and T. Gridley, The mouse snail gene encodes a key regulator of the epithelialmesenchymal transition, Mol Cell Biol, vol.21, pp.8184-8188, 2001.

I. Castanon and M. K. Baylies, A Twist in fate: evolutionary comparison of Twist structure and function, Gene, vol.287, pp.11-22, 2002.

I. Chambers, D. Colby, M. Robertson, J. Nichols, S. Lee et al., Functional expression cloning of nanog, a pluripotency sustaining factor in embryonic stem cells, Cell, vol.113, pp.643-55, 2003.

A. T. Chang, Y. Liu, K. Ayyanathan, C. Benner, Y. Jiang et al., An evolutionarily conserved DNA architecture determines target specificity of the TWIST family bHLH transcription factors, Genes Dev, vol.29, pp.603-616, 2015.

S. C. Chapman, J. Collignon, G. C. Schoenwolf, and A. Lumsden, Improved method for chick whole-embryo culture using a filter paper carrier, Dev Dyn, vol.220, pp.284-289, 2001.

L. Chavez, J. Jozefczuk, C. Grimm, J. Dietrich, B. Timmermann et al., Computational analysis of genome-wide DNA methylation during the differentiation of human embryonic stem cells along the endodermal lineage, Genome, vol.20, pp.1441-1450, 2010.

L. R. Chen, Y. L. Shiue, L. Bertolini, J. F. Medrano, R. H. Bondurant et al., Establishment of pluripotent cell lines from porcine preimplantation embryos, Theriogenology, vol.52, issue.2, pp.195-212, 1999.

Y. Chen, L. Shi, L. Zhang, R. Li, J. Liang et al., The molecular mechanism governing the oncogenic potential of SOX2 in breast cancer, J Biol Chem, vol.283, pp.17969-17978, 2008.

Z. F. Chen and R. R. Behringer, twist is required in head mesenchyme for cranial neural tube morphogenesis, Genes Dev, vol.9, pp.686-699, 1995.

J. Comijn, G. Berx, P. Vermassen, K. Verschueren, L. Van-grunsven et al., The two-handed E box binding zinc finger protein SIP1 downregulates E-cadherin and induces invasion, Mol Cell, vol.7, pp.1267-1278, 2001.

E. P. Consortium, The ENCODE (ENCyclopedia Of DNA Elements) Project. Science306, pp.636-640, 2004.

I. C. Consortium, Sequence and comparative analysis of the chicken genome provide unique perspectives on vertebrate evolution, Nature, vol.432, pp.695-716, 2004.

D. Coronado, M. Godet, P. Y. Bourillot, Y. Tapponnier, A. Bernat et al., A short G1 phase is an intrinsic determinant of naïve embryonic stem cell pluripotency, Stem Cell Res, vol.10, pp.118-131, 2013.

T. Cremer and C. Cremer, Chromosome territories, nuclear architecture and gene regulation in mammalian cells, Nat Rev Genet, vol.2, pp.292-301, 2001.

N. Dave, S. Guaita-esteruelas, S. Gutarra, À. Frias, M. Beltran et al., Functional cooperation between Snail1 and twist in the regulation of ZEB1 expression during epithelial to mesenchymal transition, J Biol Chem, vol.286, pp.12024-12032, 2011.

J. D. Delhanty, D. K. Griffin, A. H. Handyside, J. Harper, G. H. Atkinson et al., Detection of aneuploidy and chromosomal mosaicism in human embryos during preimplantation sex determination by fluorescent in situ hybridisation, (FISH), Hum Mol Genet, vol.2, issue.8, pp.1183-1188, 1993.

H. Driesch, Entwicklungsmechanische Studien I. Der Wert der beiden ersten Furchungszellen in der Echinodermenentwicklung, Zeitschrift für wissenschaftliche Zoologie, vol.53, pp.160-183, 1891.

