, L'intervention de l'enzyme 11?-hydroxylase de type 1 (CYP11B1) sur le 11-déoxycortisol conduit à la production de cortisol, 2004.

N. Gallo-payet, Adrenal and extra-adrenal functions of ACTH, J. Mol. Endocrinol, 2016.

S. Espiard, M. J. Knape, K. Bathon, G. Assié, M. Rizk-rabin et al., Activating PRKACB somatic mutation in cortisol-producing adenomas, JCI Insight, vol.3, issue.8, 2018.

D. Calebiro, G. D. Dalmazi, K. Bathon, C. L. Ronchi, and F. Beuschlein, cAMP signaling in cortisol-producing adrenal adenoma, Eur J Endocrinol, vol.173, issue.4, pp.99-106, 2015.

Z. Bram, E. Louiset, B. Ragazzon, S. Renouf, J. Wils et al., PKA regulatory subunit 1A inactivating mutation induces serotonin signaling in primary pigmented nodular adrenal disease, JCI Insight, vol.1, issue.15, 2016.
URL : https://hal.archives-ouvertes.fr/hal-02108040

E. Louiset, C. Duparc, J. Young, S. Renouf, T. Nomigni et al., Intraadrenal corticotropin in bilateral macronodular adrenal hyperplasia, N. Engl. J. Med, vol.369, issue.22, pp.2115-2125, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01908272

E. Louiset, V. Contesse, L. Groussin, D. Cartier, C. Duparc et al., Expression of serotonin7 receptor and coupling of ectopic receptors to protein kinase A and ionic currents in adrenocorticotropin-independent macronodular adrenal hyperplasia causing Cushing's syndrome, J. Clin. Endocrinol. Metab, vol.91, issue.11, pp.4578-4586, 2006.
URL : https://hal.archives-ouvertes.fr/hal-01928836

D. C. Lefebvre, G. Prévost, M. C. Zennaro, J. Bertherat, and E. Louiset, Paracrine control of steroidogenesis by serotonin in adrenocortical neoplasms, Mol. Cell. Endocrinol, vol.408, pp.198-204, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01931135

M. St-jean, N. E. Ghorayeb, I. Bourdeau, and A. Lacroix, Aberrant G-protein coupled hormone receptor in adrenal diseases, Best Pract. Res. Clin. Endocrinol. Metab, vol.32, issue.2, pp.165-187, 2018.

H. Lefebvre, V. Contesse, C. Delarue, M. Feuilloley, F. Hery et al., Serotonin-induced stimulation of cortisol secretion from human adrenocortical tissue is mediated through activation of a serotonin4 receptor subtype, Neuroscience, vol.47, issue.4, pp.999-1007, 1992.

H. Lefebvre, V. Contesse, C. Delarue, A. Legrand, J. M. Kuhn et al., The serotonin-4 receptor agonist cisapride and angiotensin-II exert additive effects on aldosterone secretion in normal man, J Clin Endocrinol Metab, vol.80, issue.2, pp.504-507, 1995.

H. Lefebvre, P. Compagnon, V. Contesse, C. Delarue, C. Thuillez et al., Production and metabolism of serotonin (5-HT) by the human adrenal cortex: paracrine stimulation of aldosterone secretion by 5-HT, J. Clin. Endocrinol. Metab, vol.86, issue.10, pp.5001-5007, 2001.

C. A. Stratakis, J. A. Carney, L. S. Kirschner, H. S. Willenberg, S. Brauer et al., Synaptophysin immunoreactivity in primary pigmented nodular adrenocortical disease: neuroendocrine properties of tumors associated with Carney complex, J. Clin. Endocrinol. Metab, vol.84, issue.3, pp.1122-1128, 1999.

Y. T. Chang, G. Mues, and K. Hyland, Alternative splicing in the coding region of human aromatic L-amino acid decarboxylase mRNA, Neurosci. Lett, vol.202, issue.3, pp.157-160, 1996.

C. Drelon, A. Berthon, I. Sahut-barnola, M. Mathieu, T. Dumontet et al., PKA inhibits WNT signalling in adrenal cortex zonation and prevents malignant tumour development, Nat Commun, vol.7, 2016.
URL : https://hal.archives-ouvertes.fr/hal-02108042

Y. M. Ulrich-lai, H. F. Figueiredo, M. M. Ostrander, D. C. Choi, W. C. Engeland et al., Chronic stress induces adrenal hyperplasia and hypertrophy in a subregion-specific manner, American Journal of Physiology-Endocrinology and Metabolism, vol.291, issue.5, pp.965-973, 2006.

I. P. Cavalcante, M. Nishi, M. Zerbini, M. Q. Almeida, V. B. Brondani et al., The role of ARMC5 in human cell cultures from nodules of primary macronodular adrenocortical hyperplasia (PMAH), Mol. Cell. Endocrinol, vol.460, pp.36-46, 2018.

V. R. Falkenberg and M. S. Rajeevan, Identification of a potential molecular link between the glucocorticoid and serotonergic signaling systems, J. Mol. Neurosci, vol.41, issue.2, pp.322-327, 2010.

C. Graveleau, H. J. Paust, D. Schmidt-grimminger, and A. K. Mukhopadhyay, Presence of a 5-HT7 receptor positively coupled to adenylate cyclase activation in human granulosa-lutein cells, J. Clin. Endocrinol. Metab, vol.85, issue.3, pp.1277-1286, 2000.

K. A. Krobert, T. Bach, T. Syversveen, A. M. Kvingedal, and F. O. Levy, The cloned human 5-HT7 receptor splice variants: a comparative characterization of their pharmacology, function and distribution, Naunyn Schmiedebergs Arch. Pharmacol, vol.363, issue.6, pp.620-632, 2001.

C. L. Ronchi, D. Dalmazi, G. Faillot, S. Sbiera, S. Assié et al., Genetic Landscape of Sporadic Unilateral Adrenocortical Adenomas Without PRKACA p.Leu206Arg Mutation, J. Clin. Endocrinol. Metab, vol.101, issue.9, pp.3526-3538, 2016.

L. Tsai and J. A. Beavo, The roles of cyclic nucleotide phosphodiesterases (PDEs) in steroidogenesis, Curr Opin Pharmacol, vol.11, issue.6, pp.670-675, 2011.

J. Meffre, S. Chaumont-dubel, M. La-cour, C. Loiseau, F. Watson et al., 5-HT(6) receptor recruitment of mTOR as a mechanism for perturbed cognition in schizophrenia, EMBO Mol Med, vol.4, issue.10, pp.1043-1056, 2012.

C. De-joussineau, I. Sahut-barnola, F. Tissier, T. Dumontet, C. Drelon et al., mTOR pathway is activated by PKA in adrenocortical cells and participates in vivo to apoptosis resistance in primary pigmented nodular adrenocortical disease (PPNAD), Hum. Mol. Genet, vol.23, issue.20, pp.5418-5428, 2014.
URL : https://hal.archives-ouvertes.fr/hal-02108057

R. Kohen, L. A. Fashingbauer, D. E. Heidmann, C. R. Guthrie, and M. W. Hamblin, Cloning of the mouse 5-HT6 serotonin receptor and mutagenesis studies of the third cytoplasmic loop, Brain Res. Mol. Brain Res, vol.90, issue.2, pp.110-117, 2001.

K. A. Krobert and F. O. Levy, The human 5-HT7 serotonin receptor splice variants: constitutive activity and inverse agonist effects, Br. J. Pharmacol, vol.135, issue.6, pp.1563-1571, 2002.

R. E. Kuc, C. A. Parker, A. D. Gee, Y. P. Bao, M. J. Brown et al., 5-HT4 receptor distribution in the human adrenal revealed by 3H-GR113808 A 5-HT4 receptor antagonist, Proc. Brit. Parmacol. Soc, vol.3, p.68, 2005.

B. B. García-iglesias, M. E. Mendoza-garrido, G. Gutiérrez-ospina, C. Rangel-barajas, M. Noyola-díaz et al., Sensitization of restraint-induced corticosterone secretion after chronic restraint in rats: involvement of 5-HT? receptors, Neuropharmacology, vol.71, pp.216-227, 2013.

H. Kim, Y. Toyofuku, F. C. Lynn, E. Chak, T. Uchida et al., Serotonin regulates pancreatic beta cell mass during pregnancy, Nat. Med, vol.16, issue.7, pp.804-808, 2010.

M. Michalowska, B. Znorko, T. Kaminski, E. Oksztulska-kolanek, and D. Pawlak, New insights into tryptophan and its metabolites in the regulation of bone metabolism, J. Physiol. Pharmacol, vol.66, issue.6, pp.779-791, 2015.

M. Matsuda, T. Imaoka, A. J. Vomachka, G. A. Gudelsky, Z. Hou et al., Serotonin regulates mammary gland development via an autocrine-paracrine loop, Dev. Cell, vol.6, issue.2, pp.193-203, 2004.

S. C. Wyler, C. C. Lord, S. Lee, J. K. Elmquist, and C. Liu, Serotonergic Control of Metabolic Homeostasis, Front Cell Neurosci, vol.11, p.277, 2017.

A. Lacroix, I. Bourdeau, A. Lampron, T. L. Mazzuco, J. Tremblay et al., Aberrant G-protein coupled receptor expression in relation to adrenocortical overfunction, Clin. Endocrinol. (Oxf.), 2010.

A. Lecoq, C. A. Stratakis, S. Viengchareun, R. Chaligné, L. Tosca et al., Adrenal GIPR expression and chromosome 19q13 microduplications in GIP-dependent Cushing's syndrome, JCI Insight, vol.2, issue.18, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01904330

F. Beuschlein, M. Fassnacht, G. Assié, D. Calebiro, C. A. Stratakis et al., Constitutive activation of PKA catalytic
URL : https://hal.archives-ouvertes.fr/inserm-01068048

. Med, , vol.370, pp.1019-1028, 2014.

F. Hannah-shmouni, F. R. Faucz, and C. A. Stratakis, Alterations of Phosphodiesterases in Adrenocortical Tumors, Front. Endocrinol, vol.7, p.111, 2016.

Z. Bram, E. Louiset, B. Ragazzon, S. Renouf, J. Wils et al., PKA regulatory subunit 1A inactivating mutation induces serotonin signaling in primary pigmented nodular adrenal disease, J. Clin. Investig. Insight, vol.1, issue.15, p.87958, 2016.
URL : https://hal.archives-ouvertes.fr/hal-02108040

C. Gao, Y. Wang, R. Broaddus, L. Sun, F. Xue et al., Exon 3 mutations of CTNNB1 drive tumorigenesis: a review, Oncotarget, vol.9, issue.4, pp.5492-5508, 2018.

P. J. Morin, A. B. Sparks, V. Korinek, N. Barker, H. Clevers et al., Activation of beta-catenin-Tcf signaling in colon cancer by mutations in beta-catenin or APC, Science, vol.275, issue.5307, pp.1787-1790, 1997.

P. Galiatsatos and W. D. Foulkes, Familial adenomatous polyposis, Am. J. Gastroenterol, vol.101, issue.2, pp.385-398, 2006.

S. Gaujoux, S. Pinson, A. Gimenez-roqueplo, L. Amar, B. Ragazzon et al., Inactivation of the APC gene is constant in adrenocortical tumors from patients with familial adenomatous polyposis but not frequent in sporadic adrenocortical cancers, Clin. Cancer Res, vol.16, issue.21, pp.5133-5141, 2010.

S. Faillot and G. Assie, Endocrine tumours: The genomics of adrenocortical tumors
URL : https://hal.archives-ouvertes.fr/inserm-01291562

, J. Endocrinol, vol.174, issue.6, pp.249-265, 2016.

A. Teo, S. Garg, L. H. Shaikh, J. Zhou, K. Frankl et al., Pregnancy, Primary Aldosteronism, and Adrenal CTNNB1 Mutations, N. Engl. J. Med, vol.373, issue.15, pp.1429-1436, 2015.

V. Wu, S. Wang, S. Chueh, Y. Huang, K. Lin et al., The prevalence of CTNNB1 mutations in primary aldosteronism and consequences for clinical outcomes, Sci. Rep, vol.7, p.39121, 2017.

N. Gagnon, K. Caceres, G. Corbeil, E. Ghorayeb, N. Ludwig et al., Genetic Characterization of GnRH/LH-Responsive Primary Aldosteronism

, Clin. Endocrinol. Metab, 2018.

M. Fassnacht, W. Arlt, I. Bancos, H. Dralle, J. Newell-price et al., Management of adrenal incidentalomas: European Society of Endocrinology Clinical Practice Guideline in collaboration with the European Network for the Study of Adrenal Tumors, Eur. J. Endocrinol, vol.175, issue.2, pp.1-34, 2016.

T. Nomigni, M. Ouzounian, S. Benoit, A. Vadrot, J. Tissier et al., Steroidogenic enzyme profile in an androgen-secreting adrenocortical oncocytoma associated with hirsutism, Endocr. Connect, vol.4, issue.2, pp.117-127, 2015.

H. Lefebvre, V. Contesse, C. Delarue, M. Feuilloley, F. Hery et al., Serotonin-induced stimulation of cortisol secretion from human adrenocortical tissue is mediated through activation of a serotonin4 receptor subtype, Neuroscience, vol.47, issue.4, pp.999-1007, 1992.

