B. Levavi-sivan, J. Bogerd, E. L. Mananos, A. Gomez, and J. J. Lareyre, Perspectives on fish gonadotropins and their receptors, Gen Comp Endocrinol, vol.165, issue.3, pp.412-449, 2010.
URL : https://hal.archives-ouvertes.fr/hal-01204994

R. W. Schulz, L. R. De-franca, J. J. Lareyre, L. Gac, F. Chiarini-garcia et al., Gen Comp Endocrinol, vol.165, issue.3, pp.390-411, 2010.

Y. Zohar, J. A. Munoz-cueto, A. Elizur, and O. Kah, Neuroendocrinology of reproduction in teleost fish, Gen Comp Endocrinol, vol.165, issue.3, pp.438-55, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00461596

V. L. Trudeau, Facing the challenges of neuropeptide gene knockouts: why do they not inhibit reproduction in adult teleost fish?, Front Neurosci, vol.12, p.302, 2018.

L. Chu, J. Li, Y. Liu, W. Hu, and C. H. Cheng, Targeted gene disruption in zebrafish reveals noncanonical functions of LH signaling in reproduction, Mol Endocrinol, vol.28, issue.11, pp.1785-95, 2014.

Z. Zhang, S. W. Lau, L. Zhang, and W. Ge, Disruption of zebrafish follicle-stimulating hormone receptor (fshr) but not luteinizing hormone receptor (lhcgr) gene by TALEN leads to failed follicle activation in females followed by sexual reversal to males, Endocrinology, vol.156, issue.10, pp.3747-62, 2015.

Z. Zhang, B. Zhu, and W. Ge, Genetic analysis of zebrafish gonadotropin (FSH and LH) functions by TALEN-mediated gene disruption, Mol Endocrinol, vol.29, issue.1, pp.76-98, 2015.

T. Sato, A. Suzuki, N. Shibata, M. Sakaizumi, and S. Hamaguchi, The novel mutant scl of the medaka fish, Oryzias latipes, shows no secondary sex characters, Zool Sci, vol.25, issue.3, pp.299-306, 2008.

G. Zhai, T. Shu, Y. Xia, Y. Lu, G. Shang et al., Characterization of sexual trait development in cyp17a1-deficient zebrafish, Endocrinology, vol.159, issue.10, pp.3549-62, 2018.

C. M. Crowder, C. S. Lassiter, and D. A. Gorelick, Nuclear androgen receptor regulates testes organization and oocyte maturation in zebrafish, Endocrinology, vol.159, issue.2, pp.980-93, 2018.

H. Tang, Y. Chen, L. Wang, Y. Yin, G. Li et al., Fertility impairment with defective spermatogenesis and steroidogenesis in male zebrafish lacking androgen receptor, Biol Reprod, vol.98, issue.2, pp.227-265, 2018.

K. De-gendt, J. V. Swinnen, P. T. Saunders, L. Schoonjans, M. Dewerchin et al., A Sertoli cell-selective knockout of the androgen receptor causes spermatogenic arrest in meiosis, Proc Natl Acad Sci U S A, vol.101, issue.5, pp.1327-1359, 2004.

X. Ma, Y. Dong, M. M. Matzuk, and T. R. Kumar, Targeted disruption of luteinizing hormone beta-subunit leads to hypogonadism, defects in gonadal steroidogenesis, and infertility, Proc Natl Acad Sci U S A, vol.101, issue.49, pp.17294-17303, 2004.

O. O. Oduwole, H. Peltoketo, A. Poliandri, L. Vengadabady, M. Chrusciel et al., Constitutively active follicle-stimulating hormone receptor enables androgen-independent spermatogenesis, J Clin Invest, vol.128, issue.5, pp.1787-92, 2018.

M. C. Leal, P. P. De-waal, A. Garcia-lopez, S. X. Chen, J. Bogerd et al., Zebrafish primary testis tissue culture: an approach to study testis function ex vivo, Gen Comp Endocrinol, vol.162, issue.2, pp.134-142, 2009.

T. Miura, K. Yamauchi, H. Takahashi, and Y. Nagahama, Hormonal induction of all stages of spermatogenesis in vitro in the male Japanese eel (Anguilla japonica), Proc Natl Acad Sci, vol.88, issue.13, pp.5774-5782, 1991.

D. Crespo, L. Assis, T. Furmanek, J. Bogerd, and R. W. Schulz, Expression profiling identifies Sertoli and Leydig cell genes as Fsh targets in adult zebrafish testis, Mol Cell Endocrinol, vol.437, pp.237-51, 2016.