A. Ducos, H. M. Berland, N. Bonnet, A. Calgaro, S. Billoux et al., Chromosomal control of pig populations in France: 2002-2006 survey, Genet Sel Evol, vol.39, pp.583-597, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00894612

A. Ducos, H. M. Berland, A. Pinton, E. Guillemot, A. Seguela et al., Nine new cases of reciprocal translocation in the domestic pig (Sus scrofa domestica L.), J Hered, vol.89, pp.136-142, 1998.
URL : https://hal.archives-ouvertes.fr/hal-02699134

A. Eger, K. Aigner, S. Sonderegger, B. Dampier, S. Oehler et al., DeltaEF1 is a transcriptional repressor of E-cadherin and regulates epithelial plasticity in breast cancer cells, Oncogene, vol.24, pp.2375-2385, 2005.

M. A. Esteban, J. Xu, J. Yang, M. Peng, D. Qin et al., Generation of induced pluripotent stem cell lines from Tibetan miniature pig, J Biol Chem, vol.284, issue.26, pp.17634-17674, 2009.

M. J. Evans, The isolation and properties of a clonal tissue culture strain of pluripotent mouse teratoma cells, J Embryol Exp Morphol, vol.28, pp.163-76, 1972.

M. J. Evans and M. H. Kaufman, Establishment in culture of pluripotential cells from mouse embryos, Nature, vol.292, pp.154-160, 1981.

H. Eyal-giladi and S. Kochav, From cleavage to primitive streak formation: a complementary normal table and a new look at the first stages of the development of the chick. I. General morphology, Dev Biol, vol.49, pp.321-358, 1976.

S. Fanucchi, Y. Shibayama, S. Burd, M. S. Weinberg, and M. M. Mhlanga, Chromosomal contact permits transcription between coregulated genes, Cell, vol.155, pp.606-620, 2013.

J. Feng, T. Liu, and Y. Zhang, Using MACS to identify peaks from ChIP-Seq data, Curr Protoc Bioinformatics Chapter, 2011.

K. A. Ferguson, V. Chow, and S. Ma, Silencing of unpaired meiotic chromosomes and altered recombination patterns in an azoospermic carrier of a t(8;13) reciprocal translocation, Hum Reprod, vol.23, pp.988-995, 2008.

W. D. Foulkes, BRCA1 functions as a breast stem cell regulator, J Med Genet, vol.41, pp.1-5, 2004.

S. H. Fujishiro, K. Nakano, Y. Mizukami, T. Azami, Y. Arai et al., Generation of Naive-Like Porcine-Induced Pluripotent Stem Cells Capable of Contributing to Embryonic and Fetal Development, Stem Cells Dev, 2012.

B. Galliot, C. De-vargas, and D. Miller, Evolution of homeobox genes: Q50 Paired-like genes founded the Paired class, Dev Genes Evol, vol.209, pp.186-197, 1999.

R. L. Gardner, Mouse chimeras obtained by the injection of cells into the blastocyst, Nature, vol.220, pp.596-603, 1968.

C. M. Ghajar, H. Peinado, H. Mori, I. R. Matei, K. J. Evason et al., The perivascular niche regulates breast tumour dormancy, Nat Cell Biol, vol.15, pp.807-817, 2013.

C. A. Gifford, M. J. Ziller, H. Gu, C. Trapnell, J. Donaghey et al., Transcriptional and epigenetic dynamics during specification of human embryonic stem cells, Cell, vol.153, pp.1149-1163, 2013.

J. A. Gingold, M. Fidalgo, D. Guallar, Z. Lau, Z. Sun et al., A genome-wide RNAi screen identifies opposing functions of Snai1 and Snai2 on the Nanog dependency in reprogramming, Mol Cell, vol.56, pp.140-152, 2014.

J. Göke, X. Lu, Y. S. Chan, H. H. Ng, L. H. Ly et al., Dynamic transcription of distinct classes of endogenous retroviral elements marks specific populations of early human embryonic cells, Cell Stem Cell, vol.16, pp.135-141, 2015.

M. A. Groenen, A. L. Archibald, H. Uenishi, C. K. Tuggle, Y. Takeuchi et al., Analyses of pig genomes provide insight into porcine demography and evolution, Nature, vol.491, pp.393-398, 2012.
URL : https://hal.archives-ouvertes.fr/cea-00880676

E. J. Grow, R. A. Flynn, S. L. Chavez, N. L. Bayless, M. Wossidlo et al., Intrinsic retroviral reactivation in human preimplantation embryos and pluripotent cells, Nature, vol.522, pp.221-225, 2015.