K. Orford, C. Crockett, J. P. Jensen, A. M. Weissman, and S. W. Byers, Serine phosphorylation-regulated ubiquitination and degradation of beta-catenin, J. Biol. Chem, vol.272, issue.40, pp.24735-24738, 1997.

A. Berthon, I. Sahut-barnola, S. Lambert-langlais, C. De-joussineau, C. Damonsoubeyrand et al., Constitutive beta-catenin activation induces adrenal hyperplasia and promotes adrenal cancer development, Hum. Mol. Genet, vol.19, issue.8, pp.1561-1576, 2010.

E. Louiset, C. Duparc, S. Lenglet, C. E. Gomez-sanchez, and H. Lefebvre, Role of cAMP/PKA pathway and T-type calcium channels in the mechanism of action of serotonin in human adrenocortical cells, Mol. Cell. Endocrinol, vol.441, pp.99-107, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01931437

H. Lefebvre, C. Duparc, G. Prévost, M. Zennaro, J. Bertherat et al., Paracrine control of steroidogenesis by serotonin in adrenocortical neoplasms, Mol Cell Endocrinol, vol.408, pp.198-204, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01931135

N. C. Law, J. Weck, B. Kyriss, J. H. Nilson, and M. Hunzicker-dunn, Lhcgr expression in granulosa cells: roles for PKA-phosphorylated ?-catenin, TCF3, and FOXO1, Mol. Endocrinol, vol.27, issue.8, pp.1295-1310, 2013.

S. Vuorenoja, A. Rivero-muller, S. Kiiveri, M. Bielinska, M. Heikinheimo et al., Adrenocortical tumorigenesis, luteinizing hormone receptor and transcription factors GATA-4 and GATA-6, Mol. Cell. Endocrinol, vol.269, issue.1-2, pp.38-45, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00531903

K. Omata, S. K. Anand, D. H. Hovelson, C. Liu, Y. Yamazaki et al., Aldosterone-Producing Cell Clusters Frequently Harbor Somatic Mutations and Accumulate With Age in Normal Adrenals, J Endocr Soc, vol.1, issue.7, pp.787-799, 2017.

K. Nishimoto, K. Nakagawa, D. Li, T. Kosaka, M. Oya et al., Adrenocortical zonation in humans under normal and pathological conditions, J. Clin. Endocrinol. Metab, vol.95, issue.5, pp.2296-2305, 2010.

I. Sahut-barnola, C. Joussineau, . De, P. Val, S. Lambert-langlais et al., Cushing's Syndrome and Fetal Features Resurgence in Adrenal Cortex-Specific Prkar1a Knockout Mice, PLOS Genetics, vol.6, issue.6, p.1000980, 2010.

C. Drelon, A. Berthon, I. Sahut-barnola, M. Mathieu, T. Dumontet et al., PKA inhibits WNT signalling in adrenal cortex zonation and prevents malignant tumour development, Nat Commun, vol.7, 2016.
URL : https://hal.archives-ouvertes.fr/hal-02108042

F. Bonnet-serrano and J. Bertherat, Genetics of tumors of the adrenal cortex, Endocr. Relat. Cancer, vol.25, issue.3, pp.131-152, 2018.

Y. Xing, C. R. Parker, M. Edwards, and W. E. Rainey, Adrenocorticotrophic hormone is a potent regulator of gene expression in human adrenal cells, J Mol Endocrinol, vol.45, issue.1, pp.59-68, 2010.

A. J. Clark and A. Weber, Adrenocorticotropin insensitivity syndromes, Endocr. Rev, vol.19, issue.6, pp.828-843, 1998.

Y. M. Ulrich-lai, H. F. Figueiredo, M. M. Ostrander, D. C. Choi, W. C. Engeland et al., Chronic stress induces adrenal hyperplasia and hypertrophy in a subregion-specific manner, American Journal of Physiology-Endocrinology and Metabolism, vol.291, issue.5, pp.965-973, 2006.

A. M. Neville and A. M. Mackay, The structure of the human adrenal cortex in health and disease, Clinics in Endocrinology and Metabolism, vol.1, issue.2, pp.361-395, 1972.

D. G. Romero, L. L. Yanes, A. F. De-rodriguez, M. W. Plonczynski, B. L. Welsh et al., Disabled-2 is expressed in adrenal zona glomerulosa and is involved in aldosterone secretion, Endocrinology, vol.148, issue.6, pp.2644-2652, 2007.

K. Nishimoto, R. Harris, W. E. Rainey, and T. Seki, Sodium deficiency regulates rat adrenal zona glomerulosa gene expression, Endocrinology, vol.155, issue.4, pp.1363-1372, 2014.

A. Naccache, E. Louiset, C. Duparc, A. Laquerrière, S. Patrier et al., Temporal and spatial distribution of mast cells and steroidogenic enzymes in the human fetal adrenal, Mol. Cell. Endocrinol, vol.434, pp.69-80, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01931413

R. W. Hunter, J. R. Ivy, and M. A. Bailey, Glucocorticoids and renal Na+ transport: implications for hypertension and salt sensitivity, J. Physiol. (Lond.), vol.592, issue.8, pp.1731-1744, 2014.

R. I. Menzies, X. Zhao, L. J. Mullins, J. J. Mullins, C. Cairns et al., Transcription controls growth, cell kinetics and cholesterol supply to sustain ACTH responses, Endocr Connect, vol.6, issue.7, pp.446-457, 2017.

. Références,

S. Acton, A. Rigotti, K. T. Landschulz, S. Xu, H. H. Hobbs et al., Identification of Scavenger Receptor SR-BI as a High Density Lipoprotein Receptor, Science, vol.271, pp.518-520, 1996.

F. Aguilar, M. Lo, B. Claustrat, J. M. Saez, J. Sassard et al., Hypersensitivity of the adrenal cortex to trophic and secretory effects of angiotensin II in Lyon genetically-hypertensive rats, Hypertens Dallas Tex, vol.43, pp.87-93, 1979.

A. H. Ahmed, M. Calvird, R. D. Gordon, P. J. Taylor, G. Ward et al., Effects of two selective serotonin reuptake inhibitor antidepressants, sertraline and escitalopram, on aldosterone/renin ratio in normotensive depressed male patients, J Clin Endocrinol Metab, vol.96, pp.1039-1045, 2011.

M. Aiba, H. Iri, H. Suzuki, K. Kageyama, T. Kawai et al., Numerous mast cells in an 11-deoxycorticosterone-producing adrenocortical tumor. Histologic evaluation of benignancy and comparison with mast cell distribution in adrenal glands and neoplastic counterparts of 67 surgical specimens, Arch Pathol Lab Med, vol.109, pp.357-360, 1985.

T. Åkerström, R. Maharjan, S. Willenberg, H. Cupisti, K. Ip et al., Activating mutations in CTNNB1 in aldosterone producing adenomas, 2016.

A. Adriana, T. Marily, and R. Martin, Genetics of Cushing's disease, Clin Endocrinol (Oxf), vol.88, pp.3-12, 2017.

N. M. Albiger, P. Sartorato, B. Mariniello, M. Iacobone, I. Finco et al., A case of primary aldosteronism in pregnancy: do LH and GNRH receptors have a potential role in regulating aldosterone secretion?, Eur J Endocrinol, vol.164, pp.405-412, 2011.

T. B. Aldrich, A preliminary report on the active principle of the suprarenal gland, Am J PhysiolLeg Content, vol.5, pp.457-461, 1901.

E. A. Al-dujaili, M. Boscaro, and C. R. Edwards, An in vitro stimulatory effect of indoleamines on aldosterone biosynthesis in the rat, J Steroid Biochem, vol.17, pp.351-355, 1982.

G. A. Alencar, A. M. Lerario, M. Y. Nishi, P. Mariani-bm-de, M. Q. Almeida et al., ARMC5 mutations are a frequent cause of primary macronodular adrenal Hyperplasia, J Clin Endocrinol Metab, vol.99, pp.1501-1509, 2014.

M. Q. Almeida, M. F. Azevedo, P. Xekouki, E. I. Bimpaki, A. Horvath et al., Activation of Cyclic AMP Signaling Leads to Different Pathway Alterations in Lesions of the Adrenal Cortex Caused by Germline PRKAR1A Defects versus Those due to Somatic GNAS Mutations, J Clin Endocrinol Metab, vol.97, pp.687-693, 2012.

M. Q. Almeida and C. A. Stratakis, Carney complex and other conditions associated with micronodular adrenal hyperplasias, Best Pract Res Clin Endocrinol Metab, vol.24, pp.907-914, 2010.

A. H. Amin, T. B. Crawford, and J. H. Gaddum, The distribution of substance P and 5-hydroxytryptamine in the central nervous system of the dog, J Physiol, vol.126, pp.596-618, 1954.

K. W. Andressen, A. H. Ulsund, K. A. Krobert, M. J. Lohse, M. Bünemann et al., Related GPCRs couple differently to Gs: preassociation between G protein and 5-HT7 serotonin receptor reveals movement of G?s upon receptor activation, FASEB J Off Publ Fed Am Soc Exp Biol, vol.32, pp.1059-1069, 2018.

F. Arakane, S. R. King, Y. Du, C. B. Kallen, L. P. Walsh et al., Phosphorylation of Steroidogenic Acute Regulatory Protein (StAR) Modulates Its Steroidogenic Activity, J Biol Chem, vol.272, pp.32656-32662, 1997.

W. Arlt and P. M. Stewart, Adrenal Corticosteroid Biosynthesis, Metabolism, and Action, Endocrinol Metab Clin North Am, vol.34, pp.293-313, 2005.

J. Arnold, Ein beitrag zu der feineren structur und dem chemismus der nebennieren, Virchows Arch Pathol Anat Physiol, vol.39, pp.64-117, 1866.

G. Assié, R. Libé, S. Espiard, M. Rizk-rabin, A. Guimier et al., ARMC5 mutations in macronodular adrenal hyperplasia with Cushing's syndrome, N Engl J Med, vol.369, pp.2105-2114, 2013.

G. Assie, E. Louiset, N. Sturm, F. René-corail, L. Groussin et al., Systematic analysis of G protein-coupled receptor gene expression in adrenocorticotropin-independent macronodular adrenocortical hyperplasia identifies novel targets for pharmacological control of adrenal Cushing's syndrome, J Clin Endocrinol Metab, vol.95, pp.253-262, 2010.

S. Azhar and R. E. , Scavenger receptor class BI and selective cholesteryl ester uptake: partners in the regulation of steroidogenesis, Mol Cell Endocrinol, vol.195, pp.1-26, 2002.

C. Ballav, A. Naziat, R. Mihai, N. Karavitaki, O. Ansorge et al., Mini-review: pheochromocytomas causing the ectopic ACTH syndrome, Endocrine, vol.42, pp.69-73, 2012.

R. Bandiera, V. Vidal, F. J. Motamedi, M. Clarkson, I. Sahut-barnola et al., WT1 maintains adrenal-gonadal primordium identity and marks a population of AGP-like progenitors within the adrenal gland, Dev Cell, vol.27, pp.5-18, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00875587

C. W. Bardin, C. L. Chen, P. L. Morris, I. Gerendai, C. Boitani et al., Proopiomelanocortin-derived peptides in testis, ovary, and tissues of reproduction, Recent Prog Horm Res, vol.43, pp.1-28, 1987.

A. Belgorosky, G. Guercio, C. Pepe, N. Saraco, and R. Ma, Genetic and Clinical Spectrum of Aromatase Deficiency in Infancy, Childhood and Adolescence, Horm Res Paediatr, vol.72, pp.321-330, 2009.

S. Ben-david, N. Zuckerman-levin, M. Epelman, Z. Shen-orr, M. Levin et al., Parturition Itself Is the Basis for Fetal Adrenal Involution, J Clin Endocrinol Metab, vol.92, pp.93-97, 2007.

M. Berger, J. A. Gray, and R. Bl, The expanded biology of serotonin, Annu Rev Med, vol.60, pp.355-366, 2009.

J. Bertherat, V. Contesse, E. Louiset, G. Barrande, C. Duparc et al., In vivo and in vitro screening for illegitimate receptors in adrenocorticotropin-independent macronodular adrenal hyperplasia causing Cushing's syndrome: identification of two cases of gonadotropin/gastric inhibitory polypeptide-dependent hypercortisolism, J Clin Endocrinol Metab, vol.90, pp.1302-1310, 2005.
URL : https://hal.archives-ouvertes.fr/hal-01928788

J. Bertherat, L. Groussin, F. Sandrini, L. Matyakhina, T. Bei et al., Molecular and Functional Analysis of PRKAR1A and its Locus (17q22-24) in Sporadic Adrenocortical Tumors: 17q Losses, Somatic Mutations, and Protein Kinase A Expression and Activity, Cancer Res, vol.63, pp.5308-5319, 2003.

J. Bertherat, A. Horvath, L. Groussin, S. Grabar, S. Boikos et al., Mutations in regulatory subunit type 1A of cyclic adenosine 5'-monophosphate-dependent protein kinase (PRKAR1A): phenotype analysis in 353 patients and 80 different genotypes, J Clin Endocrinol Metab, vol.94, pp.2085-2091, 2009.

