, in Endocrinology | www.frontiersin, 2018.

J. Conlon and D. Larhammar, The evolution of neuroendocrine peptides, General and Comparative Endocrinology, vol.142, issue.1-2, pp.53-62, 2005.
DOI : 10.1016/j.ygcen.2004.11.016

W. Zhou, S. Li, Y. Liu, X. Qi, H. Chen et al., The evolution of tachykinin/tachykinin receptor (TAC/TACR) in vertebrates and molecular identification of the TAC3/TACR3 system in zebrafish (Danio rerio), Molecular and Cellular Endocrinology, vol.361, issue.1-2
DOI : 10.1016/j.mce.2012.04.007

, Mol Cell Endocrinol, vol.361, pp.202-214, 2012.

C. Maggi, The mammalian tachykinin receptors, General Pharmacology: The Vascular System, vol.26, issue.5, pp.911-955, 1995.
DOI : 10.1016/0306-3623(94)00292-U

A. Topaloglu, F. Reimann, M. Guclu, A. Yalin, L. Kotan et al., TAC3 and TACR3 mutations in familial hypogonadotropic hypogonadism reveal a key role for Neurokinin B in the central control of reproduction, Nature Genetics, vol.260, issue.3, pp.354-362, 2009.
DOI : 10.1016/j.phrs.2004.07.002

T. Guran, G. Tolhurst, A. Bereket, N. Rocha, K. Porter et al., Hypogonadotropic Hypogonadism due to a Novel Missense Mutation in the First Extracellular Loop of the Neurokinin B Receptor, The Journal of Clinical Endocrinology & Metabolism, vol.94, issue.10, pp.3633-3642, 2009.
DOI : 10.1210/jc.2009-0551

E. Gianetti, C. Tusset, S. Noel, M. Au, A. Dwyer et al., Mutations Reveal Preferential Activation of Gonadotropin-Releasing Hormone Release by Neurokinin B in Neonatal Life Followed by Reversal in Adulthood, The Journal of Clinical Endocrinology & Metabolism, vol.95, issue.6, pp.2857-67, 2009.
DOI : 10.1210/jc.2009-2320

C. True, S. Alam, K. Cox, Y. Chan, and S. Seminara, Neurokinin B Is Critical for Normal Timing of Sexual Maturation but Dispensable for Adult Reproductive Function in Female Mice, Endocrinology, vol.156, issue.4, pp.1386-97102014, 1210.
DOI : 10.1210/en.2014-1862

J. Yang, C. Caligioni, Y. Chan, and S. Seminara, Uncovering Novel Reproductive Defects in Neurokinin B Receptor Null Mice: Closing the Gap Between Mice and Men, Endocrinology, vol.153, issue.3, pp.1498-508, 1210.
DOI : 10.1210/en.2011-1949

V. Navarro, M. Gottsch, M. Wu, D. García-galiano, S. Hobbs et al., Regulation of NKB Pathways and Their Roles in the Control of Kiss1 Neurons in the Arcuate Nucleus of the Male Mouse, Endocrinology, vol.152, issue.11, pp.4265-752011, 2011.
DOI : 10.1210/en.2011-1143

V. Navarro, M. Bosch, S. León, S. Simavli, C. True et al., The Integrated Hypothalamic Tachykinin-Kisspeptin System as a Central Coordinator for Reproduction, Endocrinology, vol.156, issue.2, pp.627-3710, 1210.
DOI : 10.1210/en.2014-1651

URL : https://academic.oup.com/endo/article-pdf/156/2/627/8991619/endo0627.pdf

H. Billings, J. Connors, S. Altman, S. Hileman, I. Holaskova et al., Neurokinin B Acts via the Neurokinin-3 Receptor in the Retrochiasmatic Area to Stimulate Luteinizing Hormone Secretion in Sheep, Endocrinology, vol.151, issue.8, pp.3836-462010, 2010.
DOI : 10.1210/en.2010-0174

S. Ramaswamy, S. Seminara, A. B. Ciofi, P. Amin, N. Plant et al., ) and Is Colocalized with Kisspeptin in the Arcuate Nucleus, Endocrinology, vol.151, issue.9, pp.4494-5032010, 2010.
DOI : 10.1210/en.2010-0223

URL : https://academic.oup.com/endo/article-pdf/151/9/4494/9014475/endo4494.pdf

K. Skorupskaite, J. George, J. Veldhuis, and R. Anderson, Neurokinin B Regulates Gonadotropin Secretion, Ovarian Follicle Growth, and the Timing of Ovulation in Healthy Women, The Journal of Clinical Endocrinology & Metabolism, vol.97, issue.2, pp.95-104102017, 1210.
DOI : 10.1159/000336376

