M. Zuccotti, V. Merico, S. Cecconi, C. Redi, and S. Garagna, What does it take to make a developmentally competent mammalian egg? Hum Reprod Updat, pp.525-540, 2011.

R. Gilchrist, M. Lane, and J. Thompson, Oocyte-secreted factors: regulators of cumulus cell function and oocyte quality, Human Reproduction Update, vol.14, issue.2, pp.159-177, 2008.
DOI : 10.1093/humupd/dmm040

P. Mermillod, R. Dalbies-tran, S. Uzbekova, A. Thelie, J. Traverso et al., Factors affecting oocyte quality: who is driving the follicle? Reproduction in domestic animals = Zuchthygiene, pp.43393-400, 2008.

G. Kidder and B. Vanderhyden, Bidirectional communication between oocytes and follicle cells: ensuring oocyte developmental competence, Canadian Journal of Physiology and Pharmacology, vol.88, issue.4, pp.399-413, 2010.
DOI : 10.1139/Y10-009

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3025001

R. Taft, J. Denegre, F. Pendola, and J. Eppig, Identification of Genes Encoding Mouse Oocyte Secretory and Transmembrane Proteins by a Signal Sequence Trap1, Biology of Reproduction, vol.67, issue.3, pp.953-960, 2002.
DOI : 10.1095/biolreprod.102.005546

M. Conti, M. Hsieh, M. Zamah, A. Oh, and J. , Novel signaling mechanisms in the ovary during oocyte maturation and ovulation, Molecular and Cellular Endocrinology, vol.356, issue.1-2, pp.65-73, 2012.
DOI : 10.1016/j.mce.2011.11.002

N. Kimura, Y. Hoshino, K. Totsukawa, and E. Sato, Cellular and molecular events during oocyte maturation in mammals: molecules of cumulus-oocyte complex matrix and signalling pathways regulating meiotic progression. Society of Reproduction and Fertility supplement, pp.327-342, 2007.

R. Norris, M. Freudzon, V. Nikolaev, and L. Jaffe, Epidermal growth factor receptor kinase activity is required for gap junction closure and for part of the decrease in ovarian follicle cGMP in response to LH, Reproduction, vol.140, issue.5, pp.655-662, 2010.
DOI : 10.1530/REP-10-0288

V. Stetina, J. Orr-weaver, and T. , Developmental control of oocyte maturation and egg activation in metazoan models. Cold Spring Harbor perspectives in biology, p.5553, 2011.

G. De-montalembert, B. Jalabert, and C. Bry, Precocious induction of maturation and ovulation in northern pike (Esox lucius), Annales de Biologie Animale Biochimie Biophysique, vol.18, issue.4, pp.969-975, 1978.
DOI : 10.1051/rnd:19780533

URL : https://hal.archives-ouvertes.fr/hal-00897375

B. Jalabert, Production of fertilizable oocytes from follicles of rainbow trout (Salmo gairdnerii) following in vitro maturation and ovulation, Annales de Biologie Animale Biochimie Biophysique, vol.18, issue.2B, pp.461-470, 1978.
DOI : 10.1051/rnd:19780317

URL : https://hal.archives-ouvertes.fr/hal-00897310

B. Jalabert, B. Breton, and A. Fostier, Precocious induction of oocyte maturation and ovulation in rainbow trout (Salmo gairdneri) : problems when using 17 ??-hydroxy-20 ??-dihydroprogesterone, Annales de Biologie Animale Biochimie Biophysique, vol.18, issue.4, pp.977-984, 1978.
DOI : 10.1051/rnd:19780534

URL : https://hal.archives-ouvertes.fr/hal-00897376

J. Bobe, T. Nguyen, and A. Fostier, Ovarian function of the trout preovulatory ovary: New insights from recent gene expression studies, Comparative Biochemistry and Physiology Part A: Molecular & Integrative Physiology, vol.153, issue.1, pp.63-68, 2009.
DOI : 10.1016/j.cbpa.2008.10.021