A. Berthon, C. Drelon, B. Ragazzon, S. Boulkroun, F. Tissier et al., WNT/?-catenin signalling is activated in aldosterone-producing adenomas and controls aldosterone production, Hum Mol Genet, vol.23, pp.889-905, 2014.
URL : https://hal.archives-ouvertes.fr/hal-02108060

B. Annabel, F. Faucz, J. Bertherat, and C. A. Stratakis, Analysis of ARMC5 expression in human tissues, Mol Cell Endocrinol, vol.441, pp.140-145, 2017.

A. Berthon, F. R. Faucz, S. Espiard, L. Drougat, J. Bertherat et al., Age-dependent effects of Armc5 haploinsufficiency on adrenocortical function, Hum Mol Genet, vol.26, pp.3495-3507, 2017.

A. Berthon, A. Martinez, J. Bertherat, and V. P. , Wnt/?-catenin signalling in adrenal physiology and tumour development, Mol Cell Endocrinol, vol.351, pp.87-95, 2012.

A. Berthon, I. Sahut-barnola, S. Lambert-langlais, C. De-joussineau, C. Damon-soubeyrand et al., Constitutive beta-catenin activation induces adrenal hyperplasia and promotes adrenal cancer development, Hum Mol Genet, vol.19, pp.1561-1576, 2010.

S. Betancourt-calle, R. A. Calle, C. M. Isales, S. White, H. Rasmussen et al., Differential effects of agonists of aldosterone secretion on steroidogenic acute regulatory phosphorylation, Mol Cell Endocrinol, vol.173, pp.87-94, 2001.

F. Beuschlein, M. Fassnacht, G. Assié, D. Calebiro, C. A. Stratakis et al., Constitutive activation of PKA catalytic subunit in adrenal Cushing's syndrome, N Engl J Med, vol.370, pp.1019-1028, 2014.

H. S. Beyer, S. G. Matta, and B. M. Sharp, Regulation of the Messenger Ribonucleic Acid for Corticotropin-Releasing Factor in the Paraventricular Nucleus and Other Brain Sites of the Rat, Endocrinology, vol.123, pp.2117-2123, 1988.

R. F. Bing and D. Schulster, Steroidogenesis in isolated rat adrenal glomerulosa cells: response to physiological concentrations of angiotensin II and effects of potassium, serotonin and [Sar1,Ala8]-angiotensin II, J Endocrinol, vol.74, pp.261-272, 1977.

J. Bockaert, S. Claeysen, V. Compan, and A. Dumuis, ) receptors, a place in the sun: act two, Curr Opin Pharmacol, vol.11, issue.4, pp.87-93, 2011.

W. B. Bollag, Regulation of aldosterone synthesis and secretion, Compr Physiol, vol.4, pp.1017-1055, 2014.

R. J. Bolt, M. M. Van-weissenbruch, H. N. Lafeber, D. De-waal, and H. A. , Glucocorticoids and lung development in the fetus and preterm infant, Pediatr Pulmonol, vol.32, pp.76-91, 2001.

E. Boonen, S. R. Bornstein, and G. Van-den-berghe, New insights into the controversy of adrenal function during critical illness, Lancet Diabetes Endocrinol, vol.3, pp.805-815, 2015.

E. Boonen, H. Vervenne, P. Meersseman, R. Andrew, L. Mortier et al., Reduced cortisol metabolism during critical illness, N Engl J Med, vol.368, pp.1477-1488, 2013.

S. R. Bornstein, W. C. Engeland, M. Ehrhart-bornstein, and J. P. Herman, Dissociation of ACTH and glucocorticoids, Trends Endocrinol Metab TEM, vol.19, pp.175-180, 2008.

S. R. Bornstein, J. A. Gonzalez-hernandez, M. Ehrhart-bornstein, G. Adler, and S. Wa, Intimate contact of chromaffin and cortical cells within the human adrenal gland forms the cellular basis for important intraadrenal interactions, J Clin Endocrinol Metab, vol.78, pp.225-232, 1994.

S. R. Bornstein, H. Rutkowski, and I. Vrezas, Cytokines and steroidogenesis, Mol Cell Endocrinol, vol.215, pp.135-141, 2004.

J. Böse, L. Grotewold, and U. Rüther, Pallister-Hall syndrome phenotype in mice mutant for Gli3, Hum Mol Genet, vol.11, pp.1129-1135, 2002.

B. A. Boston, S. Mandel, S. Lafranchi, and M. Bliziotes, Activating mutation in the stimulatory guanine nucleotide-binding protein in an infant with Cushing's syndrome and nodular adrenal hyperplasia, J Clin Endocrinol Metab, vol.79, pp.890-893, 1994.

S. Boulkroun, B. Samson-couterie, G. Amar, L. Plouin, P. Sibony et al., AldosteroneProducing Adenoma Formation in the Adrenal Cortex Involves Expression of Stem/Progenitor Cell Markers, Endocrinology, vol.152, pp.4753-4763, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00728048

I. Bourdeau, D. 'amour, P. Hamet, P. Boutin, J. M. Lacroix et al., Aberrant membrane hormone receptors in incidentally discovered bilateral macronodular adrenal hyperplasia with subclinical Cushing's syndrome, J Clin Endocrinol Metab, vol.86, pp.5534-5540, 2001.

I. Bourdeau, L. Matyakhina, S. G. Stergiopoulos, F. Sandrini, S. Boikos et al., chromosomal losses and alterations of protein kinase a subunit expression and activity in adrenocorticotropin-independent macronodular adrenal hyperplasia, J Clin Endocrinol Metab, vol.91, pp.3626-3632, 2006.

H. Boyer, J. Wils, S. Renouf, A. Arabo, C. Duparc et al., Dysregulation of Aldosterone Secretion in Mast Cell-Deficient Mice, Hypertens Dallas Tex, vol.70, pp.1256-1263, 1979.
URL : https://hal.archives-ouvertes.fr/hal-01928604

P. B. Bradley, G. Engel, W. Feniuk, J. R. Fozard, P. P. Humphrey et al., Proposals for the classification and nomenclature of functional receptors for 5-hydroxytryptamine, Neuropharmacology, vol.25, pp.563-576, 1986.

Z. Bram, E. Louiset, B. Ragazzon, S. Renouf, J. Wils et al., PKA regulatory subunit 1A inactivating mutation induces serotonin signaling in primary pigmented nodular adrenal disease, JCI Insight, vol.1, 2016.
URL : https://hal.archives-ouvertes.fr/hal-02108040

T. Brattelid, E. Qvigstad, J. A. Lynham, P. Molenaar, H. Aass et al., Functional serotonin 5-HT4 receptors in porcine and human ventricular myocardium with increased 5-HT4 mRNA in heart failure, Naunyn Schmiedebergs Arch Pharmacol, vol.370, pp.157-166, 2004.

L. Breault, J. Lehoux, G. , and N. , The angiotensin AT2 receptor is present in the human fetal adrenal gland throughout the second trimester of gestation, J Clin Endocrinoloy Metab, vol.81, pp.3914-3936, 1996.

B. B. Brodie and P. A. Shore, A concept for a role of serotonin and norepinephrine as chemical mediators in the brain, Ann N Y Acad Sci, vol.66, pp.631-642, 1957.

A. N. Brooks, D. M. Hagan, and D. C. Howe, Neuroendocrine regulation of pituitary-adrenal function during fetal life, Eur J Endocrinol, vol.135, pp.153-165, 1996.

C. E. Brown-séquard, On a New Therapeutic Method Consisting in the Use of Organic Liquids Extracted from Glands and Other Organs, Br Med J, vol.1, pp.1212-1214, 1893.

M. Burns and R. Gw, Intracellular cholesterol homeostasis and amyloid precursor protein processing, Biochim Biophys Acta, vol.1801, pp.853-859, 2010.

J. T. Busada and J. A. Cidlowski, Chapter Five -Mechanisms of Glucocorticoid Action During Development, Current Topics in Developmental Biology, pp.147-170, 2017.

D. W. Cain and J. A. Cidlowski, Immune regulation by glucocorticoids, Nat Rev Immunol, vol.17, pp.233-247, 2017.

D. Calebiro, G. D. Dalmazi, K. Bathon, C. L. Ronchi, and F. Beuschlein, cAMP signaling in cortisolproducing adrenal adenoma, Eur J Endocrinol, vol.173, pp.99-106, 2015.

L. A. Carlson, L. G. Ekelund, and L. Orö, Metabolic and cardio-vascular effects of serotonin, Life Sci, vol.6, pp.261-271, 1967.

G. A. Carrasco and L. D. Van-de-kar, Neuroendocrine pharmacology of stress, Eur J Pharmacol, vol.463, pp.235-272, 2003.

D. Cartier, S. Jégou, F. Parmentier, I. Lihrmann, E. Louiset et al., Expression profile of serotonin4 (5-HT4) receptors in adrenocortical aldosterone-producing adenomas, Eur J Endocrinol Eur Fed Endocr Soc, vol.153, pp.939-947, 2005.

D. Cartier, I. Lihrmann, F. Parmentier, C. Bastard, J. Bertherat et al., Overexpression of serotonin4 receptors in cisaprideresponsive adrenocorticotropin-independent bilateral macronodular adrenal hyperplasia causing Cushing's syndrome, J Clin Endocrinol Metab, vol.88, pp.248-254, 2003.

R. F. Carvalho, R. A. Ribeiro, R. A. Falcão, R. C. Lima, R. Leitão et al., Angiotensin II potentiates inflammatory edema in rats: Role of mast cell degranulation, Eur J Pharmacol, vol.540, pp.175-182, 2006.

I. P. Cavalcante, E. Clauser, A. Vaczlavik, L. Drougat, C. Lotfi et al., Cullin 3 is a partner of Armadillo repeat containing 5 (ARMC5), the product of the gene responsible for primary bilateral macronodular adrenal hyperplasia, Ann Endocrinol, vol.79, pp.184-185, 2018.

I. P. Cavalcante, M. Nishi, M. Zerbini, M. Q. Almeida, V. B. Brondani et al., The role of ARMC5 in human cell cultures from nodules of primary macronodular adrenocortical hyperplasia (PMAH), Mol Cell Endocrinol, vol.460, pp.36-46, 2018.

F. Ceccato and M. Boscaro, Cushing's Syndrome: Screening and Diagnosis. High Blood Press Cardiovasc Prev Off, J Ital Soc Hypertens, vol.23, pp.209-215, 2016.

F. Cerrito, M. P. Lazzaro, E. Gaudio, P. Arminio, and A. G. , 5HT2-receptors and serotonin release: their role in human platelet aggregation, Life Sci, vol.53, pp.209-215, 1993.

O. Chabre, Cushing syndrome: Physiopathology, etiology and principles of therapy, 2014.

P. Fr, , vol.43, pp.376-392, 1983.

J. R. Challis, D. Sloboda, S. G. Matthews, A. Holloway, N. Alfaidy et al., The fetal placental hypothalamic-pituitary-adrenal (HPA) axis, parturition and post natal health, Mol Cell Endocrinol, vol.185, pp.135-144, 2001.

R. Chanderbhan, B. J. Noland, T. J. Scallen, and G. V. Vahouny, Sterol carrier protein2. Delivery of cholesterol from adrenal lipid droplets to mitochondria for pregnenolone synthesis, J Biol Chem, vol.257, pp.8928-8934, 1982.

Y. T. Chang, G. Mues, and K. Hyland, Alternative splicing in the coding region of human aromatic L-amino acid decarboxylase mRNA, Neurosci Lett, vol.202, pp.157-160, 1996.

V. Chhajlani, R. Muceniece, and J. Wikberg, Molecular Cloning of a Novel Human Melanocortin Receptor, Biochem Biophys Res Commun, vol.195, pp.866-873, 1993.

D. Chida, S. Nakagawa, S. Nagai, H. Sagara, H. Katsumata et al., Melanocortin 2 receptor is required for adrenal gland development, steroidogenesis, and neonatal gluconeogenesis, Proc Natl Acad Sci U S A, vol.104, pp.18205-18210, 2007.

S. Ching and E. Vilain, Targeted disruption of Sonic Hedgehog in the mouse adrenal leads to adrenocortical hypoplasia, genesis, vol.47, pp.628-637, 2009.

S. Chung, G. H. Son, and K. K. , Circadian rhythm of adrenal glucocorticoid: Its regulation and clinical implications, Biochim Biophys Acta BBA -Mol Basis Dis, vol.1812, pp.581-591, 2011.

H. L. Claahsen-van-der-grinten, B. J. Otten, M. Stikkelbroeck, F. Sweep, and A. Hermus, Testicular adrenal rest tumours in congenital adrenal hyperplasia, Best Pract Res Clin Endocrinol Metab, vol.23, pp.209-220, 2009.

A. J. Clark and W. A. , Adrenocorticotropin insensitivity syndromes, Endocr Rev, vol.19, pp.828-843, 1998.

B. J. Clark, J. Wells, S. R. King, and D. M. Stocco, The purification, cloning, and expression of a novel luteinizing hormone-induced mitochondrial protein in MA-10 mouse Leydig tumor cells. Characterization of the steroidogenic acute regulatory protein (StAR), J Biol Chem, vol.269, pp.28314-28322, 1994.