K. Skorupskaite, J. George, J. Veldhuis, R. Millar, and R. Anderson, Neurokinin 3 receptor antagonism decreases gonadotropin and testosterone secretion in healthy men, Clinical Endocrinology, vol.86, issue.6, pp.748-56, 2017.
DOI : 10.1111/cen.13308

URL : https://repository.up.ac.za/bitstream/2263/62606/1/Skorupskaite_Neurokinin_2017.pdf

V. Navarro, J. Castellano, S. Mcconkey, R. Pineda, F. Ruiz-pino et al., Interactions between kisspeptin and neurokinin B in the control of GnRH secretion in the female rat, American Journal of Physiology-Endocrinology and Metabolism, vol.6, issue.1, pp.202-212, 2010.
DOI : 10.1210/jc.2009-2600

V. Navarro, New Insights into the Control of Pulsatile GnRH Release: The Role of Kiss1/Neurokinin B Neurons, Frontiers in Endocrinology, vol.3, 2012.
DOI : 10.3389/fendo.2012.00048

B. Enright, M. Leach, G. Pelletier, F. Labrie, B. Mcintyre et al., Effects of an antagonist of neurokinin receptors 1, 2 and 3 on reproductive hormones in male beagle dogs, Birth Defects Research Part B: Developmental and Reproductive Toxicology, vol.30, issue.6, pp.517-542, 2010.
DOI : 10.1164/ajrccm/148.6_Pt_1.1628

G. Fraser, H. Hoveyda, I. Clarke, S. Ramaswamy, T. Plant et al., The NK3 Receptor Antagonist ESN364 Interrupts Pulsatile LH Secretion and Moderates Levels of Ovarian Hormones Throughout the Menstrual Cycle, Endocrinology, vol.156, issue.11, pp.4214-252015, 2015.
DOI : 10.1210/en.2015-1409

P. Grachev, R. Millar, O. Byrne, and K. , The Role of Neurokinin B Signalling in Reproductive Neuroendocrinology, Neuroendocrinology, vol.99, issue.1, pp.7-17, 2014.
DOI : 10.1159/000357734

URL : http://www.karger.com/Article/Pdf/357734

C. Fergani and V. Navarro, Expanding the role of tachykinins in the neuroendocrine control of reproduction, Reproduction, vol.106, issue.1, pp.1-14, 2017.
DOI : 10.1007/978-3-211-98811-4_39

M. Lehman, L. Coolen, and R. Goodman, Minireview: Kisspeptin/Neurokinin B/Dynorphin (KNDy) Cells of the Arcuate Nucleus: A Central Node in the Control of Gonadotropin-Releasing Hormone Secretion, Endocrinology, vol.151, issue.8, pp.3479-892010, 2010.
DOI : 10.1210/en.2010-0022

M. Mittelman-smith, S. Krajewski-hall, N. Mcmullen, and N. Rance, Ablation of KNDy Neurons Results in Hypogonadotropic Hypogonadism and Amplifies the Steroid-Induced LH Surge in Female Rats, Endocrinology, vol.157, issue.5, pp.2015-27102015, 1210.
DOI : 10.1210/en.2015-1740

J. Biran, M. Golan, N. Mizrahi, S. Ogawa, I. Parhar et al., ), Endocrinology, vol.155, issue.12, pp.4831-4210, 1210.
DOI : 10.1210/en.2013-2114

X. Qi, W. Zhou, S. Li, Y. Liu, G. Ye et al., Goldfish neurokinin B: Cloning, tissue distribution, and potential role in regulating reproduction, General and Comparative Endocrinology, vol.221, pp.267-77, 2015.
DOI : 10.1016/j.ygcen.2014.10.017

H. Chen, L. Xiao, Y. Liu, S. Li, G. Li et al., Neurokinin B signaling in hermaphroditic species, a study of the orange-spotted grouper ( Epinephelus coioides ), General and Comparative Endocrinology, vol.260, pp.125-160, 2018.
DOI : 10.1016/j.ygcen.2018.01.009

N. Zmora, T. Wong, J. Stubblefield, B. Levavi-sivan, and Y. Zohar, Neurokinin B regulates reproduction via inhibition of kisspeptin in a teleost, the striped bass, Journal of Endocrinology, vol.295, issue.2, pp.159-74, 2017.
DOI : 10.1095/biolreprod.115.131870