W. Guo, Z. Keckesova, J. L. Donaher, T. Shibue, V. Tischler et al., Slug and Sox9 cooperatively determine the mammary stem cell state, Cell, vol.148, pp.1015-1028, 2012.

V. J. Hall and P. Hyttel, Breaking down pluripotency in the porcine embryo reveals both a premature and reticent stem cell state in the inner cell mass and unique expression profiles of the naive and primed stem cell States, Stem Cells Dev, vol.23, issue.17, pp.2030-2075, 2014.

V. J. Hall, J. Christensen, Y. Gao, M. H. Schmidt, and P. Hyttel, Porcine pluripotency cell signaling develops from the inner cell mass to the epiblast during early development, Dev Dyn, vol.238, issue.8, pp.2014-2038, 2009.

V. J. Hall, J. V. Jacobsen, M. A. Rasmussen, and P. Hyttel, Ultrastructural and molecular distinctions between the porcine inner cell mass and epiblast reveal unique pluripotent cell states, Dev Dyn, vol.239, issue.11, pp.2911-2931, 2010.

V. Hamburger and H. L. Hamilton, A series of normal stages in the development of the chick embryo, Dev Dyn, vol.195, pp.231-72, 1951.

S. S. Hammoud, J. Purwar, C. Pflueger, B. R. Cairns, and D. T. Carrell, Alterations in sperm DNA methylation patterns at imprinted loci in two classes of infertility, Fertil, vol.94, pp.1728-1733, 2010.

J. H. Hanna, K. Saha, J. , and R. , Pluripotency and cellular reprogramming: facts, hypotheses, unresolved issues, Cell, vol.143, pp.508-525, 2010.

E. D. Hay, Organization and fine structure of epithelium and mesenchyme in the developing chick embryo, pp.31-55, 1968.

S. Houshdaran, V. K. Cortessis, K. Siegmund, A. Yang, P. W. Laird et al., Widespread epigenetic abnormalities suggest a broad DNA methylation erasure defect in abnormal human sperm, PLoS One, vol.2, 1289.

Y. Huang, Y. Li, D. W. Burt, H. Chen, Y. Zhang et al., The duck genome and transcriptome provide insight into an avian influenza virus reservoir species, Nat Genet, vol.45, pp.776-783, 2013.
URL : https://hal.archives-ouvertes.fr/hal-02652041

D. Huss, B. Benazeraf, A. Wallingford, M. Filla, J. Yang et al., A transgenic quail model that enables dynamic imaging of amniote embryogenesis, Development, vol.142, pp.2850-2859, 2015.

W. W. Hwang-verslues, P. H. Chang, Y. M. Jeng, W. H. Kuo, P. H. Chiang et al., Loss of corepressor PER2 under hypoxia up-regulates OCT1-mediated EMT gene expression and enhances tumor malignancy, Proc Natl Acad Sci U S A, vol.110, pp.12331-12336, 2013.

D. Ioannou and D. K. Griffin, Male fertility, chromosome abnormalities, and nuclear organization, Cytogenet Genome Res, vol.133, pp.269-279, 2011.

B. W. Kahan and B. Ephrussi, Developmental potentialities of clonal in vitro cultures of mouse testicular teratoma, J Natl Cancer Inst, vol.44, pp.1015-1051, 1970.

J. Kim, W. Yu, K. Kovalski, and L. Ossowski, Requirement for specific proteases in cancer cell intravasation as revealed by a novel semiquantitative PCR-based assay, Cell, vol.94, pp.353-362, 1998.

D. J. Kleinjan and V. Van-heyningen, Position effect in human genetic disease, Hum Mol Genet, vol.7, pp.1611-1618, 1998.

H. Kobayashi, A. Sato, E. Otsu, H. Hiura, C. Tomatsu et al., Aberrant DNA methylation of imprinted loci in sperm from oligospermic patients, Hum Mol Genet, vol.16, pp.2542-2551, 2007.

W. A. Kues, D. Herrmann, B. Barg-kues, S. Haridoss, M. Nowak-imialek et al., Derivation and characterization of sleeping beauty transposon-mediated porcine induced pluripotent stem cells, Stem Cells Dev, vol.22, issue.1, pp.124-159, 2013.