B. F. Clem, E. A. Hudson, and C. Bj, Cyclic Adenosine 3?,5?-Monophosphate (cAMP) Enhances cAMP-Responsive Element Binding (CREB) Protein Phosphorylation and Phospho-CREB Interaction with the Mouse Steroidogenic Acute Regulatory Protein Gene Promoter, Endocrinology, vol.146, pp.1348-1356, 2005.

M. T. Conconi, R. Spinazzi, and G. G. Nussdorfer, Endogenous ligands of PACAP/VIP receptors in the autocrine-paracrine regulation of the adrenal gland, Int Rev Cytol, vol.249, pp.1-51, 2006.

V. Contesse, C. Hamel, H. Lefebvre, A. Dumuis, H. Vaudry et al., Activation of 5-hydroxytryptamine4 receptors causes calcium influx in adrenocortical cells: involvement of calcium in 5-hydroxytryptamine-induced steroid secretion, Mol Pharmacol, vol.49, pp.481-493, 1996.

V. Contesse, H. Lefebvre, S. Lenglet, J. M. Kuhn, C. Delarue et al., Role of 5-HT in the regulation of the brain-pituitary-adrenal axis: effects of 5-HT on adrenocortical cells, Can J Physiol Pharmacol, vol.78, pp.967-983, 2000.

V. Contesse, Y. Reznik, E. Louiset, C. Duparc, D. Cartier et al., Abnormal sensitivity of cortisol-producing adrenocortical adenomas to serotonin: in vivo and in vitro studies, J Clin Endocrinol Metab, vol.90, pp.2843-2850, 2005.
URL : https://hal.archives-ouvertes.fr/hal-01928824

J. D. Crane, R. Palanivel, E. P. Mottillo, A. L. Bujak, H. Wang et al., Inhibiting peripheral serotonin synthesis reduces obesity and metabolic dysfunction by promoting brown adipose tissue thermogenesis, Nat Med, vol.21, pp.166-172, 2015.

R. E. Crowder, The development of the adrenal gland in man, with special reference to origin and ultimate location of cell types and evidence in favor of the "cell migration" theory, Contributions to Embryology pp 193-210, 1957.

M. D. Crowell, Role of serotonin in the pathophysiology of the irritable bowel syndrome, Br J Pharmacol, vol.141, pp.1285-1293, 2004.

M. Da-prada and P. Gb, Content and subcellular localization of catecholamines and 5-hydroxytryptamine in human and animal blood platelets: monoamine distribution between platelets and plasma, Br J Pharmacol, vol.65, pp.653-662, 1979.

A. Dahlstroem and K. Fuxe, Evidence for the existence of monoamine-containing neurons in the central nervous system, Acta Physiol Scand Suppl SUPPL, vol.232, pp.1-55, 1964.

H. Daidoh, H. Morita, T. Mune, M. Murayama, J. Hanafusa et al., Responses of plasma adrenocortical steroids to low dose ACTH in normal subjects, Clin Endocrinol (Oxf), vol.43, pp.311-315, 1995.

A. P. Davenport, S. L. Hoskins, R. E. Kuc, and C. Plumpton, Differential distribution of endothelin peptides and receptors in human adrenal gland, Histochem J, vol.28, pp.779-789, 1996.

C. De-joussineau, I. Sahut-barnola, I. Levy, E. Saloustros, P. Val et al., The cAMP pathway and the control of adrenocortical development and growth, Mol Cell Endocrinol, vol.351, pp.28-36, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01922116

C. De-joussineau, I. Sahut-barnola, F. Tissier, T. Dumontet, C. Drelon et al., mTOR pathway is activated by PKA in adrenocortical cells and participates in vivo to apoptosis resistance in primary pigmented nodular adrenocortical disease (PPNAD), Hum Mol Genet, vol.23, pp.5418-5428, 2014.
URL : https://hal.archives-ouvertes.fr/hal-02108057

M. L. De-marchis, F. Tonelli, D. Quaresmini, D. Lovero, D. Della-morte et al., Desmoid Tumors in Familial Adenomatous Polyposis, Anticancer Res, vol.37, pp.3357-3366, 2017.

M. Degawa-yamauchi, K. A. Moss, J. E. Bovenkerk, S. S. Shankar, C. L. Morrison et al., Regulation of adiponectin expression in human adipocytes: effects of adiposity, glucocorticoids, and tumor necrosis factor alpha, Obes Res, vol.13, pp.662-669, 2005.

A. P. Deiraldi, L. M. Zieher, and D. E. , The 5-hydroxytryptamine content and synthesis of normal and denervated pineal gland, Life Sci, vol.9, pp.691-696, 1962.

C. Delarue, J. M. Conlon, R. Fournier, A. Vaudry, and H. , Endothelins as local activators of adrenocortical cells, J Mol Endocrinol, vol.32, pp.1-7, 2004.

C. Delarue, H. Lefebvre, S. Idres, F. Leboulenger, G. Homo-delarche et al., Serotonin stimulates corticosteroid secretion by frog adrenocortical tissue in vitro, J Steroid Biochem, vol.29, pp.519-525, 1988.

D. Dalmazi, G. Kisker, C. Calebiro, D. Mannelli, M. Canu et al., Novel Somatic Mutations in the Catalytic Subunit of the Protein Kinase A as a Cause of Adrenal Cushing's Syndrome: A European Multicentric Study, J Clin Endocrinol Metab, vol.99, pp.2093-2100, 2014.

D. Cat, A. N. Friederich-persson, M. White, A. Touyz, and R. M. , Adipocytes, aldosterone and obesity-related hypertension, J Mol Endocrinol, vol.57, pp.7-21, 2016.

Y. Doi, K. Atarashi, R. Franco-saenz, and M. Pj, Effect of changes in sodium or potassium balance, and nephrectomy, on adrenal renin and aldosterone concentrations, Hypertens Dallas Tex, vol.6, pp.124-129, 1979.

J. Dörner, M. Rodriguez, V. Ziegler, R. Röhrig, T. Cochran et al., GLI1+ progenitor cells in the adrenal capsule of the adult mouse give rise to heterotopic gonadal-like tissue, Mol Cell Endocrinol, vol.441, pp.164-175, 2017.

M. Doroszko, M. Chrusciel, J. Stelmaszewska, T. Slezak, A. Rivero-muller et al., Luteinizing Hormone and GATA4 Action in the Adrenocortical Tumorigenesis of Gonadectomized Female Mice, Cell Physiol Biochem Int J Exp Cell Physiol Biochem Pharmacol, vol.43, pp.1064-1076, 2017.

A. Dostert, Negative glucocorticoid receptor response elements and their role in glucocorticoid action, Curr Pharm Des, vol.10, pp.2807-2816, 2004.

C. Drelon, A. Berthon, I. Sahut-barnola, M. Mathieu, T. Dumontet et al., PKA inhibits WNT signalling in adrenal cortex zonation and prevents malignant tumour development, Nat Commun, vol.7, 2016.
URL : https://hal.archives-ouvertes.fr/hal-02108042

L. Drougat, S. Espiard, and J. Bertherat, Genetics of primary bilateral macronodular adrenal hyperplasia: a model for early diagnosis of Cushing's syndrome?, Eur J Endocrinol, vol.173, pp.121-131, 2015.

A. Dumuis, R. Bouhelal, M. Sebben, R. Cory, and J. Bockaert, A nonclassical 5-hydroxytryptamine receptor positively coupled with adenylate cyclase in the central nervous system, Mol Pharmacol, vol.34, pp.880-887, 1988.

J. Dunn, Y. Chu, H. H. Qin, and T. Zupekan, Transplantation of Adrenal Cortical Progenitor Cells Enriched by Nile Red, J Surg Res, vol.156, pp.317-324, 2009.

C. Duparc, L. Moreau, J. Dzib, H. Boyer, T. Nomigni et al., Mast Cell Hyperplasia Is Associated With Aldosterone Hypersecretion in a Subset of Aldosterone-Producing Adenomas, J Clin Endocrinol Metab, vol.100, pp.550-560, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01544140

R. M. Eglen, E. H. Wong, A. Dumuis, and J. Bockaert, Central 5-HT4 receptors, Trends Pharmacol Sci, vol.16, pp.391-398, 1995.

M. Ehrhart-bornstein, A. Haidan, S. Alesci, and B. Sr, Neurotransmitters and neuropeptides in the differential regulation of steroidogenesis in adrenocortical-chromaffin cocultures, Endocr Res, vol.26, pp.833-842, 2000.

M. Ehrhart-bornstein, J. P. Hinson, S. R. Bornstein, W. A. Scherbaum, and G. P. Vinson, Intraadrenal Interactions in the Regulation of Adrenocortical Steroidogenesis, Endocr Rev, vol.19, pp.101-143, 1998.

M. Ehrhart-bornstein, V. Lamounier-zepter, A. Schraven, J. Langenbach, H. S. Willenberg et al., Human adipocytes secrete mineralocorticoid-releasing factors, Proc Natl Acad Sci U S A, vol.100, pp.14211-14216, 2003.

N. El-ghorayeb, I. Bourdeau, and A. Lacroix, Role of ACTH and Other Hormones in the Regulation of Aldosterone Production in Primary Aldosteronism, Front Endocrinol, vol.7, p.72, 2016.

A. El-wakil and L. E. , The Wnt/beta-catenin pathway in adrenocortical development and cancer, Mol Cell Endocrinol, vol.332, pp.32-37, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00731010

U. Elbelt, A. Trovato, M. Kloth, E. Gentz, R. Finke et al., Molecular and clinical evidence for an ARMC5 tumor syndrome: concurrent inactivating germline and somatic mutations are associated with both primary macronodular adrenal hyperplasia and meningioma, J Clin Endocrinol Metab, vol.100, pp.119-128, 2015.

R. El-merahbi, M. Löffler, A. Mayer, and G. Sumara, The roles of peripheral serotonin in metabolic homeostasis, FEBS Lett, vol.589, pp.1728-1734, 2015.

P. Elustondo, L. A. Martin, and B. Karten, Mitochondrial cholesterol import, Biochim Biophys Acta BBA -Mol Cell Biol Lipids, vol.1862, pp.90-101, 2017.

M. Engels, P. N. Span, R. T. Mitchell, J. Heuvel, M. A. Marijnissen-van-zanten et al., GATA transcription factors in testicular adrenal rest tumours, Endocr Connect, vol.6, pp.866-875, 2017.

V. Erspamer and A. B. , Identification of enteramine, the specific hormone of the enterochromaffin cell system, as 5-hydroxytryptamine, Nature, vol.169, pp.800-801, 1952.

S. Espiard, L. Drougat, R. Libé, G. Assié, K. Perlemoine et al., ARMC5 Mutations in a Large Cohort of Primary Macronodular Adrenal Hyperplasia: Clinical and Functional Consequences, J Clin Endocrinol Metab, vol.100, pp.926-935, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01813998

S. Espiard, M. J. Knape, K. Bathon, G. Assié, M. Rizk-rabin et al., Activating PRKACB somatic mutation in cortisol-producing adenomas, JCI Insight, vol.3, 2018.

S. Faillot and A. G. , ENDOCRINE TUMOURS: The genomics of adrenocortical tumors, Eur J Endocrinol, vol.174, pp.249-265, 2016.
URL : https://hal.archives-ouvertes.fr/inserm-01291562

H. Falhammar and A. Nordenström, Nonclassic congenital adrenal hyperplasia due to 21-hydroxylase deficiency: clinical presentation, diagnosis, treatment, and outcome, Endocrine, vol.50, pp.32-50, 2015.

J. J. Feige and B. A. , Growth factor regulation of adrenal cortex growth and function, Prog Growth Factor Res, vol.3, pp.103-113, 1991.

F. L. Fernandes-rosa, S. Boulkroun, and Z. , Somatic and inherited mutations in primary aldosteronism, J Mol Endocrinol, vol.59, pp.47-63, 2017.

P. Ferrari, The role of 11?-hydroxysteroid dehydrogenase type 2 in human hypertension, Biochim Biophys Acta BBA -Mol Basis Dis, vol.1802, pp.1178-1187, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00636205

B. D. Freedman, P. B. Kempna, D. L. Carlone, M. Shah, N. A. Guagliardo et al., Adrenocortical zonation results from lineage conversion of differentiated zona glomerulosa cells, Dev Cell, vol.26, pp.666-673, 2013.

J. W. Funder, R. M. Carey, C. Fardella, C. E. Gomez-sanchez, F. Mantero et al., Case Detection, Diagnosis, and Treatment of Patients with Primary Aldosteronism: An Endocrine Society Clinical Practice Guideline, J Clin Endocrinol Metab, vol.93, pp.3266-3281, 2008.

N. Gagnon, K. Caceres, G. Corbeil, E. Ghorayeb, N. Ludwig et al., Genetic Characterization of GnRH/LH-Responsive Primary Aldosteronism, J Clin Endocrinol Metab, 2018.

N. Gallo-payet, Adrenal and extra-adrenal functions of ACTH, J Mol Endocrinol, 2016.

N. Gallo-payet, P. Pothier, and H. Isler, On the presence of chromaffin cells in the adrenal cortex: their possible role in adrenocortical function, Biochem Cell Biol Biochim Biol Cell, vol.65, pp.588-592, 1987.