D. Safian, N. Ryane, J. Bogerd, and R. W. Schulz, Fsh stimulates Leydig cell Wnt5a production, enriching zebrafish type a spermatogonia, J Endocrinol, vol.239, issue.3, pp.351-63, 2018.

T. Miura, C. Miura, Y. Konda, and K. Yamauchi, Spermatogenesis-preventing substance in Japanese eel, Development, vol.129, issue.11, pp.2689-97, 2002.

R. H. Nobrega, R. D. Morais, D. Crespo, P. P. De-waal, L. R. De-franca et al., Fsh stimulates Spermatogonial proliferation and differentiation in zebrafish via Igf3, Endocrinology, vol.156, issue.10, pp.3804-3821, 2015.

K. S. Skaar, R. H. Nobrega, A. Magaraki, L. C. Olsen, R. W. Schulz et al., Proteolytically activated, recombinant anti-mullerian hormone inhibits androgen secretion, proliferation, and differentiation of spermatogonia in adult zebrafish testis organ cultures, Endocrinology, vol.152, issue.9, pp.3527-3567, 2011.

C. D. Green, Q. Ma, G. L. Manske, A. N. Shami, X. Zheng et al., A comprehensive roadmap of murine spermatogenesis defined by single-cell RNA-Seq, Dev Cell, vol.46, issue.5, pp.651-667, 2018.

J. J. Koskenniemi, H. E. Virtanen, and J. Toppari, Testicular growth and development in puberty, Curr Opin Endocrinol Diabetes Obes, vol.24, issue.3, pp.215-239, 2017.

D. G. De-rooij, The nature and dynamics of spermatogonial stem cells, Development, vol.144, issue.17, pp.3022-3052, 2017.

E. Kjaerner-semb, F. Ayllon, L. Kleppe, E. Sorhus, K. Skaftnesmo et al., Vgll3 and the hippo pathway are regulated in Sertoli cells upon entry and during puberty in Atlantic salmon testis, Sci Rep, vol.8, issue.1, p.1912, 2018.

M. C. Melo, E. Andersson, P. G. Fjelldal, J. Bogerd, L. R. Franca et al., Salinity and photoperiod modulate pubertal development in Atlantic salmon (Salmo salar), J Endocrinol, vol.220, issue.3, pp.319-351, 2014.

K. Shearer, P. Parkins, B. Gadberry, B. Beckman, and P. Swanson, Effects of growth rate/body size and a low lipid diet on the incidence of early sexual maturation in juvenile male spring Chinook salmon (Oncorhynchus tshawytscha), Aquaculture, vol.252, pp.545-56, 2006.

P. G. Fjelldal, T. Hansen, and T. Huang, Continuous light and elevated temperature can trigger maturation both during and immediately after smoltification in male Atlantic salmon (Salmo salar), Aquaculture, vol.321, pp.93-100, 2011.

P. G. Fjelldal, R. Schulz, T. O. Nilsen, E. Andersson, B. Norberg et al., Sexual maturation and smoltification in domesticated Atlantic salmon (Salmo salar L.) -is there a developmental conflict, Physiol Rep, vol.6, issue.17, p.13809, 2018.

G. L. Taranger, M. Carrillo, R. W. Schulz, P. Fontaine, S. Zanuy et al., Control of puberty in farmed fish, Gen Comp Endocrinol, vol.165, issue.3, pp.483-515, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00107062

B. Campbell, J. T. Dickey, and P. Swanson, Endocrine changes during onset of puberty in male spring Chinook salmon, Oncorhynchus tshawytscha, Biol Reprod, vol.69, issue.6, pp.2109-2126, 2003.

J. M. Gomez, C. Weil, M. Ollitrault, P. Y. Lebail, B. 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), Gen Comp Endocrinol, vol.113, pp.413-441, 1999.

T. Ohta, H. Miyake, C. Miura, H. Kamei, A. K. Miura et al., Follicle-stimulating hormone induces spermatogenesis mediated by androgen production in Japanese eel, Anguilla japonica, Biol Reprod, vol.77, issue.6, pp.970-977, 2007.

A. Garcia-lopez, H. De-jonge, R. H. Nobrega, P. P. De-waal, W. Van-dijk et al., Studies in zebrafish reveal unusual cellular expression patterns of gonadotropin receptor messenger ribonucleic acids in the testis and unexpected functional differentiation of the gonadotropins, Endocrinology, vol.151, issue.5, pp.2349-60, 2010.

J. V. Planas and P. Swanson, Maturation-associated changes in the response of the salmon testis to the steroidogenic actions of gonadotropins (GTH I and GTH II) in vitro, Biol Reprod, vol.52, pp.697-704, 1995.