D. J. Kwon, H. Jeon, K. B. Oh, S. A. Ock, G. S. Im et al., Generation of leukemia inhibitory factor-dependent induced pluripotent stem cells from the massachusetts general hospital miniature pig, Biomed Res Int, p.140639, 2013.

R. Lander, T. Nasr, S. D. Ochoa, K. Nordin, M. S. Prasad et al., Interactions between Twist and other core epithelial-mesenchymal transition factors are controlled by GSK3-mediated phosphorylation, Nat Commun, vol.4, p.1542, 2013.

R. Li, J. Liang, S. Ni, T. Zhou, X. Qing et al., A mesenchymal-to-epithelial transition initiates and is required for the nuclear reprogramming of mouse fibroblasts, Cell Stem Cell, vol.7, pp.51-63, 2010.

E. Lieberman-aiden, N. L. Van-berkum, L. Williams, M. Imakaev, T. Ragoczy et al., Comprehensive mapping of long-range interactions reveals folding principles of the human genome, Science, vol.326, pp.289-293, 2009.

J. C. Litten-brown, A. M. Corson, and L. Clarke, Porcine models for the metabolic syndrome, digestive and bone disorders: a general overview, Animal, vol.4, issue.6, pp.899-920, 2010.

S. Liu, C. Ginestier, E. Charafe-jauffret, H. Foco, C. G. Kleer et al., BRCA1 regulates human mammary stem/progenitor cell fate, Proc Natl Acad Sci U S A, vol.105, pp.1680-1685, 2008.
URL : https://hal.archives-ouvertes.fr/hal-01431959

C. C. Lu, J. Brennan, and E. J. Robertson, From fertilization to gastrulation: axis formation in the mouse embryo, Curr Opin Genet Dev, vol.11, pp.384-92, 2001.

M. F. Lu, H. T. Cheng, M. J. Kern, S. S. Potter, B. Tran et al., prx-1 functions cooperatively with another paired-related homeobox gene, prx-2, to maintain cell fates within the craniofacial mesenchyme, Development, vol.126, pp.495-504, 1999.

J. K. Lunney, Advances in swine biomedical model genomics, Int J Biol Sci, vol.3, issue.3, pp.179-84, 2007.

T. S. Macfarlan, W. D. Gifford, S. Driscoll, K. Lettieri, H. M. Rowe et al., Embryonic stem cell potency fluctuates with endogenous retrovirus activity, Nature, vol.487, pp.57-63, 2012.

S. A. Mani, W. Guo, M. J. Liao, E. N. Eaton, A. Ayyanan et al., The epithelial-mesenchymal transition generates cells with properties of stem cells, Cell, vol.133, pp.704-715, 2008.

A. Mantzouratou and J. D. Delhanty, Aneuploidy in the human cleavage stage embryo, Cytogenet Genome Res, vol.133, issue.2-4, pp.141-149, 2011.

C. J. Marques, P. Costa, B. Vaz, F. Carvalho, S. Fernandes et al., Abnormal methylation of imprinted genes in human sperm is associated with oligozoospermia, Mol Hum Reprod, vol.14, pp.67-74, 2008.

G. R. Martin, Isolation of a pluripotent cell line from early mouse embryos cultured in medium conditioned by teratocarcinoma stem cells, Proc Natl Acad Sci U S A, vol.78, pp.7634-7642, 1981.

G. R. Martin and M. J. Evans, Differentiation of clonal lines of teratocarcinoma cells: formation of embryoid bodies in vitro, Proc Natl Acad Sci U S A, vol.72, pp.1441-1446, 1975.

G. Marzullo, Production of chick chimaeras, Nature, vol.225, pp.72-75, 1970.

Y. Matsui, K. Zsebo, and B. L. Hogan, Derivation of pluripotential embryonic stem cells from murine primordial germ cells in culture, Cell, vol.70, pp.841-848, 1992.

D. Medina, Of mice and women: A short history of mouse mammary cancer research with an emphasis on the paradigms inspired by the transplantation method, Cold Spring Harb Perspect Biol, vol.2, p.4523, 2010.