B. B. García-iglesias, M. E. Mendoza-garrido, G. Gutiérrez-ospina, C. Rangel-barajas, M. Noyola-díaz et al., Sensitization of restraint-induced corticosterone secretion after chronic restraint in rats: involvement of 5-HT? receptors, Neuropharmacology, vol.71, pp.216-227, 2013.

S. Garinet, M. Néou, B. De-la-villéon, S. Faillot, J. Sakat et al., Calling Chromosome Alterations, DNA Methylation Statuses, and Mutations in Tumors by Simple Targeted Next-Generation Sequencing: A Solution for Transferring Integrated Pangenomic Studies into Routine Practice?, J Mol Diagn JMD, vol.19, pp.776-787, 2017.

S. Gaujoux, S. Pinson, A. Gimenez-roqueplo, L. Amar, B. Ragazzon et al., Inactivation of the APC Gene Is Constant in Adrenocortical Tumors from Patients with Familial Adenomatous Polyposis but Not Frequent in Sporadic Adrenocortical Cancers, Clin Cancer Res, vol.16, pp.5133-5141, 2010.

S. Gaujoux, F. Tissier, L. Groussin, R. Libé, B. Ragazzon et al., Wnt/beta-catenin and 3',5'-cyclic adenosine 5'-monophosphate/protein kinase A signaling pathways alterations and somatic beta-catenin gene mutations in the progression of adrenocortical tumors, J Clin Endocrinol Metab, vol.93, pp.4135-4140, 2008.

J. Gautam, S. Banskota, S. C. Regmi, S. Ahn, Y. H. Jeon et al., Tryptophan hydroxylase 1 and 5-HT7 receptor preferentially expressed in triplenegative breast cancer promote cancer progression through autocrine serotonin signaling, Mol Cancer, vol.15, p.75, 2016.

A. F. Gazdar, H. K. Oie, C. H. Shackleton, T. R. Chen, T. J. Triche et al., Establishment and characterization of a human adrenocortical carcinoma cell line that expresses multiple pathways of steroid biosynthesis, Cancer Res, vol.50, pp.5488-5496, 1990.

A. Glasow and B. Sr, Leptin and the adrenal gland, Eur J Clin Invest, vol.30, pp.39-45, 2000.

A. Glasow, A. Haidan, U. Hilbers, M. Breidert, J. Gillespie et al., Expression of Ob receptor in normal human adrenals: differential regulation of adrenocortical and adrenomedullary function by leptin, J Clin Endocrinol Metab, vol.83, pp.4459-4466, 1998.

G. Goh, U. I. Scholl, J. M. Healy, M. Choi, M. L. Prasad et al., Recurrent activating mutation in PRKACA in cortisolproducing adrenal tumors, Nat Genet, vol.46, pp.613-617, 2014.

D. C. Gomes, L. F. Leal, L. M. Mermejo, C. A. Scrideli, C. E. Martinelli et al., Sonic Hedgehog Signaling Is Active in Human Adrenal Cortex Development and Deregulated in Adrenocortical Tumors, J Clin Endocrinol Metab, vol.99, pp.1209-1216, 2014.

R. A. Gomez, Fate of Renin Cells During Development and Disease, Hypertens Dallas Tex, vol.69, pp.387-395, 1979.

J. A. González-hernández, S. R. Bornstein, M. Ehrhart-bornstein, J. E. Gschwend, A. Gwosdow et al., IL-1 is expressed in human adrenal gland in vivo. Possible role in a local immune-adrenal axis, Clin Exp Immunol, vol.99, pp.137-141, 1995.

J. A. Gonzalez-hernandez, S. R. Bornstein, M. Ehrhart-bornstein, E. Späth-schwalbe, G. Jirikowski et al., Interleukin-6 messenger ribonucleic acid expression in human adrenal gland in vivo: new clue to a paracrine or autocrine regulation of adrenal function, J Clin Endocrinol Metab, vol.79, pp.1492-1497, 1994.

J. A. González-hernández, M. Ehrhart-bornstein, E. Späth-schwalbe, W. A. Scherbaum, and B. Sr, Human adrenal cells express tumor necrosis factor-alpha messenger ribonucleic acid: evidence for paracrine control of adrenal function, J Clin Endocrinol Metab, vol.81, pp.807-813, 1996.

, Gottschau M (1883) Struktur und embryonale Entwicklung der Nebennieren bei Säugetieren. Arch Anat Physiol Lpz, pp.412-458

C. Graveleau, H. J. Paust, D. Schmidt-grimminger, and A. K. Mukhopadhyay, Presence of a 5-HT7 receptor positively coupled to adenylate cyclase activation in human granulosa-lutein cells, J Clin Endocrinol Metab, vol.85, pp.1277-1286, 2000.

R. O. Greep and D. Hw, Histological, cytochemical and physiological observations on the regeneration of the rat's adrenal gland following enucleation, Endocrinology, vol.45, pp.42-56, 1949.

E. J. Groen, A. Roos, F. L. Muntinghe, R. H. Enting, J. Vries et al., Extra-Intestinal Manifestations of Familial Adenomatous Polyposis, Ann Surg Oncol, vol.15, pp.2439-2450, 2008.

L. Groussin, L. S. Kirschner, C. Vincent-dejean, K. Perlemoine, E. Jullian et al., Molecular Analysis of the Cyclic AMP-Dependent Protein Kinase A (PKA) Regulatory Subunit 1A (PRKAR1A) Gene in Patients with Carney Complex and Primary Pigmented Nodular Adrenocortical Disease (PPNAD) Reveals Novel Mutations and Clues For Pathophysiology: Augmented PKA Signaling is Associated with Adrenal Tumorigenesis in PPNAD, Am J Hum Genet, vol.71, pp.1433-1442, 2002.

L. Groussin, J. F. Massias, X. Bertagna, and J. Bertherat, Loss of expression of the ubiquitous transcription factor cAMP response element-binding protein (CREB) and compensatory overexpression of the activator CREMtau in the human adrenocortical cancer cell line H295R, J Clin Endocrinol Metab, vol.85, pp.345-354, 2000.

L. Guasti, A. Paul, E. Laufer, and K. P. , Localization of Sonic Hedgehog secreting and receiving cells in the developing and adult rat adrenal cortex, Mol Cell Endocrinol, vol.336, pp.117-122, 2011.

A. Guimier, B. Ragazzon, G. Assié, F. Tissier, B. Dousset et al., AXIN genetic analysis in adrenocortical carcinomas updated, J Endocrinol Invest, vol.36, pp.1000-1003, 2013.

H. Güse-behling, M. Ehrhart-bornstein, S. R. Bornstein, M. R. Waterman, W. A. Scherbaum et al., Regulation of adrenal steroidogenesis by adrenaline: expression of cytochrome P450 genes, J Endocrinol, vol.135, pp.229-237, 1992.

M. Haase, H. S. Willenberg, and B. Sr, Update on the Corticomedullary Interaction in the Adrenal Gland, Pediatr Adrenal Dis, vol.20, pp.28-37, 2011.

A. Haidan, S. R. Bornstein, Z. Liu, L. P. Walsh, D. M. Stocco et al., Expression of adrenocortical steroidogenic acute regulatory (StAR) protein is influenced by chromaffin cells, Mol Cell Endocrinol, vol.165, pp.25-32, 2000.

J. G. Hall, P. D. Pallister, S. K. Clarren, J. B. Beckwith, F. W. Wiglesworth et al., Congenital hypothalamic hamartoblastoma, hypopituitarism, imperforate anus and postaxial polydactyly--a new syndrome? Part I: clinical, causal, and pathogenetic considerations, Am J Med Genet, vol.7, pp.47-74, 1980.

M. Hamon and H. Gozlan, Les récepteurs centraux de la sérotonine, Médecine/Sciences, vol.9, pp.21-30, 1993.

J. Hannon and D. Hoyer, Molecular biology of 5-HT receptors, Behav Brain Res, vol.195, pp.198-213, 2008.

M. J. Harding, H. F. Mcgraw, and A. Nechiporuk, The roles and regulation of multicellular rosette structures during morphogenesis, Dev Camb Engl, vol.141, pp.2549-2558, 2014.

O. Hatano, A. Takakusu, M. Nomura, and K. Morohashi, Identical origin of adrenal cortex and gonad revealed by expression profiles of Ad4BP/SF-1, Genes Cells Devoted Mol Cell Mech, vol.1, pp.663-671, 1996.

N. Hattangady, L. Olala, W. B. Bollag, and R. We, Acute and Chronic Regulation of Aldosterone Production, Mol Cell Endocrinol, vol.350, pp.151-162, 2012.

P. B. Hedlund and J. G. Sutcliffe, Functional, molecular and pharmacological advances in 5-HT7 receptor research, Trends Pharmacol Sci, vol.25, pp.481-486, 2004.

M. Heikkilä, H. Peltoketo, J. Leppäluoto, M. Ilves, O. Vuolteenaho et al., Wnt-4 deficiency alters mouse adrenal cortex function, reducing aldosterone production, Endocrinology, vol.143, pp.4358-4365, 2002.

P. S. Hench and K. Ec, The effect of a hormone of the adrenal cortex (17-hydroxy-11-dehydrocorticosterone; compound E) and of pituitary adrenocorticotropic hormone on rheumatoid arthritis, Proc Staff Meet Mayo Clin, vol.24, pp.181-197, 1949.

J. P. Hinson, L. A. Cameron, A. Purbrick, and S. Kapas, The role of neuropeptides in the regulation of adrenal zona glomerulosa function: effects of substance P, neuropeptide Y, neurotensin, Metenkephalin, Leu-enkephalin and corticotrophin-releasing hormone on aldosterone secretion in the intact perfused rat adrenal, J Endocrinol, vol.140, pp.91-96, 1994.

J. Hofland, P. J. Delhanty, J. Steenbergen, L. J. Hofland, P. M. Van-koetsveld et al., Melanocortin 2 receptor-associated protein (MRAP) and MRAP2 in human adrenocortical tissues: regulation of expression and association with ACTH responsiveness, J Clin Endocrinol Metab, vol.97, pp.747-754, 2012.

A. Horvath, S. Boikos, C. Giatzakis, A. Robinson-white, L. Groussin et al., A genome-wide scan identifies mutations in the gene encoding phosphodiesterase 11A4 (PDE11A) in individuals with adrenocortical hyperplasia, Nat Genet, vol.38, pp.794-800, 2006.

D. Hoyer, 5-HT Receptor Nomenclature: Naming Names, Does It Matter? A Tribute to Maurice Rapport, ACS Chem Neurosci, vol.8, pp.908-919, 2017.

D. Hoyer, Molecular pharmacology and biology of 5-HT1C receptors, Trends Pharmacol Sci, vol.9, pp.89-94, 1988.

D. Hoyer and M. G. , 5-HT receptor classification and nomenclature: towards a harmonization with the human genome, Neuropharmacology, vol.36, pp.419-428, 1997.

L. Hu, X. Lin, H. Lu, B. Chen, and Y. Bai, An overview of hedgehog signaling in fibrosis, Mol Pharmacol, vol.87, pp.174-182, 2015.

C. Huang, S. Miyagawa, D. Matsumaru, K. L. Parker, and Y. , Progenitor Cell Expansion and Organ Size of Mouse Adrenal Is Regulated by Sonic Hedgehog, Endocrinology, vol.151, pp.1119-1128, 2010.

J. H. Humphrey and R. Jaques, The histamine and serotonin content of the platelets and polymorphonuclear leucocytes of various species, J Physiol, vol.124, pp.305-310, 1954.

P. P. Humphrey, P. Hartig, and D. Hoyer, A proposed new nomenclature for 5-HT receptors, Trends Pharmacol Sci, vol.14, pp.233-236, 1993.

R. W. Hunter, J. R. Ivy, and B. Ma, Glucocorticoids and renal Na+ transport: implications for hypertension and salt sensitivity, J Physiol, vol.592, pp.1731-1744, 2014.

J. Hureau, G. Hidden, and M. Att, Vascularisation des glandes surrénales, Anat Clin, vol.2, pp.127-146, 1979.

S. Idres, C. Delarue, H. Lefebvre, A. Larcher, M. Feuilloley et al., Mechanism of action of serotonin on frog adrenal cortex, J Steroid Biochem, vol.34, pp.547-550, 1989.

G. K. Isbister, N. A. Buckley, and I. M. Whyte, Serotonin toxicity: a practical approach to diagnosis and treatment, Med J Aust, vol.187, pp.361-365, 2007.

H. Ishimoto and J. Rb, Development and Function of the Human Fetal Adrenal Cortex: A Key Component in the Feto-Placental Unit, Endocr Rev, vol.32, pp.317-355, 2011.

K. Itcho, K. Oki, K. Kobuke, Y. Yoshii, H. Ohno et al., Aberrant G proteinreceptor expression is associated with DNA methylation in aldosterone-producing adenoma, Mol Cell Endocrinol, vol.461, pp.100-104, 2018.

R. Ivell, M. Balvers, R. Domagalski, H. Ungefroren, N. Hunt et al., Relaxin-like factor: a highly specific and constitutive new marker for Leydig cells in the human testis, Mol Hum Reprod, vol.3, pp.459-466, 1997.