M. J. Mazon, A. Gomez, O. Yilmaz, M. Carrillo, and S. Zanuy, Administration of follicle-stimulating hormone in vivo triggers testicular recrudescence of juvenile European sea bass (Dicentrarchus labrax), Biol Reprod, vol.90, issue.1, p.6, 2014.

M. C. Melo, P. Van-dijk, E. Andersson, T. O. Nilsen, P. G. Fjelldal et al., Androgens directly stimulate spermatogonial differentiation in juvenile Atlantic salmon (Salmo salar), Gen Comp Endocrinol, vol.211, pp.52-61, 2015.

E. Sambroni, J. J. Lareyre, L. Gac, and F. , Fsh controls gene expression in fish both independently of and through steroid mediation, PLoS One, vol.8, issue.10, p.76684, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00934838

B. Borg, Androgens in teleost fishes, Comp Biochem Physiol C: Pharmacol Toxicol Endocrinol, vol.109, issue.3, pp.219-264, 1994.

L. H. De-castro-assis, R. H. De-nobrega, N. E. Gomez-gonzalez, J. Bogerd, and R. W. Schulz, Estrogen-induced inhibition of spermatogenesis in zebrafish is largely reversed by androgen, J Mol Endocrinol, vol.60, issue.4, pp.273-84, 2018.

J. E. Cavaco, C. Vilrokx, V. L. Trudeau, R. W. Schulz, and H. J. Goos, Sex steroids and the initiation of puberty in male African catfish (Clarias gariepinus), Am J Phys, vol.275, issue.6, pp.1793-802, 1998.

M. Blazquez, P. Medina, B. Crespo, A. Gomez, and S. Zanuy, Identification of conserved genes triggering puberty in European sea bass males (Dicentrarchus labrax) by microarray expression profiling, BMC Genomics, vol.18, issue.1, p.441, 2017.

A. D. Rolland, J. J. Lareyre, A. S. Goupil, J. Montfort, M. J. Ricordel et al., Expression profiling of rainbow trout testis development identifies evolutionary conserved genes involved in spermatogenesis, BMC Genomics, vol.10, p.546, 2009.
URL : https://hal.archives-ouvertes.fr/inserm-00663561

E. Sambroni, A. D. Rolland, and J. J. Lareyre, Le Gac F: FSH and LH have common and distinct effects on gene expression in rainbow trout testis, J Mol Endocrinol, vol.50, issue.1, pp.1-18, 2013.

X. Wang, Q. Liu, S. Xu, Y. Xiao, Y. Wang et al., Transcriptome dynamics during turbot spermatogenesis predicting the potential key genes regulating Male germ cell proliferation and maturation, Sci Rep, vol.8, issue.1, p.15825, 2018.

S. S. Hammoud, D. H. Low, C. Yi, D. T. Carrell, E. Guccione et al., Chromatin and transcription transitions of mammalian adult germline stem cells and spermatogenesis, Cell Stem Cell, vol.15, issue.2, pp.239-53, 2014.

S. S. Hammoud, D. H. Low, C. Yi, C. L. Lee, J. M. Oatley et al., Transcription and imprinting dynamics in developing postnatal male germline stem cells, Genes Dev, vol.29, issue.21, pp.2312-2336, 2015.

A. D. Rolland, A. Lardenois, A. S. Goupil, J. J. Lareyre, R. Houlgatte et al., Profiling of androgen response in rainbow trout pubertal testis: relevance to male gonad development and spermatogenesis, PLoS One, vol.8, issue.1, p.53302, 2013.
URL : https://hal.archives-ouvertes.fr/inserm-00825763

D. Martinovic-weigelt, R. L. Wang, D. L. Villeneuve, D. C. Bencic, J. Lazorchak et al., Gene expression profiling of the androgen receptor antagonists flutamide and vinclozolin in zebrafish (Danio rerio) gonads, Aquat Toxicol, vol.101, issue.2, pp.447-58, 2011.

K. De-gendt and G. Verhoeven, Tissue-and cell-specific functions of the androgen receptor revealed through conditional knockout models in mice, Mol Cell Endocrinol, vol.352, issue.1-2, pp.13-25, 2012.

L. H. Assis, D. Crespo, R. D. Morais, L. R. Franca, J. Bogerd et al., INSL3 stimulates spermatogonial differentiation in testis of adult zebrafish (Danio rerio), Cell Tissue Res, vol.363, issue.2, pp.579-88, 2016.

T. Miura, C. Miura, K. Yamauchi, and Y. Nagahama, Human recombinant activin induces proliferation of spermatogonia in vitro in the Japanese eel Anguilla japonica, Fish Sci, vol.61, pp.434-441, 1995.