A. Meissner, A. Gnirke, G. W. Bell, B. Ramsahoye, E. S. Lander et al., Reduced representation bisulfite sequencing for comparative high-resolution DNA methylation analysis, Nucleic Acids Res, vol.33, pp.5868-5877, 2005.

J. Mejlvang, M. Kriajevska, C. Vandewalle, T. Chernova, A. E. Sayan et al., Direct repression of cyclin D1 by SIP1 attenuates cell cycle progression in cells undergoing an epithelial mesenchymal transition, Mol Biol Cell, vol.18, pp.4615-4624, 2007.

B. Mintz, Formation of genotypically mosaic mouse embryos, 1962.

B. Mintz and K. Illmensee, Normal genetically mosaic mice produced from malignant teratocarcinoma cells, Proc Natl Acad Sci U S A, vol.72, pp.3585-3594, 1975.

M. M. Mitalipova, R. R. Rao, D. M. Hoyer, J. A. Johnson, L. F. Meisner et al., Preserving the genetic integrity of human embryonic stem cells, Nat Biotechnol, vol.23, issue.1, pp.19-20, 2005.

K. Mitsui, Y. Tokuzawa, H. Itoh, K. Segawa, M. Murakami et al., The Homeoprotein Nanog Is Required for Maintenance of Pluripotency in Mouse Epiblast and ES Cells, Cell, vol.113, pp.631-673, 2003.

D. Monk, Germline-derived DNA methylation and early embryo epigenetic reprogramming: The selected survival of imprints, Int J Biochem Cell Biol, vol.67, pp.128-138, 2015.

D. Montjean, A. Zini, C. Ravel, S. Belloc, A. Dalleac et al., Sperm global DNA methylation level: association with semen parameters and genome integrity, Andrology, vol.3, pp.235-240, 2015.

N. Montserrat, E. G. Bahima, L. Batlle, S. Häfner, A. M. Rodrigues et al., Generation of pig iPS cells: a model for cell therapy, J Cardiovasc Transl Res, vol.4, issue.2, pp.121-151, 2011.

N. Montserrat, L. De-oñate, E. Garreta, F. González, A. Adamo et al., Generation of feeder-free pig induced pluripotent stem cells without Pou5f1, Cell Transplant, vol.21, issue.5, pp.70-73, 2012.

J. Nichols and A. Smith, Naive and primed pluripotent states, Cell Stem Cell, vol.4, pp.487-492, 2009.

J. Nichols, B. Zevnik, K. Anastassiadis, H. Niwa, D. Klewe-nebenius et al., Formation of pluripotent stem cells in the mammalian embryo depends on the POU transcription factor Oct4, Cell, vol.95, pp.379-91, 1998.

M. A. Nieto, M. G. Sargent, D. G. Wilkinson, and J. Cooke, Control of cell behavior during vertebrate development by Slug, a zinc finger gene, Science, vol.264, pp.835-839, 1994.

R. A. Norris, K. K. Scott, C. S. Moore, G. Stetten, C. R. Brown et al., Human PRRX1 and PRRX2 genes: cloning, expression, genomic localization, and exclusion as disease genes for Nager syndrome, Mamm, vol.11, pp.1000-1005, 2000.

E. Notarianni, S. Laurie, R. M. Moor, and M. J. Evans, Maintenance and differentiation in culture of pluripotential embryonic cell lines from pig blastocysts, J Reprod Fertil Suppl, vol.41, pp.51-57, 1990.

O. H. Ocaña, R. Córcoles, A. Fabra, G. Moreno-bueno, H. Acloque et al., Metastatic colonization requires the repression of the epithelial-mesenchymal transition inducer Prrx1, Cancer Cell, vol.22, pp.709-724, 2012.

O. Oestrup, V. Hall, S. G. Petkov, X. A. Wolf, S. Hyldig et al., From zygote to implantation: morphological and molecular dynamics during embryo development in the pig, Reprod Domest Anim, vol.44, pp.39-49, 2009.

M. Oliver-bonet, E. Ko, and R. H. Martin, Male infertility in reciprocal translocation carriers: the sex body affair, Cytogenet Genome Res, vol.111, pp.343-346, 2005.