M. Iwata, Y. Oki, T. Okazawa, S. Ishizawa, C. Taka et al., A rare case of adrenocorticotropic hormone (ACTH)-independent macroadrenal hyperplasia showing ectopic production of ACTH, Intern Med Tokyo Jpn, vol.51, pp.2181-2187, 2012.

J. H. Jeon, K. Kim, J. H. Kim, A. Baek, H. Cho et al., A novel adipokine CTRP1 stimulates aldosterone production, FASEB J Off Publ Fed Am Soc Exp Biol, vol.22, pp.1502-1511, 2008.

I. K. Johnsen, M. Slawik, I. Shapiro, M. F. Hartmann, S. A. Wudy et al., Gonadectomy in mice of the inbred strain CE/J induces proliferation of sub-capsular adrenal cells expressing gonadal marker genes, J Endocrinol, vol.190, pp.47-57, 2006.

H. Jørgensen, U. Knigge, A. Kjaer, M. Møller, and J. Warberg, Serotonergic stimulation of corticotropin-releasing hormone and pro-opiomelanocortin gene expression, J Neuroendocrinol, vol.14, pp.788-795, 2002.

A. M. Judd, G. B. Call, M. Barney, C. J. Mcilmoil, A. G. Balls et al., Possible function of IL-6 and TNF as intraadrenal factors in the regulation of adrenal steroid secretion, Ann N Y Acad Sci, vol.917, pp.628-637, 2000.

J. Kaludjerovic and W. We, The Interplay between Estrogen and Fetal Adrenal Cortex, J Nutr Metab, p.837901, 2012.

C. Kamrath, M. F. Hartmann, and W. Sa, Androgen synthesis in patients with congenital adrenal hyperplasia due to 21-hydroxylase deficiency, Horm Metab Res Horm Stoffwechselforschung Horm Metab, vol.45, pp.86-91, 2013.

S. Kang, J. M. Graham, A. H. Olney, and L. G. Biesecker, GLI3 frameshift mutations cause autosomal dominant Pallister-Hall syndrome, Nat Genet, vol.15, pp.266-268, 1997.

A. J. Kaumann, L. Sanders, A. M. Brown, K. J. Murray, and M. J. Brown, A 5-hydroxytryptamine receptor in human atrium, Br J Pharmacol, vol.100, pp.879-885, 1990.

M. Keene and E. E. Hewer, Observations on the Development of the Human Suprarenal Gland, J Anat, vol.61, pp.302-324, 1927.

A. Kim and G. D. Hammer, Adrenocortical Cells with Stem/Progenitor Cell Properties: Recent Advances, Mol Cell Endocrinol, vol.265, pp.10-16, 2007.

A. C. Kim, A. L. Reuter, M. Zubair, T. Else, K. Serecky et al., Targeted disruption of ?-catenin in Sf1-expressing cells impairs development and maintenance of the adrenal cortex, Development, vol.135, pp.2593-2602, 2008.

H. Kim, Y. Toyofuku, F. C. Lynn, E. Chak, T. Uchida et al., Serotonin regulates pancreatic beta cell mass during pregnancy, Nat Med, vol.16, pp.804-808, 2010.

P. King, A. Paul, and E. Laufer, Shh signaling regulates adrenocortical development and identifies progenitors of steroidogenic lineages, Proc Natl Acad Sci U S A, vol.106, pp.21185-21190, 2009.

P. J. King, L. Guasti, and E. Laufer, Hedgehog signalling in endocrine development and disease, J Endocrinol, vol.198, pp.439-450, 2008.

R. Kohen, L. A. Fashingbauer, D. E. Heidmann, C. R. Guthrie, and M. W. Hamblin, Cloning of the mouse 5-HT6 serotonin receptor and mutagenesis studies of the third cytoplasmic loop, Brain Res Mol Brain Res, vol.90, pp.110-117, 2001.

T. Komori, Glucocorticoid Signaling and Bone Biology, Horm Metab Res, vol.48, pp.755-763, 2016.

F. B. Kraemer, Adrenal cholesterol utilization, Mol Cell Endocrinol, vol.265, pp.42-45, 2007.

K. A. Krobert, T. Bach, T. Syversveen, A. M. Kvingedal, and L. Fo, The cloned human 5-HT7 receptor splice variants: a comparative characterization of their pharmacology, function and distribution, Naunyn Schmiedebergs Arch Pharmacol, vol.363, pp.620-632, 2001.

K. A. Krobert and L. Fo, The human 5-HT7 serotonin receptor splice variants: constitutive activity and inverse agonist effects, Br J Pharmacol, vol.135, pp.1563-1571, 2002.

W. K. Kroeze, K. Kristiansen, and R. Bl, Molecular biology of serotonin receptors structure and function at the molecular level, Curr Top Med Chem, vol.2, pp.507-528, 2002.

A. W. Krug, K. Vleugels, S. Schinner, V. Lamounier-zepter, C. G. Ziegler et al., Human adipocytes induce an ERK1/2 MAP kinases-mediated upregulation of steroidogenic acute regulatory protein (StAR) and an angiotensin II-sensitization in human adrenocortical cells, Int J Obes, vol.31, pp.1605-1616, 2005.

R. E. Kuc, C. A. Parker, A. D. Gee, Y. P. Bao, M. J. Brown et al., 5-HT4 receptor distribution in the human adrenal revealed by 3H-GR113808 A 5-HT4 receptor antagonist, Proc Brit Parmacol Soc, vol.3, p.68, 2005.

A. Lacroix, V. Baldacchino, I. Bourdeau, P. Hamet, and J. Tremblay, Cushing's syndrome variants secondary to aberrant hormone receptors, Trends Endocrinol Metab TEM, vol.15, pp.375-382, 2004.

A. Lacroix, E. Bolté, J. Tremblay, J. Dupré, P. Poitras et al., Gastric inhibitory polypeptide-dependent cortisol hypersecretion--a new cause of Cushing's syndrome, N Engl J Med, vol.327, pp.974-980, 1992.

A. Lacroix, R. A. Feelders, C. A. Stratakis, and N. Lk, Cushing's syndrome, Lancet Lond Engl, vol.386, pp.913-927, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01904330

A. Lacroix, P. Hamet, and J. M. Boutin, Leuprolide acetate therapy in luteinizing hormone--dependent Cushing's syndrome, N Engl J Med, vol.341, pp.1577-1581, 1999.

A. Lacroix, N. Ndiaye, J. Tremblay, and P. Hamet, Ectopic and abnormal hormone receptors in adrenal Cushing's syndrome, Endocr Rev, vol.22, pp.75-110, 2001.

B. Lacroix, L. Isabelle, M. Antoine, L. Tânia, J. Tremblay et al., Aberrant G-protein coupled receptor expression in relation to adrenocortical overfunction, Clin Endocrinol (Oxf), vol.73, pp.1-15, 2009.

K. Laenen, G. Haegeman, and P. Vanhoenacker, Structure of the human 5-HT7 receptor gene and characterization of its promoter region, Gene, vol.391, pp.252-263, 2007.

D. S. Lala, D. A. Rice, and P. Kl, Steroidogenic factor I, a key regulator of steroidogenic enzyme expression, is the mouse homolog of fushi tarazu-factor I, Mol Endocrinol Baltim Md, vol.6, pp.1249-1258, 1992.

E. Lalli, J. Barhanin, M. Zennaro, and R. Warth, Local Control of Aldosterone Production and Primary Aldosteronism, Trends Endocrinol Metab TEM, vol.27, pp.123-131, 2016.

B. Lamolet, A. M. Pulichino, T. Lamonerie, Y. Gauthier, T. Brue et al., A pituitary cell-restricted T box factor, Tpit, activates POMC transcription in cooperation with Pitx homeoproteins, Cell, vol.104, pp.849-859, 2001.
URL : https://hal.archives-ouvertes.fr/hal-00023729

A. Lampron, I. Bourdeau, P. Hamet, J. Tremblay, and A. Lacroix, Whole genome expression profiling of glucose-dependent insulinotropic peptide (GIP)-and adrenocorticotropindependent adrenal hyperplasias reveals novel targets for the study of GIP-dependent Cushing's syndrome, J Clin Endocrinol Metab, vol.91, pp.3611-3618, 2006.

C. Langer, C. Piper, J. Vogt, J. Heintze, T. Butz et al., Atrial fibrillation in carcinoid heart disease: The role of serotonin. A review of the literature, Clin Res Cardiol Off J Ger Card Soc, vol.96, pp.114-118, 2007.

E. Laufer, D. Kesper, A. Vortkamp, and K. P. , Sonic hedgehog signaling during adrenal development, Mol Cell Endocrinol, vol.351, pp.19-27, 2012.

N. C. Law, J. Weck, B. Kyriss, J. H. Nilson, and M. Hunzicker-dunn, Lhcgr expression in granulosa cells: roles for PKA-phosphorylated ?-catenin, TCF3, and FOXO1, Mol Endocrinol Baltim Md, vol.27, pp.1295-1310, 2013.

L. F. Leal, E. Szarek, F. Faucz, and C. A. Stratakis, Phosphodiesterase 8B and cyclic AMP signaling in the adrenal cortex, Endocrine, vol.50, pp.27-31, 2015.

A. Lecoq, C. A. Stratakis, S. Viengchareun, R. Chaligné, L. Tosca et al.,

P. Kamenický, Adrenal GIPR expression and chromosome 19q13 microduplications in GIP-dependent Cushing's syndrome, JCI Insight, vol.2, 2017.

D. Lefebvre, . Céline, G. Prévost, J. Bertherat, and L. E. , Cell-to-cell communication in bilateral macronodular adrenal hyperplasia causing hypercortisolism, Front Endocrinol, vol.6, p.34, 2015.
URL : https://hal.archives-ouvertes.fr/inserm-01154353

D. C. Lefebvre, G. Prévost, M. C. Zennaro, J. Bertherat, and L. E. , Paracrine control of steroidogenesis by serotonin in adrenocortical neoplasms, Mol Cell Endocrinol, vol.408, pp.198-204, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01931135

H. Lefebvre, D. Cartier, C. Duparc, I. Lihrmann, V. Contesse et al., Characterization of serotonin(4) receptors in adrenocortical aldosterone-producing adenomas: in vivo and in vitro studies, J Clin Endocrinol Metab, vol.87, pp.1211-1216, 2002.
URL : https://hal.archives-ouvertes.fr/hal-01928563

H. Lefebvre, P. Compagnon, V. Contesse, C. Delarue, C. Thuillez et al., Production and metabolism of serotonin (5-HT) by the human adrenal cortex: paracrine stimulation of aldosterone secretion by 5-HT, J Clin Endocrinol Metab, vol.86, pp.5001-5007, 2001.

H. Lefebvre, V. Contesse, C. Delarue, M. Feuilloley, F. Hery et al., Serotonin-induced stimulation of cortisol secretion from human adrenocortical tissue is mediated through activation of a serotonin4 receptor subtype, Neuroscience, vol.47, pp.999-1007, 1992.

H. Lefebvre, V. Contesse, C. Delarue, A. Legrand, J. M. Kuhn et al., The serotonin-4 receptor agonist cisapride and angiotensin-II exert additive effects on aldosterone secretion in normal man, J Clin Endocrinol Metab, vol.80, pp.504-507, 1995.

H. Lefebvre, V. Contesse, C. Delarue, C. Soubrane, A. Legrand et al., Effect of the serotonin-4 receptor agonist zacopride on aldosterone secretion from the human adrenal cortex: in vivo and in vitro studies, J Clin Endocrinol Metab, vol.77, pp.1662-1666, 1993.

H. Lefebvre, V. Contesse, C. Delarue, H. Vaudry, and J. M. Kuhn, Serotonergic Regulation of Adrenocortical Function, Horm Metab Res, vol.30, pp.398-403, 1998.

H. Lefebvre, C. Duparc, N. Chartrel, S. Jegou, A. Pellerin et al., Intraadrenal adrenocorticotropin production in a case of bilateral macronodular adrenal hyperplasia causing Cushing's syndrome, J Clin Endocrinol Metab, vol.88, pp.3035-3042, 2003.
URL : https://hal.archives-ouvertes.fr/hal-01928681

H. Lefebvre, K. N. Gonzalez, V. Contesse, C. Delarue, and H. Vaudry, Effect of prolonged administration of the serotonin4 (5-HT4) receptor agonist cisapride on aldosterone secretion in healthy volunteers, Endocr Res, vol.24, pp.749-752, 1998.

H. Lefebvre, G. Prévost, and L. E. , Autocrine/paracrine regulatory mechanisms in adrenocortical neoplasms responsible for primary adrenal hypercorticism, Eur J Endocrinol Eur Fed Endocr Soc, vol.169, pp.115-138, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01946266

H. Lefebvre, M. Thomas, C. Duparc, J. Bertherat, and L. E. , Role of ACTH in the Interactive/Paracrine Regulation of Adrenal Steroid Secretion in Physiological and Pathophysiological Conditions, 2016.
URL : https://hal.archives-ouvertes.fr/inserm-01354821

S. Lenglet, E. Louiset, C. Delarue, H. Vaudry, and C. V. , Activation of 5-HT(7) receptor in rat glomerulosa cells is associated with an increase in adenylyl cyclase activity and calcium influx through T-type calcium channels, Endocrinology, vol.143, pp.1748-1760, 2002.