E. Lubzens, G. Young, J. Bobe, and J. Cerda, Oogenesis in teleosts: How fish eggs are formed, General and Comparative Endocrinology, vol.165, issue.3, pp.367-389, 2010.
DOI : 10.1016/j.ygcen.2009.05.022

URL : https://hal.archives-ouvertes.fr/hal-01205005

R. Patino and C. Sullivan, Ovarian follicle growth, maturation, and ovulation in teleost fish, Fish Physiology and Biochemistry, vol.26, issue.1, pp.57-70, 2002.
DOI : 10.1023/A:1023311613987

M. Monaco, E. Villecco, and S. Sanchez, Summary, Zygote, vol.21, issue.02, pp.149-157, 2007.
DOI : 10.3109/15419060109080740

M. Rebagliati and I. Dawid, Expression of Activin Transcripts in Follicle Cells and Oocytes of Xenopus laevis, Developmental Biology, vol.159, issue.2, pp.574-580, 1993.
DOI : 10.1006/dbio.1993.1265

A. Fukui, R. Shiurba, and M. Asashima, Activin incorporation into vitellogenic oocytes of Xenopus laevis, Cell Mol Biol, vol.45, pp.545-554, 1999.

P. Sretarugsa and R. Wallace, The developing Xenopus oocyte specifies the type of gonadotropin-stimulated steroidogenesis performed by its associated follicle cells, Development, Growth and Differentiation, vol.118, issue.1, pp.87-97, 1997.
DOI : 10.1210/en.133.1.423

L. Li, C. Stoeckert, . Jr, and D. Roos, OrthoMCL: Identification of Ortholog Groups for Eukaryotic Genomes, Genome Research, vol.13, issue.9, pp.2178-2189, 2003.
DOI : 10.1101/gr.1224503

A. Balboula, K. Yamanaka, M. Sakatani, A. Hegab, S. Zaabel et al., Cathepsin B activity is related to the quality of bovine cumulus oocyte complexes and its inhibition can improve their developmental competence, Molecular Reproduction and Development, vol.438, issue.83, pp.439-448, 2010.
DOI : 10.1002/mrd.21164

S. Oksjoki, M. Soderstrom, E. Vuorio, and L. Anttila, Differential expression patterns of cathepsins B, H, K, L and S in the mouse ovary, Molecular Human Reproduction, vol.7, issue.1, pp.27-34, 2001.
DOI : 10.1093/molehr/7.1.27

C. Howe, G. Overton, J. Sawicki, D. Solter, P. Stein et al., Expression of SPARC/osteonectin transcript in murine embryos and gonads. Differentiation; research in biological diversity, pp.20-25, 1988.

M. Raty, E. Ketoja, T. Pitkanen, V. Ahola, K. Kananen et al., In vitro maturation supplements affect developmental competence of bovine cumulus???oocyte complexes and embryo quality after vitrification, Cryobiology, vol.63, issue.3, pp.245-255, 2011.
DOI : 10.1016/j.cryobiol.2011.09.134

Y. Sidis, T. Fujiwara, L. Leykin, K. Isaacson, T. Toth et al., Characterization of Inhibin/Activin Subunit, Activin Receptor, and Follistatin Messenger Ribonucleic Acid in Human and Mouse Oocytes: Evidence for Activin's Paracrine Signaling from Granulosa Cells to Oocytes1, Biology of Reproduction, vol.59, issue.4, pp.807-812, 1998.
DOI : 10.1095/biolreprod59.4.807

O. Patel, A. Bettegowda, J. Ireland, P. Coussens, P. Lonergan et al., Functional genomics studies of oocyte competence: evidence that reduced transcript abundance for follistatin is associated with poor developmental competence of bovine oocytes, Reproduction, vol.133, issue.1, pp.95-106, 2007.
DOI : 10.1530/rep.1.01123

C. Rico, C. Medigue, S. Fabre, P. Jarrier, M. Bontoux et al., Regulation of Anti-M??llerian Hormone Production in the Cow: A Multiscale Study at Endocrine, Ovarian, Follicular, and Granulosa Cell Levels1, Biology of Reproduction, vol.84, issue.3, pp.560-571, 2011.
DOI : 10.1095/biolreprod.110.088187