D. Olmeda, A. Montes, G. Moreno-bueno, J. M. Flores, F. Portillo et al., Snai1 and Snai2 collaborate on tumor growth and metastasis properties of mouse skin carcinoma cell lines, Oncogene, vol.27, pp.4690-4701, 2008.

T. Oskarsson, S. Acharyya, X. H. Zhang, S. Vanharanta, S. F. Tavazoie et al., Breast cancer cells produce tenascin C as a metastatic niche component to colonize the lungs, Nat Med, vol.17, pp.867-874, 2011.

S. E. Pacheco, E. A. Houseman, B. C. Christensen, C. J. Marsit, K. T. Kelsey et al., Integrative DNA methylation and gene expression analyses identify DNA packaging and epigenetic regulatory genes associated with low motility sperm, PLoS One, vol.6, 2011.

B. Pain, M. E. Clark, M. Shen, H. Nakazawa, M. Sakurai et al., Long-term in vitro culture and characterisation of avian embryonic stem cells with multiple morphogenetic potentialities, Development, vol.122, pp.2339-2387, 1996.
URL : https://hal.archives-ouvertes.fr/hal-02695273

V. E. Papaioannou, M. W. Mcburney, R. L. Gardner, and M. J. Evans, Fate of teratocarcinoma cells injected into early mouse embryos, 1975.

H. Peinado, D. Olmeda, and A. Cano, Snail, Zeb and bHLH factors in tumour progression: an alliance against the epithelial phenotype?, Nat Rev Cancer, vol.7, pp.415-428, 2007.

C. Peña, J. M. García, V. García, J. Silva, G. Domínguez et al., The expression levels of the transcriptional regulators p300 and CtBP modulate the correlations between SNAIL, ZEB1, E-cadherin and vitamin D receptor in human colon carcinomas, Int J Cancer, vol.119, pp.2098-2104, 2006.

J. N. Petitte, M. E. Clark, G. Liu, A. M. Verrinder-gibbins, and R. J. Etches, Production of somatic and germline chimeras in the chicken by transfer of early blastodermal cells, Development, vol.108, pp.185-194, 1990.

F. M. Piccolo and A. G. Fisher, Getting rid of DNA methylation, Trends Cell Biol, vol.24, pp.136-143, 2014.

K. Polyak, Heterogeneity in breast cancer, J Clin Invest, vol.121, pp.3786-3788, 2011.

B. D. Pope, T. Ryba, V. Dileep, F. Yue, W. Wu et al., Topologically associating domains are stable units of replication-timing regulation, Nature, vol.515, pp.402-405, 2014.

T. A. Proia, P. J. Keller, P. B. Gupta, I. Klebba, A. D. Jones et al., Genetic predisposition directs breast cancer phenotype by dictating progenitor cell fate, Cell Stem Cell, vol.8, pp.149-163, 2011.

T. A. Quach, D. A. Villagómez, G. Coppola, A. Pinton, E. J. Hart et al., A cytogenetic study of breeding boars in Canada, Cytogenet Genome Res, vol.126, pp.271-280, 2009.
URL : https://hal.archives-ouvertes.fr/hal-02668874

E. J. Radford, M. Ito, H. Shi, J. A. Corish, K. Yamazawa et al., In utero effects. In utero undernourishment perturbs the adult sperm methylome and intergenerational metabolism, Science, vol.345, p.1255903, 2014.

S. S. Rao, M. H. Huntley, N. C. Durand, E. K. Stamenova, I. D. Bochkov et al., A 3D map of the human genome at kilobase resolution reveals principles of chromatin looping, Cell, vol.159, pp.1665-1680, 2014.

A. Rodríguez, C. Allegrucci, and R. Alberio, Modulation of pluripotency in the porcine embryo and iPS cells, PLoS One, vol.7, issue.11, p.49079, 2012.

H. M. Rowe, A. Kapopoulou, A. Corsinotti, L. Fasching, T. S. Macfarlan et al., TRIM28 repression of retrotransposon-based enhancers is necessary to preserve transcriptional dynamics in embryonic stem cells, Genome Res, vol.23, pp.452-461, 2013.