M. Lesurtel, R. Graf, B. Aleil, D. J. Walther, Y. Tian et al., Platelet-derived serotonin mediates liver regeneration, Science, vol.312, pp.104-107, 2006.

D. J. Leszczyszyn, J. A. Jankowski, O. H. Viveros, E. J. Diliberto, J. A. Near et al., Secretion of catecholamines from individual adrenal medullary chromaffin cells, J Neurochem, vol.56, pp.1855-1863, 1991.

J. D. Lever, Observations on the adrenal blood vessels in the rat, J Anat, vol.86, pp.459-467, 1952.

. Lezoualc'h-f, 5-HT4 receptor and Alzheimer's disease: the amyloid connection, Exp Neurol, vol.205, pp.325-329, 2007.

Q. Li, H. Johansson, and L. Grimelius, Innervation of human adrenal gland and adrenal cortical lesions, Virchows Arch Int J Pathol, vol.435, pp.580-589, 1999.

P. Lian, H. Li, Y. Zhang, and J. Z. , Effects of activity of 11?-hydroxysteroid dehydrogenase type 2 on serum potassium levels in Cushing, 2015.

, Zhonghua Yi Xue Za Zhi, vol.95, pp.929-932

J. E. Lim, C. S. Porteus, and N. J. Bernier, Serotonin directly stimulates cortisol secretion from the interrenals in goldfish, Gen Comp Endocrinol, vol.192, pp.246-255, 2013.

V. Locatelli, E. Bresciani, L. Tamiazzo, and A. Torsello, Central nervous system-acting drugs influencing hypothalamic-pituitary-adrenal axis function, Endocr Dev, vol.17, pp.108-120, 2010.

M. Lodish and C. A. Stratakis, A genetic and molecular update on adrenocortical causes of Cushing syndrome, Nat Rev Endocrinol, vol.12, pp.255-262, 2016.

P. B. Loughrey, D. Zhang, and A. P. Heaney, New Treatments for the Carcinoid Syndrome, Endocrinol Metab Clin North Am, vol.47, pp.557-576, 2018.

E. Louiset, V. Contesse, L. Groussin, D. Cartier, C. Duparc et al., Expression of serotonin7 receptor and coupling of ectopic receptors to protein kinase A and ionic currents in adrenocorticotropin-independent macronodular adrenal hyperplasia causing Cushing's syndrome, J Clin Endocrinol Metab, vol.91, pp.4578-4586, 2006.
URL : https://hal.archives-ouvertes.fr/hal-01928836

E. Louiset, C. Duparc, L. Groussin, F. Gobet, R. Desailloud et al., Abnormal Sensitivity to Glucagon and Related Peptides in Primary Adrenal Cushing's Syndrome, Horm Metab Res Horm Stoffwechselforschung Horm Metab, vol.46, pp.876-882, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01931119

E. Louiset, C. Duparc, S. Lenglet, C. E. Gomez-sanchez, and H. Lefebvre, Role of cAMP/PKA pathway and T-type calcium channels in the mechanism of action of serotonin in human adrenocortical cells, Mol Cell Endocrinol, vol.441, pp.99-107, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01931437

E. Louiset, C. Duparc, J. Young, S. Renouf, T. Nomigni et al., Intraadrenal corticotropin in bilateral macronodular adrenal hyperplasia, N Engl J Med, vol.369, pp.2115-2125, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01908272

E. Louiset, K. Isvi, J. M. Gasc, C. Duparc, B. Cauliez et al., Ectopic expression of serotonin7 receptors in an adrenocortical carcinoma co-secreting renin and cortisol, Endocr Relat Cancer, vol.15, pp.1025-1034, 2008.
URL : https://hal.archives-ouvertes.fr/hal-01929382

J. R. Lundblad and J. L. Roberts, Regulation of Proopiomelanocortin Gene Expression in Pituitary, Endocr Rev, vol.9, pp.135-158, 1988.

X. Luo, Y. Ikeda, and P. Kl, A cell-specific nuclear receptor is essential for adrenal and gonadal development and sexual differentiation, Cell, vol.77, pp.481-490, 1994.

B. T. Macdonald, K. Tamai, and X. He, Wnt/beta-catenin signaling: components, mechanisms, and diseases, Dev Cell, vol.17, pp.9-26, 2009.

M. Manchia, B. Carpiniello, F. Valtorta, and S. Comai, Serotonin Dysfunction, Aggressive Behavior, and Mental Illness: Exploring the Link Using a Dimensional Approach, ACS Chem Neurosci, vol.8, pp.961-972, 2017.

H. Mandel, R. Shemer, Z. U. Borochowitz, M. Okopnik, C. Knopf et al., SERKAL syndrome: an autosomalrecessive disorder caused by a loss-of-function mutation in WNT4, Am J Hum Genet, vol.82, pp.39-47, 2008.

F. Mantero, G. Opocher, M. Boscaro, and A. D. , Effect of serotonin on plasma aldosterone in man, J Endocrinol Invest, vol.5, pp.97-99, 1982.

D. Marazziti, S. Baroni, A. Pirone, G. Giannaccini, L. Betti et al., Serotonin receptor of type 6 (5-HT6) in human prefrontal cortex and hippocampus post-mortem: an immunohistochemical and immunofluorescence study, Neurochem Int, vol.62, pp.182-188, 2013.

M. Matsuda, T. Imaoka, A. J. Vomachka, G. A. Gudelsky, Z. Hou et al., Serotonin regulates mammary gland development via an autocrine-paracrine loop, Dev Cell, vol.6, pp.193-203, 2004.

G. M. Mawe and J. M. Hoffman, Serotonin signalling in the gut--functions, dysfunctions and therapeutic targets, Nat Rev Gastroenterol Hepatol, vol.10, pp.473-486, 2013.

T. L. Mazzuco, M. Thomas, M. Martinie, N. Cherradi, N. Sturm et al., Cellular and molecular abnormalities of a macronodular adrenal hyperplasia causing beta-blockersensitive Cushing's syndrome, Arq Bras Endocrinol Metabol, vol.51, pp.1452-1462, 2007.

A. D. Medhurst, F. Lezoualc'h, R. Fischmeister, D. N. Middlemiss, and G. J. Sanger, Quantitative mRNA analysis of five C-terminal splice variants of the human 5-HT4 receptor in the central nervous system by TaqMan real time RT-PCR, Brain Res Mol Brain Res, vol.90, pp.125-134, 2001.

J. Meffre, S. Chaumont-dubel, M. La-cour, C. Loiseau, F. Watson et al., ) receptor recruitment of mTOR as a mechanism for perturbed cognition in schizophrenia, EMBO Mol Med, vol.4, issue.6, pp.1043-1056, 2012.

G. Mengod, M. T. Vilaró, A. Raurich, J. F. López-giménez, R. Cortés et al., 5-HT receptors in mammalian brain: receptor autoradiography and in situ hybridization studies of new ligands and newly identified receptors, Histochem J, vol.28, pp.747-758, 1996.

R. I. Menzies, X. Zhao, L. J. Mullins, J. J. Mullins, C. Cairns et al., Transcription controls growth, cell kinetics and cholesterol supply to sustain ACTH responses, Endocr Connect, vol.6, pp.446-457, 2017.

S. Mesiano, C. L. Coulter, and J. Rb, Localization of cytochrome P450 cholesterol side-chain cleavage, cytochrome P450 17 alpha-hydroxylase/17, 20-lyase, and 3 beta-hydroxysteroid dehydrogenase isomerase steroidogenic enzymes in human and rhesus monkey fetal adrenal glands: reappraisal of functional zonation, J Clin Endocrinol Metab, vol.77, pp.1184-1189, 1993.

S. Mesiano and J. Rb, Developmental and Functional Biology of the Primate Fetal Adrenal Cortex, Endocr Rev, vol.18, pp.378-403, 1997.

L. A. Metherell, J. P. Chapple, S. Cooray, A. David, C. Becker et al., Mutations in MRAP, encoding a new interacting partner of the ACTH receptor, cause familial glucocorticoid deficiency type 2, Nat Genet, vol.37, pp.166-170, 2005.

I. A. Michaelson and W. Vp, The subcellular localization of 5-hydroxytryptamine in guinea pig brain, Biochem Pharmacol, vol.12, pp.203-211, 1963.

W. L. Miller, Why Nobody Has P450scc (20,22 Desmoslase) Deficiencyg, J Clin Endocrinol Metab, vol.83, pp.1399-1400, 1998.

W. L. Miller and H. S. Bose, Early steps in steroidogenesis: intracellular cholesterol trafficking Thematic Review Series: Genetics of Human Lipid Diseases, J Lipid Res, vol.52, pp.2111-2135, 2011.

M. Minami, T. Endo, M. Hirafuji, N. Hamaue, Y. Liu et al., Pharmacological aspects of anticancer drug-induced emesis with emphasis on serotonin release and vagal nerve activity, Pharmacol Ther, vol.99, pp.149-165, 2003.

M. C. Miquel, E. Doucet, M. Riad, A. J. Vergé, D. Hamon et al., Effect of the selective lesion of serotoninergic neurons on the regional distribution of 5-HT1A receptor mRNA in the rat brain, Brain Res Mol Brain Res, vol.14, pp.357-362, 1992.

S. Mitra, S. M. Genuth, L. B. Berman, and V. Vertes, Aldosterone secretion in anephric patients, N Engl J Med, vol.286, pp.61-64, 1972.

L. F. Mohammad-zadeh, L. Moses, and G. Sm, Serotonin: a review, J Vet Pharmacol Ther, vol.31, pp.187-199, 2008.

W. S. Monkhouse and A. Khalique, The adrenal and renal veins of man and their connections with azygos and lumbar veins, J Anat, vol.146, pp.105-115, 1986.

J. More, J. Young, Y. Reznik, G. Raverot, F. Borson-chazot et al., Ectopic ACTH Syndrome in Children and Adolescents, J Clin Endocrinol Metab, vol.96, pp.1213-1222, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00852073

J. Müller and W. H. Ziegler, Stimulation of aldosterone biosynthesis in vitro by serotonin, Acta Endocrinol (Copenh), vol.59, pp.23-35, 1968.

K. Murao, V. Terpstra, S. R. Green, N. Kondratenko, D. Steinberg et al., Characterization of CLA-1, a Human Homologue of Rodent Scavenger Receptor BI, as a Receptor for High Density Lipoprotein and Apoptotic Thymocytes, J Biol Chem, vol.272, pp.17551-17557, 1997.

K. Murphy, X. Zhang, R. Gainetdinov, J. M. Beaulieu, and M. G. Caron, A regulatory domain in the N terminus of tryptophan hydroxylase 2 controls enzyme expression, J. Biol. Chem, vol.283, pp.13216-13224, 2008.

A. Naccache, E. Louiset, C. Duparc, A. Laquerrière, S. Patrier et al., Temporal and spatial distribution of mast cells and steroidogenic enzymes in the human fetal adrenal, Mol Cell Endocrinol, vol.434, pp.69-80, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01931413

Y. Nakamura, N. G. Hattangady, P. Ye, F. Satoh, R. Morimoto et al., Aberrant gonadotropin-releasing hormone receptor (GnRHR) expression and its regulation of CYP11B2 expression and aldosterone production in adrenal aldosterone-producing adenoma (APA), Mol Cell Endocrinol, vol.384, pp.102-108, 2014.

R. Natarajan, S. Ploszaj, R. Horton, and J. Nadler, Tumor necrosis factor and interleukin-1 are potent inhibitors of angiotensin-II-induced aldosterone synthesis, Endocrinology, vol.125, pp.3084-3089, 1989.

A. M. Neville and A. M. Mackay, The structure of the human adrenal cortex in health and disease, Clin Endocrinol Metab, vol.1, pp.361-395, 1972.

J. Newell-price, Proopiomelanocortin gene expression and DNA methylation: implications for Cushing's syndrome and beyond, J Endocrinol, vol.177, pp.365-372, 2003.

W. Ni and S. W. Watts, 5-hydroxytryptamine in the cardiovascular system: focus on the serotonin transporter (SERT), Clin Exp Pharmacol Physiol, vol.33, pp.575-583, 2006.

K. Nishimoto, R. Harris, W. E. Rainey, and T. Seki, Sodium deficiency regulates rat adrenal zona glomerulosa gene expression, Endocrinology, vol.155, pp.1363-1372, 2014.

A. Nishiyama, K. , and S. , New Approaches to Blockade of the Renin-AngiotensinAldosterone System: Overview of Regulation of the Renin-Angiotensin-Aldosterone System, J Pharmacol Sci, vol.113, pp.289-291, 2010.

M. Noda, H. Higashida, S. Aoki, and K. Wada, Multiple signal transduction pathways mediated by 5-HT receptors, Mol Neurobiol, vol.29, pp.31-39, 2004.

G. G. Nussdorfer and M. Lk, Role of tachykinins in the regulation of the hypothalamopituitary-adrenal axis, Peptides, vol.19, pp.949-968, 1998.