J. Taieb, M. Grynberg, A. Pierre, N. Arouche, P. Massart et al., FSH and Its Second Messenger cAMP Stimulate the Transcription of Human Anti-M??llerian Hormone in Cultured Granulosa Cells, Molecular Endocrinology, vol.25, issue.4, pp.645-655, 2011.
DOI : 10.1210/me.2010-0297

W. Baarends, J. Uilenbroek, P. Kramer, J. Hoogerbrugge, E. Van-leeuwen et al., Anti-mullerian hormone and anti-mullerian hormone type II receptor messenger ribonucleic acid expression in rat ovaries during postnatal development, the estrous cycle, and gonadotropin-induced follicle growth, Endocrinology, vol.136, pp.4951-4962, 1995.

Y. Mishina, R. Rey, M. Finegold, M. Matzuk, N. Josso et al., Genetic analysis of the Mullerian-inhibiting substance signal transduction pathway in mammalian sexual differentiation., Genes & Development, vol.10, issue.20, pp.2577-2587, 1996.
DOI : 10.1101/gad.10.20.2577

M. Mihm, P. Baker, J. Ireland, G. Smith, P. Coussens et al., Molecular Evidence That Growth of Dominant Follicles Involves a Reduction in Follicle-Stimulating Hormone Dependence and an Increase in Luteinizing Hormone Dependence in Cattle1, Biology of Reproduction, vol.74, issue.6, pp.1051-1059, 2006.
DOI : 10.1095/biolreprod.105.045799

J. Park, F. Richard, S. Chun, J. Park, E. Law et al., Phosphodiesterase Regulation Is Critical for the Differentiation and Pattern of Gene Expression in Granulosa Cells of the Ovarian Follicle, Molecular Endocrinology, vol.17, issue.6, pp.1117-1130, 2003.
DOI : 10.1210/me.2002-0435

M. Sasseville, F. Albuz, N. Cote, C. Guillemette, R. Gilchrist et al., Characterization of Novel Phosphodiesterases in the Bovine Ovarian Follicle1, Biology of Reproduction, vol.81, issue.2, pp.415-425, 2009.
DOI : 10.1095/biolreprod.108.074450

R. Thomas, D. Armstrong, and R. Gilchrist, Bovine Cumulus Cell-Oocyte Gap Junctional Communication During In Vitro Maturation in Response to Manipulation of Cell-Specific Cyclic Adenosine 3???,5???-Monophosophate Levels1, Biology of Reproduction, vol.70, issue.3, pp.548-556, 2004.
DOI : 10.1095/biolreprod.103.021204

M. Byers, G. Kuiper, J. Gustafsson, and O. Park-sarge, Estrogen receptor-beta mRNA expression in rat ovary: down-regulation by gonadotropins, Mol Endocrinol, vol.11, pp.172-182, 1997.

J. Couse, J. Lindzey, K. Grandien, J. Gustafsson, and K. Korach, Tissue distribution and quantitative analysis of estrogen receptor-alpha (ERalpha) and estrogen receptor-beta (ERbeta) messenger ribonucleic acid in the wild-type and ERalpha-knockout mouse, Endocrinology, vol.138, pp.4613-4621, 1997.

C. Chase, . Jr, C. Kirby, A. Hammond, T. Olson et al., Patterns of ovarian growth and development in cattle with a growth hormone receptor deficiency., Journal of Animal Science, vol.76, issue.1, pp.212-219, 1998.
DOI : 10.2527/1998.761212x

D. Zaczek, J. Hammond, L. Suen, S. Wandji, D. Service et al., Impact of Growth Hormone Resistance on Female Reproductive Function: New Insights from Growth Hormone Receptor Knockout Mice1, Biology of Reproduction, vol.67, issue.4, pp.1115-1124, 2002.
DOI : 10.1095/biolreprod67.4.1115