E. Rozbicki, M. Chuai, A. I. Karjalainen, F. Song, H. M. Sang et al., , 2015.

, Myosin-II-mediated cell shape changes and cell intercalation contribute to primitive streak formation, Nat Cell Biol, vol.17, pp.397-408

P. Samavarchi-tehrani, A. Golipour, L. David, H. K. Sung, T. A. Beyer et al., Functional genomics reveals a BMP-driven mesenchymal-to-epithelial transition in the initiation of somatic cell reprogramming, Cell Stem Cell, vol.7, pp.64-77, 2010.

M. G. Santoro, M. Fukushima, A. Benedetto, A. , and C. , PGJ2, a new antiviral prostaglandin: inhibition of Sendai virus replication and alteration of virus protein synthesis, J Gen Virol, vol.68, pp.1153-1158, 1987.

F. Seidel, Die Entwickunlspotenzen einer isolierten blastomere der Zweizellenstadiums in Saugetier Ei, Naturwissenschaften, vol.39, 1952.

J. Silva, O. Barrandon, J. Nichols, J. Kawaguchi, T. W. Theunissen et al., Promotion of reprogramming to ground state pluripotency by signal inhibition, PLoS Biol, vol.6, p.253, 2008.

P. Simpson, Maternal-Zygotic Gene Interactions during Formation of the Dorsoventral Pattern in Drosophila Embryos, Genetics, vol.105, pp.615-632, 1983.

A. G. Smith, J. K. Heath, D. D. Donaldson, G. G. Wong, J. Moreau et al., Inhibition of pluripotential embryonic stem cell differentiation by purified polypeptides, Nature, vol.336, pp.688-90, 1988.

P. Soriano, G. Friedrich, and P. Lawinger, Promoter interactions in retrovirus vectors introduced into fibroblasts and embryonic stem cells, J Virol, vol.65, pp.2314-2323, 1991.

H. Spemann, Embryonic Development and induction, 1938.

B. T. Spike, D. D. Engle, J. C. Lin, S. K. Cheung, J. La et al., A mammary stem cell population identified and characterized in late embryogenesis reveals similarities to human breast cancer, Cell Stem Cell, vol.10, pp.183-197, 2012.

L. C. Stevens, C. C. , and L. , Spontaneous testicular teratomas in an inbred strain of mice, Proc Natl Acad Sci U S A, vol.40, pp.1080-1087, 1954.

C. Stocking and C. A. Kozak, Murine endogenous retroviruses, Cell Mol Life Sci, vol.65, pp.3383-3398, 2008.

A. Streit, A. J. Berliner, C. Papanayotou, A. Sirulnik, and C. D. Stern, Initiation of neural induction by FGF signalling before gastrulation, Nature, vol.406, pp.74-82, 2000.

K. Takahashi, Y. , and S. , Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors, Cell, vol.126, pp.663-676, 2006.

Y. Takashima, G. Guo, R. Loos, J. Nichols, G. Ficz et al., Resetting transcription factor control circuitry toward ground-state pluripotency in human, Cell, vol.158, pp.1254-1269, 2014.

A. K. Tarkowski, Mouse chimeraes developed from fused eggs, Nature, vol.190, pp.857-860, 1961.

B. P. Telugu, T. Ezashi, and R. M. Roberts, Porcine induced pluripotent stem cells analogous to naïve and primed embryonic stem cells of the mouse', Int J Dev Biol, vol.54, pp.1703-1714, 2010.

P. J. Tesar, J. G. Chenoweth, F. A. Brook, T. J. Davies, E. P. Evans et al., New cell lines from mouse epiblast share defining features with human embryonic stem cells, Nature, vol.448, pp.196-199, 2007.

T. W. Theunissen, B. E. Powell, H. Wang, M. Mitalipova, D. A. Faddah et al., Systematic identification of culture conditions for induction and maintenance of naive human pluripotency, Cell Stem Cell, vol.15, pp.471-487, 2014.

J. P. Thiery, H. Acloque, R. Y. Huang, and M. A. Nieto, Epithelial-mesenchymal transitions in development and disease, Cell, vol.139, pp.871-890, 2009.

J. A. Thomson, J. Itskovitz-eldor, S. S. Shapiro, M. A. Waknitz, J. J. Swiergiel et al., Embryonic stem cell lines derived from human blastocysts, Science, vol.282, issue.5391, pp.1145-1152, 1998.