S. Ogawa, P. Ramadasan, M. Goschorska, A. Anantharajah, K. Ng et al., Cloning and expression of tachykinins and their association with kisspeptins in the brains of zebrafish, The Journal of Comparative Neurology, vol.90, issue.13, pp.2991-3012, 2012.
DOI : 10.1016/j.nlm.2008.04.013

S. Dufour, E. Burzawa-gerard, L. Belle, N. Sbaihi, M. Vidal et al., Reproductive Endocrinology of the European Eel, Anguilla anguilla, Eel Biology, pp.373-83, 2003.
DOI : 10.1007/978-4-431-65907-5_25

S. Dufour, M. Sebert, F. Weltzien, K. Rousseau, and C. Pasqualini, Neuroendocrine control by dopamine of teleost reproduction, Journal of Fish Biology, vol.84, issue.1, pp.129-60, 2010.
DOI : 10.1007/978-1-4615-3042-8_12

URL : https://hal.archives-ouvertes.fr/hal-00585568

K. Rousseau, A. Lafont, J. Pasquier, G. Maugars, C. Jolly et al., Advances in Eel Reproductive Physiology and Endocrinology, Eel Physiology. Boca Raton, pp.1-43, 2013.
DOI : 10.1201/b15365-2

J. Pasquier, C. Cabau, T. Nguyen, E. Jouanno, D. Severac et al., Gene evolution and gene expression after whole genome duplication in fish: the PhyloFish database, BMC Genomics, vol.40, issue.Web Server issu, pp.368-378, 2016.
DOI : 10.1093/nar/gks576

URL : https://hal.archives-ouvertes.fr/hal-01487023

C. Henkel, E. Burgerhout, D. De-wijze, R. Dirks, Y. Minegishi et al., Primitive Duplicate Hox Clusters in the European Eel's Genome, PLoS ONE, vol.6, issue.4, 2012.
DOI : 10.1371/journal.pone.0032231.s008

URL : https://doi.org/10.1371/journal.pone.0032231

H. Jansen, M. Liem, S. Jong-raadsen, S. Dufour, F. Weltzien et al., Rapid de novo assembly of the European eel genome from nanopore sequencing reads, Sci Rep, vol.7, pp.1-13, 2017.
DOI : 10.1038/s41598-017-07650-6

URL : https://www.nature.com/articles/s41598-017-07650-6.pdf

S. Pavey, M. Laporte, E. Normandeau, J. Gaudin, L. Letourneau et al., ), Molecular Ecology Resources, vol.14, issue.9, pp.806-817, 2017.
DOI : 10.1186/1471-2164-14-604

M. Gouy, S. Guindon, and O. Gascuel, SeaView Version 4: A Multiplatform Graphical User Interface for Sequence Alignment and Phylogenetic Tree Building, Molecular Biology and Evolution, vol.24, issue.8, pp.221-225, 2010.
DOI : 10.1093/molbev/msm092

URL : https://hal.archives-ouvertes.fr/lirmm-00705187

A. Stamatakis, RAxML version 8: a tool for phylogenetic analysis and post-analysis of large phylogenies, Bioinformatics, vol.57, issue.9, pp.1312-1315, 2014.
DOI : 10.1080/10635150802429642

URL : https://academic.oup.com/bioinformatics/article-pdf/30/9/1312/17345185/btu033.pdf

T. Petersen, S. Brunak, G. Von-heijne, and H. Nielsen, SignalP 4.0: discriminating signal peptides from transmembrane regions, Nature Methods, vol.6, issue.10, pp.785-791, 2011.
DOI : 10.1016/0005-2795(75)90109-9

B. Southey, A. Amare, T. Zimmerman, S. Rodriguez-zas, and J. Sweedler, NeuroPred: a tool to predict cleavage sites in neuropeptide precursors and provide the masses of the resulting peptides, Nucleic Acids Research, vol.34, issue.Web Server, pp.267-72, 2006.
DOI : 10.1093/nar/gkl161

J. Pasquier, A. Lafont, F. Denis, B. Lefranc, C. Dubessy et al., Table_1.DOCX, Frontiers in Endocrinology, vol.211, 2018.
DOI : 10.3389/fendo.2017.00353.s001

J. Yang, R. Yan, A. Roy, D. Xu, J. Poisson et al., The I-TASSER Suite: protein structure and function prediction, Nature Methods, vol.12, issue.1, pp.7-8, 2014.
DOI : 10.1002/prot.24341