J. Silva, J. Figueiredo, and R. Van-den-hurk, Involvement of growth hormone (GH) and insulin-like growth factor (IGF) system in ovarian folliculogenesis, Theriogenology, vol.71, issue.8, pp.1193-1208, 2009.
DOI : 10.1016/j.theriogenology.2008.12.015

J. Gomez, C. Weil, M. Ollitrault, L. Bail, P. Breton et al., Growth Hormone (GH) and Gonadotropin Subunit Gene Expression and Pituitary and Plasma Changes during Spermatogenesis and Oogenesis in Rainbow Trout (Oncorhynchus mykiss), General and Comparative Endocrinology, vol.113, issue.3, pp.413-428, 1999.
DOI : 10.1006/gcen.1998.7222

O. Onagbesan, V. Bruggeman, and E. Decuypere, Intra-ovarian growth factors regulating ovarian function in avian species: A review, Animal Reproduction Science, vol.111, issue.2-4, pp.121-140, 2009.
DOI : 10.1016/j.anireprosci.2008.09.017

K. Roby and P. Terranova, on Steroidogenesis of Healthy and Atretic Follicles of the Rat: Theca as a Target*, Endocrinology, vol.126, issue.5, pp.2711-2718, 1990.
DOI : 10.1210/endo-126-5-2711

A. Amsterdam, I. Keren-tal, D. Aharoni, A. Dantes, A. Land-bracha et al., Steroidogenesis and apoptosis in the mammalian ovary, Steroids, vol.68, issue.10-13, pp.861-867, 2003.
DOI : 10.1016/j.steroids.2003.09.003

L. Clark, M. Wei, G. Cattoretti, C. Mendelsohn, and B. Tycko, The Tnfrh1 (Tnfrsf23) gene is weakly imprinted in several organs and expressed at the trophoblast-decidua interface, BMC Genetics, vol.3, issue.1, p.11, 2002.
DOI : 10.1186/1471-2156-3-11

J. Bridgham and A. Johnson, Alternatively Spliced Variants of Gallus gallus TNFRSF23 Are Expressed in the Ovary and Differentially Regulated by Cell Signaling Pathways1, Biology of Reproduction, vol.70, issue.4, pp.972-979, 2004.
DOI : 10.1095/biolreprod.103.023614

J. Bobe and F. Goetz, A Tumor Necrosis Factor Decoy Receptor Homologue Is Up-Regulated in the Brook Trout (Salvelinus fontinalis) Ovary at the Completion of Ovulation1, Biology of Reproduction, vol.62, issue.2, pp.420-426, 2000.
DOI : 10.1095/biolreprod62.2.420

T. Ding, Z. Li, T. Hailemariam, S. Mukherjee, F. Maxfield et al., SMS overexpression and knockdown: impact on cellular sphingomyelin and diacylglycerol metabolism, and cell apoptosis, The Journal of Lipid Research, vol.49, issue.2, pp.376-385, 2008.
DOI : 10.1194/jlr.M700401-JLR200

S. Perlman, T. Bouquin, B. Van-den-hazel, T. Jensen, H. Schambye et al., Transcriptome analysis of FSH and FSH variant stimulation in granulosa cells from IVM patients reveals novel regulated genes, Molecular Human Reproduction, vol.12, issue.3, pp.135-144, 2006.
DOI : 10.1093/molehr/gah247

H. Lu and D. Bilder, Endocytic control of epithelial polarity and proliferation in Drosophila, Nature Cell Biology, vol.11, issue.12, pp.1232-1239, 2005.
DOI : 10.1242/dev.00643

M. Cuajungco, C. Grimm, K. Oshima, D. Hoedt, D. Nilius et al., PACSINs Bind to the TRPV4 Cation Channel: PACSIN 3 MODULATES THE SUBCELLULAR LOCALIZATION OF TRPV4, Journal of Biological Chemistry, vol.281, issue.27, pp.18753-18762, 2006.
DOI : 10.1074/jbc.M602452200