T. Miura and C. Miura, Japanese eel: a model for analysis of spermatogenesis, Zool Sci, vol.18, pp.1055-63, 2001.

D. P. Brazil, R. H. Church, S. Surae, C. Godson, and F. Martin, BMP signalling: agony and antagony in the family, Trends Cell Biol, vol.25, issue.5, pp.249-64, 2015.

J. C. Neumann, G. L. Chandler, V. A. Damoulis, N. J. Fustino, K. Lillard et al., Mutation in the type IB bone morphogenetic protein receptor Alk6b impairs germ-cell differentiation and causes germ-cell tumors in zebrafish, Proc Natl Acad Sci, vol.108, issue.32, pp.13153-13161, 2011.

B. Mulloy and C. C. Rider, The bone morphogenetic proteins and their antagonists, Vitam Horm, vol.99, pp.63-90, 2015.

B. Christen, A. M. Rodrigues, M. B. Monasterio, C. F. Roig, I. Belmonte et al., Transient downregulation of bmp signalling induces extra limbs in vertebrates, Development, vol.139, issue.14, pp.2557-65, 2012.

C. Morinaga, D. Saito, S. Nakamura, T. Sasaki, S. Asakawa et al., The hotei mutation of medaka in the anti-Mullerian hormone receptor causes the dysregulation of germ cell and sexual development, Proc Natl Acad Sci, vol.104, issue.23, pp.9691-9697, 2007.

Q. Lin, J. Mei, Z. Li, X. Zhang, L. Zhou et al., Distinct and cooperative roles of amh and dmrt1 in self-renewal and differentiation of Male germ cells in zebrafish, Genetics, vol.207, issue.3, pp.1007-1029, 2017.

R. Morais, D. Crespo, R. H. Nobrega, M. S. Lemos, H. Van-de-kant et al., Antagonistic regulation of spermatogonial differentiation in zebrafish (Danio rerio) by Igf3 and Amh, Mol Cell Endocrinol, vol.454, pp.112-136, 2017.

S. Yamaguchi, C. Miura, K. Kikuchi, F. T. Celino, T. Agusa et al., Zinc is an essential trace element for spermatogenesis, Proc Natl Acad Sci, vol.106, issue.26, pp.10859-64, 2009.

F. T. Celino, S. Yamaguchi, C. Miura, T. Ohta, Y. Tozawa et al., Tolerance of spermatogonia to oxidative stress is due to high levels of Zn and cu/Zn superoxide dismutase, PLoS One, vol.6, issue.2, p.16938, 2011.

N. Shyh-chang, G. Q. Daley, and L. C. Cantley, Stem cell metabolism in tissue development and aging, Development, vol.140, issue.12, pp.2535-2582, 2013.

K. Ito, Metabolism and the control of cell fate decisions and stem cell renewal, Annu Rev Cell Dev Biol, vol.32, pp.399-409, 2016.

Y. Kitajima, Y. Tashiro, N. Suzuki, H. Warita, M. Kato et al., Proteasome dysfunction induces muscle growth defects and protein aggregation, J Cell Sci, vol.127, pp.5204-5221, 2014.

N. Hedhli and C. Depre, Proteasome inhibitors and cardiac cell growth, Cardiovasc Res, vol.85, issue.2, pp.321-330, 2010.

L. Bedford, D. Hay, A. Devoy, S. Paine, D. G. Powe et al., Depletion of 26S proteasomes in mouse brain neurons causes neurodegeneration and Lewy-like inclusions resembling human pale bodies, J Neurosci, vol.28, issue.33, pp.8189-98, 2008.

A. Mofers, P. Pellegrini, S. Linder, D. 'arcy, and P. , Proteasome-associated deubiquitinases and cancer, Cancer Metastasis Rev, vol.36, issue.4, pp.635-53, 2017.

V. Murillo-garzon, I. Gorrono-etxebarria, M. Akerfelt, M. C. Puustinen, L. Sistonen et al., Frizzled-8 integrates Wnt-11 and transforming growth factor-beta signaling in prostate cancer, Nat Commun, vol.9, issue.1, p.1747, 2018.

A. A. Sharov, G. Falco, Y. Piao, S. Poosala, K. G. Becker et al., Effects of aging and calorie restriction on the global gene expression profiles of mouse testis and ovary, BMC Biol, vol.6, p.24, 2008.

C. M. Rodgers, B. D. Neff, and R. Knapp, Androgen-mediated nurturing and aggressive behaviors during paternal care in bluegill sunfish (Lepomis macrochirus), Horm Behav, vol.63, issue.3, pp.454-61, 2013.