URL : http://europepmc.org/articles/pmc4428668?pdf=render

M. Montero, L. Belle, N. Vidal, B. Dufour, and S. , Primary Cultures of Dispersed Pituitary Cells from Estradiol-Pretreated Female Silver Eels (Anguilla anguillaL.): Immunocytochemical Characterization of Gonadotropic Cells and Stimulation of Gonadotropin Release, General and Comparative Endocrinology, vol.104, issue.1, pp.103-150146, 1996.
DOI : 10.1006/gcen.1996.0146

J. Pasquier, A. Lafont, J. Leprince, H. Vaudry, K. Rousseau et al., First evidence for a direct inhibitory effect of kisspeptins on LH expression in the eel, Anguilla anguilla, General and Comparative Endocrinology, vol.173, issue.1, pp.216-241, 2011.
DOI : 10.1016/j.ygcen.2011.05.019

S. Aroua, F. Weltzien, L. Belle, N. Dufour, and S. , Development of real-time RT-PCR assays for eel gonadotropins and their application to the comparison of in vivo and in vitro effects of sex steroids, General and Comparative Endocrinology, vol.153, issue.1-3, pp.333-376, 2007.
DOI : 10.1016/j.ygcen.2007.02.027

URL : https://hal.archives-ouvertes.fr/hal-00145334

G. Maugars, S. Dufour, J. Cohen-tannoudji, and B. Quérat, Multiple Thyrotropin ??-Subunit and Thyrotropin Receptor-Related Genes Arose during Vertebrate Evolution, PLoS ONE, vol.3, issue.2, 2014.
DOI : 10.1371/journal.pone.0111361.s008

URL : https://hal.archives-ouvertes.fr/hal-01366759

D. Peñaranda, I. Mazzeo, J. Hildahl, V. Gallego, R. Nourizadeh-lillabadi et al., Molecular characterization of three GnRH receptor paralogs in the European eel, Anguilla anguilla: Tissue-distribution and changes in transcript abundance during artificially induced sexual development, Molecular and Cellular Endocrinology, vol.369, issue.1-2, pp.1-14, 2013.
DOI : 10.1016/j.mce.2013.01.025

T. Almeida, J. Rojo, P. Nieto, F. Pinto, M. Hernandez et al., Tachykinins and Tachykinin Receptors: Structure and Activity Relationships, Current Medicinal Chemistry, vol.11, issue.15, pp.2045-81, 2004.
DOI : 10.2174/0929867043364748

N. Page, D. Morrish, and N. Weston-bell, Differential mRNA splicing and precursor processing of neurokinin B in neuroendocrine tissues, Peptides, vol.30, issue.8, pp.1508-1521, 2009.
DOI : 10.1016/j.peptides.2009.04.023

H. Satake, M. Ogasawara, T. Kawada, K. Masuda, M. Aoyama et al., Journal of Biological Chemistry, vol.43, issue.51, pp.53798-805, 2004.
DOI : 10.1210/endo-121-4-1555

A. Mantha, I. Chandrashekar, N. Baquer, and S. Cowsik, Three Dimensional Structure of Mammalian Tachykinin Peptide Neurokinin B Bound to Lipid Micelles, Journal of Biomolecular Structure and Dynamics, vol.42, issue.2, pp.137-184, 2004.
DOI : 10.1016/0024-3205(88)90246-9

T. Whitehead, S. Mcnair, C. Hadden, J. Young, and R. Hicks, Journal of Medicinal Chemistry, vol.41, issue.9, pp.1497-506, 1998.
DOI : 10.1021/jm970789x

N. Rance, S. Krajewski, M. Smith, M. Cholanian, and P. Dacks, Neurokinin B and the hypothalamic regulation of reproduction, Brain Research, vol.1364, pp.116-144, 2010.
DOI : 10.1016/j.brainres.2010.08.059

X. Qi, M. Salem, W. Zhou, M. Sato-shimizu, G. Ye et al., Neurokinin B Exerts Direct Effects on the Ovary to Stimulate Estradiol Production, Endocrinology, vol.157, issue.9, pp.3355-65102016, 1210.
DOI : 10.1210/en.2016-1354

A. Lecci, A. Capriati, M. Altamura, and C. Maggi, Tachykinins and tachykinin receptors in the gut, with special reference to NK2 receptors in human, Autonomic Neuroscience, vol.126, issue.127, pp.126-127232, 2006.
DOI : 10.1016/j.autneu.2006.02.014

T. Mijiddorj, H. Kanasaki, I. Purwana, A. Oride, U. Sukhbaatar et al., Role of Neurokinin B and Dynorphin A in pituitary gonadotroph and somatolactotroph cell lines, Endocrine Journal, vol.59, issue.7, pp.631-671, 2012.
DOI : 10.1507/endocrj.EJ11-0401

A. Sahu and P. Kalra, Effects of tachykinins on luteinizing hormone release in female rats: potent inhibitory action of neuropeptide K, Endocrinology, vol.130, 1992.