P. D. Tonge, A. J. Corso, C. Monetti, S. M. Hussein, M. C. Puri et al., Divergent reprogramming routes lead to alternative stem-cell states, Nature, vol.516, pp.192-197, 2014.

D. D. Tran, C. A. Corsa, H. Biswas, R. L. Aft, and G. D. Longmore, Temporal and spatial cooperation of Snail1 and Twist1 during epithelial-mesenchymal transition predicts for human breast cancer recurrence, Mol Cancer Res, vol.9, pp.1644-1657, 2011.

J. H. Tsai, J. L. Donaher, D. A. Murphy, S. Chau, Y. et al., Spatiotemporal regulation of epithelial-mesenchymal transition is essential for squamous cell carcinoma metastasis, Cancer Cell, vol.22, pp.725-736, 2012.

J. M. Turner, Meiotic sex chromosome inactivation, Development, vol.134, pp.1823-1831, 2007.
URL : https://hal.archives-ouvertes.fr/hal-01229825

J. M. Turner, O. Aprelikova, X. Xu, R. Wang, S. Kim et al., BRCA1, histone H2AX phosphorylation, and male meiotic sex chromosome inactivation, Curr Biol, vol.14, pp.2135-2142, 2004.

J. M. Turner, S. K. Mahadevaiah, P. J. Ellis, M. J. Mitchell, and P. S. Burgoyne, Pachytene asynapsis drives meiotic sex chromosome inactivation and leads to substantial postmeiotic repression in spermatids, Dev Cell, vol.10, pp.521-529, 2006.

J. J. Unternaehrer, R. Zhao, K. Kim, M. Cesana, J. T. Powers et al., The epithelial-mesenchymal transition factor SNAIL paradoxically enhances reprogramming, Stem Cell Reports, vol.3, pp.691-698, 2014.

S. Vega, A. V. Morales, O. H. Ocaña, F. Valdés, I. Fabregat et al., Snail blocks the cell cycle and confers resistance to cell death, Genes Dev, vol.18, pp.1131-1143, 2004.

F. D. West, S. L. Terlouw, D. J. Kwon, J. L. Mumaw, S. K. Dhara et al., Porcine induced pluripotent stem cells produce chimeric offspring, Stem Cells Dev, vol.19, issue.8, pp.1211-1231, 2010.

M. S. Wicha, Cancer stem cell heterogeneity in hereditary breast cancer, Breast Cancer Res, vol.10, p.105, 2008.

J. Wu, D. Okamura, M. Li, K. Suzuki, C. Luo et al., An alternative pluripotent state confers interspecies chimaeric competency, Nature, vol.521, pp.316-321, 2015.

Z. Wu, J. Chen, J. Ren, L. Bao, J. Liao et al., Generation of pig induced pluripotent stem cells with a drug-inducible system, J Mol Cell Biol, vol.1, issue.1, pp.46-54, 2009.

Z. Q. Wu, X. Y. Li, C. Y. Hu, M. Ford, C. G. Kleer et al., Canonical Wnt signaling regulates Slug activity and links epithelial-mesenchymal transition with epigenetic Breast Cancer 1, Early Onset (BRCA1) repression, Proc Natl Acad Sci U S A, vol.109, pp.16654-16659, 2012.

S. Yamashita, C. Miyagi, T. Fukada, N. Kagara, Y. S. Che et al., Zinc transporter LIVI controls epithelialmesenchymal transition in zebrafish gastrula organizer, Nature, vol.429, pp.298-302, 2004.

Q. L. Ying, J. Wray, J. Nichols, L. Batlle-morera, B. Doble et al., The ground state of embryonic stem cell self-renewal, Nature, vol.453, pp.519-523, 2008.

Y. Zhang, R. P. Mccord, Y. J. Ho, B. R. Lajoie, D. G. Hildebrand et al., Spatial organization of the mouse genome and its role in recurrent chromosomal translocations, Cell, vol.148, pp.908-921, 2012.

M. Zvelebil, E. Oliemuller, Q. Gao, O. Wansbury, A. Mackay et al., Embryonic mammary signature subsets are activated in Brca1-/-and basal-like breast cancers, Breast Cancer Res, vol.15, p.25, 2013.