S. Roth, F. Neuman-silberberg, G. Barcelo, and T. Schupbach, cornichon and the EGF receptor signaling process are necessary for both anterior-posterior and dorsal-ventral pattern formation in Drosophila, Cell, vol.81, issue.6, pp.967-978, 1995.
DOI : 10.1016/0092-8674(95)90016-0

C. Castro, D. Piscopo, T. Nakagawa, and R. Derynck, Cornichon regulates transport and secretion of TGF??-related proteins in metazoan cells, Journal of Cell Science, vol.120, issue.14, pp.2454-2466, 2007.
DOI : 10.1242/jcs.004200

F. Neuman-silberberg and T. Schupbach, The Drosophila TGF-??-like protein Gurken: expression and cellular localization during Drosophila oogenesis, Mechanisms of Development, vol.59, issue.2, pp.105-113, 1996.
DOI : 10.1016/0925-4773(96)00567-9

E. Kiss-toth, Tribbles: ???puzzling??? regulators of cell signalling, Biochemical Society Transactions, vol.33, issue.2, pp.684-687, 2011.
DOI : 10.1172/JCI41366

T. Yokoyama, Y. Kanno, Y. Yamazaki, T. Takahara, S. Miyata et al., Trib1 links the MEK1/ERK pathway in myeloid leukemogenesis, Blood, vol.116, issue.15, pp.2768-2775, 2010.
DOI : 10.1182/blood-2009-10-246264

R. Burkhardt, S. Toh, W. Lagor, A. Birkeland, M. Levin et al., Trib1 is a lipid- and myocardial infarction???associated gene that regulates hepatic lipogenesis and VLDL production in mice, Journal of Clinical Investigation, vol.120, issue.12, pp.4410-4414, 2010.
DOI : 10.1172/JCI44213DS1

M. Yamamoto, S. Uematsu, T. Okamoto, Y. Matsuura, S. Sato et al., Enhanced TLR-mediated NF-IL6 dependent gene expression by Trib1 deficiency. The Journal of experimental medicine, pp.2233-2239, 2007.
DOI : 10.1084/jem.20070183

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2118688

P. Rorth, K. Szabo, and G. Texido, The Level of C/EBP Protein Is Critical for Cell Migration during Drosophila Oogenesis and Is Tightly Controlled by Regulated Degradation, Molecular Cell, vol.6, issue.1, pp.23-30, 2000.
DOI : 10.1016/S1097-2765(00)00004-6

D. Robinson, K. Cant, and L. Cooley, Morphogenesis of Drosophila ovarian ring canals, Development, vol.120, pp.2015-2025, 1994.

S. Wang, Z. Zhou, K. Ying, R. Tang, Y. Huang et al., Cloning and characterization of KLHL5, a novel human gene encoding a kelch-related protein with a BTB domain, Biochemical Genetics, vol.39, issue.7/8, pp.227-238, 2001.
DOI : 10.1023/A:1010203114697

G. Suske, E. Bruford, and S. Philipsen, Mammalian SP/KLF transcription factors: Bring in the family, Genomics, vol.85, issue.5, pp.551-556, 2005.
DOI : 10.1016/j.ygeno.2005.01.005

B. Mcconnell and V. Yang, Mammalian Kruppel-Like Factors in Health and Diseases, Physiological Reviews, vol.90, issue.4, pp.1337-1381, 2010.
DOI : 10.1152/physrev.00058.2009

S. Natesampillai, J. Kerkvliet, P. Leung, and J. Veldhuis, Regulation of Kruppel-like factor 4, 9, and 13 genes and the steroidogenic genes LDLR, StAR, and CYP11A in ovarian granulosa cells, AJP: Endocrinology and Metabolism, vol.294, issue.2, pp.385-391, 2008.
DOI : 10.1152/ajpendo.00480.2007

P. Kezele, J. Ague, E. Nilsson, and M. Skinner, Alterations in the Ovarian Transcriptome During Primordial Follicle Assembly and Development1, Biology of Reproduction, vol.72, issue.1, pp.241-255, 2005.
DOI : 10.1095/biolreprod.104.032060