M. Corander, B. Challis, E. Thompson, Z. Jovanovic, L. Tung et al., The Effects of Neurokinin B upon Gonadotrophin Release in Male Rodents, Journal of Neuroendocrinology, vol.50, issue.3, pp.181-7237, 1159.
DOI : 10.1111/j.1365-2826.2009.01951.x

C. Nestor, A. Briscoe, S. Davis, M. Valent, R. Goodman et al., Evidence of a Role for Kisspeptin and Neurokinin B in Puberty of Female Sheep, Endocrinology, vol.153, issue.6, pp.2756-6510, 1210.
DOI : 10.1210/en.2011-2009

K. Sakamoto, K. Murata, Y. Wakabayashi, K. Yayou, S. Ohkura et al., Central Administration of Neurokinin B Activates Kisspeptin/NKB Neurons in the Arcuate Nucleus and Stimulates Luteinizing Hormone Secretion in Ewes During the Non-Breeding Season, Journal of Reproduction and Development, vol.58, issue.6, pp.700-706, 2012.
DOI : 10.1262/jrd.2011-038

R. Goodman, S. Hileman, C. Nestor, K. Porter, J. Connors et al., Kisspeptin, Neurokinin B, and Dynorphin Act in the Arcuate Nucleus to Control Activity of the GnRH Pulse Generator in Ewes, Endocrinology, vol.154, issue.11, pp.4259-692013, 2013.
DOI : 10.1210/en.2013-1331

T. Sandoval-guzmán and N. Rance, Central injection of senktide, an NK 3 receptor agonist, or neuropeptide Y inhibits LH secretion and induces different patterns of Fos expression in the rat hypothalamus, Brain Research, vol.1026, issue.2, pp.307-319, 2004.
DOI : 10.1016/j.brainres.2004.08.026

V. Navarro, M. Gottsch, C. Chavkin, H. Okamura, D. Clifton et al., Regulation of Gonadotropin-Releasing Hormone Secretion by Kisspeptin/Dynorphin/Neurokinin B Neurons in the Arcuate Nucleus of the Mouse, Journal of Neuroscience, vol.29, issue.38, pp.11859-66, 2009.
DOI : 10.1523/JNEUROSCI.1569-09.2009

Y. Wakabayashi, T. Nakada, K. Murata, S. Ohkura, K. Mogi et al., Neurokinin B and Dynorphin A in Kisspeptin Neurons of the Arcuate Nucleus Participate in Generation of Periodic Oscillation of Neural Activity Driving Pulsatile Gonadotropin-Releasing Hormone Secretion in the Goat, Journal of Neuroscience, vol.30, issue.8, pp.3124-3156, 2010.
DOI : 10.1523/JNEUROSCI.5848-09.2010

J. Henriksen, T. Saermark, H. Vilhardt, and S. Mau, Tachykinins Induce Secretion of Prolactin from Perifused Rat Anterior Pituitary Cells by Interactions with Two Different Binding Sites, Journal of Receptors and Signal Transduction, vol.120, issue.1-4, pp.529-570, 1995.
DOI : 10.1210/endo-120-1-280

C. Maggi, Principles of tachykininergic co-transmission in the peripheral and enteric nervous system, Regulatory Peptides, vol.93, issue.1-3, pp.53-64, 2000.
DOI : 10.1016/S0167-0115(00)00177-4

J. Pasquier, A. Lafont, S. Jeng, M. Morini, R. Dirks et al., Multiple Kisspeptin Receptors in Early Osteichthyans Provide New Insights into the Evolution of This Receptor Family, PLoS ONE, vol.149, issue.11, 2012.
DOI : 10.1371/journal.pone.0048931.s009

C. Pasqualini, F. Weltzien, B. Vidal, S. Baloche, C. Rouget et al., Two Distinct Dopamine D2 Receptor Genes in the European Eel: Molecular Characterization, Tissue-Specific Transcription, and Regulation by Sex Steroids, Endocrinology, vol.150, issue.3, pp.1377-922008, 2009.
DOI : 10.1210/en.2008-0578

URL : https://hal.archives-ouvertes.fr/hal-00374336

C. Jolly, K. Rousseau, L. Prézeau, C. Vol, J. Tomkiewicz et al., Functional characterisation of eel dopamine D2 receptors and involvement in the direct inhibition of pituitary gonadotrophins, J Neuroendocrinol