C. H. Slater, M. S. Fitzpatrick, and C. B. Schreck, Characterization of an androgen receptor in salmonid lymphocytes: possible link to androgen-induced immunosuppression, Gen Comp Endocrinol, vol.100, issue.2, pp.218-243, 1995.

C. Stuckenholz, L. Lu, P. Thakur, N. Kaminski, and N. Bahary, FACS-assisted microarray profiling implicates novel genes and pathways in zebrafish gastrointestinal tract development, Gastroenterology, vol.137, issue.4, pp.1321-1353, 2009.

N. Y. Storer, R. M. White, A. Uong, E. Price, G. P. Nielsen et al., Zebrafish rhabdomyosarcoma reflects the developmental stage of oncogene expression during myogenesis, Development, vol.140, issue.14, pp.3040-50, 2013.

R. M. White, J. Cech, S. Ratanasirintrawoot, C. Y. Lin, P. B. Rahl et al., DHODH modulates transcriptional elongation in the neural crest and melanoma, Nature, vol.471, issue.7339, pp.518-540, 2011.

G. S. Ducker and J. D. Rabinowitz, One-carbon metabolism in health and disease, Cell Metab, vol.25, issue.1, pp.27-42, 2017.

J. P. Etchegaray and R. Mostoslavsky, Interplay between metabolism and epigenetics: a nuclear adaptation to environmental changes, Mol Cell, vol.62, issue.5, pp.695-711, 2016.

J. Folch, M. Lees, S. Stanley, and G. H. , A simple method for the isolation and purification of total lipides from animal tissues, J Biol Chem, vol.226, issue.1, pp.497-509, 1957.

F. F. Almeida, C. Kristoffersen, G. L. Taranger, and R. W. Schulz, Spermatogenesis in Atlantic cod (Gadus morhua): a novel model of cystic germ cell development, Biol Reprod, vol.78, issue.1, pp.27-34, 2008.

A. Fostier, R. Billard, B. Breton, M. Legendre, and S. Marlot, Plasma 11-oxotestosterone and gonadotropin during the beginning of spermiation in rainbow trout (Salmo gairdneri R.), Gen Comp Endocrinol, vol.46, issue.4, pp.428-462, 1982.
URL : https://hal.archives-ouvertes.fr/hal-01600658

G. Maugars and M. Schmitz, Gene expression profiling during spermatogenesis in early maturing male Atlantic salmon parr testes, Gen Comp Endocrinol, vol.159, issue.2-3, pp.178-87, 2008.

S. G. Jantzen, D. S. Sanderson, V. Schalburg, K. R. Yasuike, M. Marass et al., A 44K microarray dataset of the changing transcriptome in developing Atlantic salmon (Salmo salar L.), BMC Res Notes, vol.4, p.88, 2011.

S. Wang, T. Furmanek, H. Kryvi, C. Krossoy, G. K. Totland et al., Transcriptome sequencing of Atlantic salmon (Salmo salar L.) notochord prior to development of the vertebrae provides clues to regulation of positional fate, chordoblast lineage and mineralisation, BMC Genomics, vol.15, p.141, 2014.

D. Merico, R. Isserlin, O. Stueker, A. Emili, and G. D. Bader, Enrichment map: a network-based method for gene-set enrichment visualization and interpretation, PLoS One, vol.5, issue.11, p.13984, 2010.

P. Shannon, A. Markiel, O. Ozier, N. S. Baliga, J. T. Wang et al., Cytoscape: a software environment for integrated models of biomolecular interaction networks, Genome Res, vol.13, issue.11, pp.2498-504, 2003.

M. Ashburner, C. A. Ball, J. A. Blake, D. Botstein, H. Butler et al., Gene ontology: tool for the unification of biology. The gene ontology consortium, Nat Genet, vol.25, issue.1, pp.25-34, 2000.

D. W. Huang, B. T. Sherman, and R. A. Lempicki, Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources, Nat Protocols, vol.4, issue.1, pp.44-57, 2008.

D. Szklarczyk, A. Franceschini, S. Wyder, K. Forslund, D. Heller et al., STRING v10: protein-protein interaction networks, integrated over the tree of life, Nucleic Acids Res, vol.43, pp.447-52, 2015.

J. Bogerd, M. Blomenrohr, E. Andersson, H. H. Van-der-putten, C. P. Tensen et al., Discrepancy between molecular structure and ligand selectivity of a testicular follicle-stimulating hormone receptor of the African catfish (Clarias gariepinus), Biol Reprod, vol.64, issue.6, pp.1633-1676, 2001.

, Publisher's Note

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