E. Jansen, J. Laven, H. Dommerholt, J. Polman, C. Van-rijt et al., Abnormal Gene Expression Profiles in Human Ovaries from Polycystic Ovary Syndrome Patients, Molecular Endocrinology, vol.18, issue.12, pp.3050-3063, 2004.
DOI : 10.1210/me.2004-0074

W. Yan, K. Burns, L. Ma, and M. Matzuk, Identification of Zfp393, a germ cell-specific gene encoding a novel zinc finger protein, Mechanisms of Development, vol.118, issue.1-2, pp.233-239, 2002.
DOI : 10.1016/S0925-4773(02)00258-7

J. Richards, Ovulation: New factors that prepare the oocyte for fertilization, Molecular and Cellular Endocrinology, vol.234, issue.1-2, pp.75-79, 2005.
DOI : 10.1016/j.mce.2005.01.004

J. Bobe, J. Montfort, T. Nguyen, and A. Fostier, Identification of new participants in the rainbow trout (Oncorhynchus mykiss) oocyte maturation and ovulation processes using cDNA microarrays, Reproductive Biology and Endocrinology, vol.4, issue.1, p.39, 2006.
DOI : 10.1186/1477-7827-4-39

J. Bobe, T. Nguyen, and B. Jalabert, Targeted Gene Expression Profiling in the Rainbow Trout (Oncorhynchus mykiss) Ovary During Maturational Competence Acquisition and Oocyte Maturation, Biology of Reproduction, vol.71, issue.1, pp.73-82, 2004.
DOI : 10.1095/biolreprod.103.025205

H. Rime, T. Nguyen, J. Bobe, A. Fostier, and G. Monod, Prochloraz-induced Oocyte Maturation in Rainbow Trout (Oncorhynchus mykiss), a Molecular and Functional Analysis, Toxicological Sciences, vol.118, issue.1, pp.61-70, 2010.
DOI : 10.1093/toxsci/kfq255

URL : https://hal.archives-ouvertes.fr/hal-01205007

J. Dumont, Oogenesis inXenopus laevis (Daudin). I. Stages of oocyte development in laboratory maintained animals, Journal of Morphology, vol.63, issue.2, pp.153-179, 1972.
DOI : 10.1002/jmor.1051360203

M. Salem, P. Kenney, C. Rexroad, and J. Yao, Development of a 37 k high-density oligonucleotide microarray: a new tool for functional genome research in rainbow trout, Journal of Fish Biology, vol.42, issue.9, pp.2187-2206, 2008.
DOI : 10.1007/s00251-004-0645-6

M. Gohin, P. Bodinier, A. Fostier, F. Chesnel, and J. Bobe, Aromatase is expressed and active in the rainbow trout oocyte during final oocyte maturation, Molecular Reproduction and Development, vol.29, issue.7, pp.510-518, 2011.
DOI : 10.1002/mrd.21335

URL : https://hal.archives-ouvertes.fr/inserm-00604822

M. Salhab, L. Tosca, C. Cabau, P. Papillier, C. Perreau et al., Kinetics of gene expression and signaling in bovine cumulus cells throughout IVM in different mediums in relation to oocyte developmental competence, cumulus apoptosis and progesterone secretion, Theriogenology, vol.75, issue.1, pp.90-104, 2011.
DOI : 10.1016/j.theriogenology.2010.07.014

URL : https://hal.archives-ouvertes.fr/hal-01129418

F. Chen, A. Mackey, C. Stoeckert, . Jr, and D. Roos, OrthoMCL-DB: querying a comprehensive multi-species collection of ortholog groups, Nucleic Acids Research, vol.34, issue.90001, pp.363-368, 2006.
DOI : 10.1093/nar/gkj123

E. Bonnet, A. Fostier, and J. Bobe, Microarray-based analysis of fish egg quality after natural or controlled ovulation, BMC Genomics, vol.8, issue.1, p.55, 2007.
DOI : 10.1186/1471-2164-8-55

D. Huang, B. Sherman, and R. Lempicki, Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources, Nature Protocols, vol.99, issue.1, pp.44-57, 2009.
DOI : 10.1038/nprot.2008.211