E. Obinu, G. Locci, C. Gerosa, V. Fanos, L. Vinci et al., Adrenal stem cell niches are located between adrenal and renal capsules, J Pediatr Neonatal Individ Med, vol.5, p.9, 2016.

K. Orford, C. Crockett, J. P. Jensen, A. M. Weissman, and S. W. Byers, Serine phosphorylationregulated ubiquitination and degradation of beta-catenin, J Biol Chem, vol.272, pp.24735-24738, 1997.

J. E. Pabon, X. Li, Z. M. Lei, J. S. Sanfilippo, M. A. Yussman et al., Novel presence of luteinizing hormone/chorionic gonadotropin receptors in human adrenal glands, J Clin Endocrinol Metab, vol.81, pp.2397-2400, 1996.

, Pal SB (1978) 6-Hydroxylation of cortisol and urinary 6beta-hydroxycortisol, Metabolism, vol.27, pp.1003-1011

R. M. Paragliola, G. Papi, A. Pontecorvi, and C. Sm, Treatment with Synthetic Glucocorticoids and the Hypothalamus-Pituitary-Adrenal Axis, Int J Mol Sci, vol.18, 2017.

G. Päth, S. R. Bornstein, M. Ehrhart-bornstein, and S. Wa, Interleukin-6 and the interleukin-6 receptor in the human adrenal gland: expression and effects on steroidogenesis, J Clin Endocrinol Metab, vol.82, pp.2343-2349, 1997.

A. H. Payne and H. Db, Overview of Steroidogenic Enzymes in the Pathway from Cholesterol to Active Steroid Hormones, Endocr Rev, vol.25, pp.947-970, 2004.

G. J. Pepe and A. E. , Actions of placental and fetal adrenal steroid hormones in primate pregnancy, Endocr Rev, vol.16, pp.608-648, 1995.

I. Perogamvros, D. W. Ray, and P. J. Trainer, Regulation of cortisol bioavailability-effects on hormone measurement and action, Nat Rev Endocrinol, vol.8, pp.717-727, 2012.

S. J. Peroutka and T. A. Howell, The molecular evolution of G protein-coupled receptors: focus on 5-hydroxytryptamine receptors, Neuropharmacology, vol.33, pp.319-324, 1994.

V. Perraudin, C. Delarue, H. Lefebvre, D. Rego, J. Vaudry et al., Evidence for a role of vasopressin in the control of aldosterone secretion in primary aldosteronism: in vitro and in vivo studies, J Clin Endocrinol Metab, vol.91, pp.1566-1572, 2006.

P. Pervanidou and C. Gp, Post-traumatic Stress Disorder in children and adolescents: from Sigmund Freud's "trauma" to psychopathology and the (Dys)metabolic syndrome, Horm Metab Res Horm Stoffwechselforschung Horm Metab, vol.39, pp.413-419, 2007.

J. Peters, Local renin-angiotensin systems in the adrenal gland, Peptides, vol.34, pp.427-432, 2012.

M. Pihlajoki, J. Dörner, R. S. Cochran, M. Heikinheimo, and W. Db, Adrenocortical zonation, renewal, and remodeling, Front Endocrinol, vol.6, p.27, 2015.

U. Plöckinger, M. Chrusciel, M. Doroszko, W. Saeger, O. Blankenstein et al., Functional Implications of LH/hCG Receptors in Pregnancy-Induced Cushing Syndrome, J Endocr Soc, vol.1, pp.57-71, 2017.

H. Raff, S. T. Sharma, and N. Lk, Physiological Basis for the Etiology, Diagnosis, and Treatment of Adrenal Disorders: Cushing's Syndrome, Adrenal Insufficiency, and Congenital Adrenal Hyperplasia, Compr Physiol, vol.4, pp.739-769, 2014.

N. A. Rahman, S. Kiiveri, A. Rivero-müller, J. Levallet, S. Vierre et al., Adrenocortical tumorigenesis in transgenic mice expressing the inhibin alpha-subunit promoter/simian virus 40 T-antigen transgene: relationship between ectopic expression of luteinizing hormone receptor and transcription factor GATA-4, Mol Endocrinol Baltim Md, vol.18, pp.2553-2569, 2004.

W. E. Rainey, Adrenal zonation: clues from 11?-hydroxylase and aldosterone synthase, Mol Cell Endocrinol, vol.151, pp.151-160, 1999.

W. E. Rainey and Y. Nakamura, Regulation of the adrenal androgen biosynthesis, J Steroid Biochem Mol Biol, vol.108, pp.281-286, 2008.

C. V. Rao, X. L. Zhou, and Z. M. Lei, Functional luteinizing hormone/chorionic gonadotropin receptors in human adrenal cortical H295R cells, Biol Reprod, vol.71, pp.579-587, 2004.

M. M. Rapport, A. A. Green, and P. Ih, Partial purification of the vasoconstrictor in beef serum, J Biol Chem, vol.174, pp.735-741, 1948.

G. Reid and R. M. , Pharmacological actions of synthetic 5-hydroxytryptamine (serotonin, thrombocytin), Nature, vol.169, pp.801-802, 1952.

Y. Reznik, V. Allali-zerah, J. A. Chayvialle, R. Leroyer, P. Leymarie et al., Food-dependent Cushing's syndrome mediated by aberrant adrenal sensitivity to gastric inhibitory polypeptide, N Engl J Med, vol.327, pp.981-986, 1992.

Y. Rhayem, L. G. Perez-rivas, A. Dietz, K. Bathon, C. Gebhard et al., PRKACA Somatic Mutations Are Rare Findings in Aldosterone-Producing Adenomas, J Clin Endocrinol Metab, vol.101, pp.3010-3017, 2016.

C. L. Ronchi, D. Dalmazi, G. Faillot, S. Sbiera, S. Assié et al., Beuschlein F, and European Network for the Study of Adrenocortical Tumors (ENSAT) (2016) Genetic Landscape of Sporadic Unilateral Adrenocortical Adenomas Without PRKACA p.Leu206Arg Mutation, J Clin Endocrinol Metab, vol.101, pp.3526-3538

H. Rosenkrantz, A direct influence of 5-hydroxytryptamine on the adrenal cortex, Endocrinology, vol.64, pp.355-362, 1959.

G. Rossi, G. Albertin, A. Belloni, L. Zanin, M. A. Biasolo et al., Gene expression, localization, and characterization of endothelin A and B receptors in the human adrenal cortex, J Clin Invest, vol.94, pp.1226-1234, 1994.

G. P. Rossi, P. G. Andreis, G. Neri, C. Tortorella, M. R. Pelizzo et al., Endothelin-1 stimulates aldosterone synthesis in Conn's adenomas via both A and B receptors coupled with the protein kinase C-and cyclooxygenase-dependent signaling pathways, J Investig Med Off Publ Am Fed Clin Res, vol.48, pp.343-350, 2000.

G. P. Rossi, D. Sticchi, L. Giuliani, P. Bernante, S. Zavattiero et al., Adiponectin receptor expression in the human adrenal cortex and aldosterone-producing adenomas, Int J Mol Med, vol.17, pp.975-980, 2006.

B. L. Roth, Drugs and valvular heart disease, N Engl J Med, vol.356, pp.6-9, 2007.

A. K. Roth-isigkeit and P. Schmucker, Postoperative dissociation of blood levels of cortisol and adrenocorticotropin after coronary artery bypass grafting surgery, Steroids, vol.62, pp.695-699, 1997.

R. B. Rothman, M. H. Baumann, J. E. Savage, L. Rauser, A. Mcbride et al., Evidence for possible involvement of 5-HT(2B) receptors in the cardiac valvulopathy associated with fenfluramine and other serotonergic medications, Circulation, vol.102, pp.2836-2841, 2000.

R. T. Rubin, T. H. Miller, M. E. Rhodes, and C. Rk, Adrenal cortical responses to low-and high-dose ACTH(1-24) administration in major depressives vs. matched controls, Psychiatry Res, vol.143, pp.43-50, 2006.

C. Ruggiero and L. E. , Impact of ACTH Signaling on Transcriptional Regulation of Steroidogenic Genes, 2016.

I. Sahut-barnola, C. Joussineau, . De, P. Val, S. Lambert-langlais et al., Cushing's Syndrome and Fetal Features Resurgence in Adrenal Cortex-Specific Prkar1a Knockout Mice, PLOS Genet, vol.6, p.1000980, 2010.

I. Sakuma, S. Higuchi, M. Fujimoto, T. Takiguchi, A. Nakayama et al., Cushing Syndrome Due to ACTH-Secreting Pheochromocytoma, Aggravated by Glucocorticoid-Driven Positive-Feedback Loop, J Clin Endocrinol Metab, vol.101, pp.841-846, 2016.

A. Salomon, M. Keramidas, C. Maisin, and T. M. , Loss of ?-catenin in adrenocortical cancer cells causes growth inhibition and reversal of epithelial-to-mesenchymal transition, Oncotarget, vol.6, pp.11421-11433, 2015.

S. A. Salyer, J. Parks, M. T. Barati, E. D. Lederer, B. J. Clark et al., Aldosterone regulates Na(+), K(+) ATPase activity in human renal proximal tubule cells through mineralocorticoid receptor, Biochim Biophys Acta, vol.1833, pp.2143-2152, 2013.

M. Sand, S. Uecker, F. G. Bechara, M. Gelos, D. Sand et al., Simultaneous ectopic adrenocorticotropic hormone syndrome and adrenal metastasis of a medullary thyroid carcinoma causing paraneoplastic Cushing's syndrome, Int Semin Surg Oncol, vol.4, p.15, 2007.

K. Saner-amigh, B. A. Mayhew, F. Mantero, F. Schiavi, P. C. White et al., Elevated expression of luteinizing hormone receptor in aldosterone-producing adenomas, J Clin Endocrinol Metab, vol.91, pp.1136-1142, 2006.

A. Santos, E. Resmini, J. C. Pascual, I. Crespo, and W. Sm, Psychiatric Symptoms in Patients with Cushing's Syndrome: Prevalence, Diagnosis and Management, Drugs, vol.77, pp.829-842, 2017.

H. Sasano, Y. Ohashi, T. Suzuki, and H. Nagura, Vascularity in human adrenal cortex, Mod Pathol Off J U S Can Acad Pathol Inc, vol.11, pp.329-333, 1998.

H. Sasano, T. Suzuki, and H. Nagura, ACTH-independent macronodular adrenocortical hyperplasia: immunohistochemical and in situ hybridization studies of steroidogenic enzymes, Mod Pathol Off J U S Can Acad Pathol Inc, vol.7, pp.215-219, 1994.

Y. Sato, S. Maekawa, R. Ishii, M. Sanada, T. Morikawa et al., Recurrent somatic mutations underlie corticotropin-independent Cushing's syndrome, Science, vol.344, pp.917-920, 2014.

H. Satoh, R. Saito, S. Hisata, J. Shiihara, S. Taniuchi et al., An ectopic ACTH-producing small cell lung carcinoma associated with enhanced corticosteroid biosynthesis in the peritumoral areas of adrenal metastasis, Lung Cancer Amst Neth, vol.76, pp.486-490, 2012.

E. A. Schäfer, Oliver-Sharpey Lectures on the present condition of our knowledge regarding the functions of the suprarenal capsules, Br Med J, vol.1, pp.1277-1281, 1908.

S. Schinner and B. Sr, Cortical-chromaffin cell interactions in the adrenal gland, Endocr Pathol, vol.16, pp.91-98, 2005.

S. Schinner, H. S. Willenberg, D. Krause, M. Schott, V. Lamounier-zepter et al., Adipocyte-derived products induce the transcription of the StAR promoter and stimulate aldosterone and cortisol secretion from adrenocortical cells through the Wnt-signaling pathway, Int J Obes, vol.31, pp.864-870, 2005.

A. Sculptoreanu, T. Scheuer, and C. Wa, Voltage-dependent potentiation of L-type Ca2+ channels due to phosphorylation by cAMP-dependent protein kinase, Nature, vol.364, pp.240-243, 1993.

M. B. Sewer and L. D. , Regulation of Steroid Hormone Biosynthesis by the Cytoskeleton, Lipids, vol.43, pp.1109-1115, 2008.

L. H. Shaikh, J. Zhou, A. Teo, S. Garg, S. G. Neogi et al., LGR5 Activates Noncanonical Wnt Signaling and Inhibits Aldosterone Production in the Human Adrenal, J Clin Endocrinol Metab, vol.100, pp.836-844, 2015.

Z. She and Y. , Molecular mechanisms involved in mammalian primary sex determination, J Mol Endocrinol, vol.53, pp.21-37, 2014.

S. Singh, Trials of new antiarrhythmic drugs for maintenance of sinus rhythm in patients with atrial fibrillation, J Interv Card Electrophysiol Int J Arrhythm Pacing, vol.10, pp.71-76, 2004.

A. J. Sleight and P. Sj, Identification of 5-hydroxytryptamine1A receptor agents using a composite pharmacophore analysis and chemical database screening, Naunyn Schmiedebergs Arch Pharmacol, vol.343, pp.109-116, 1991.

A. Spät, Glomerulosa cell-a unique sensor of extracellular K+ concentration, Mol Cell Endocrinol, vol.217, pp.23-26, 2004.

A. Spät and L. Hunyady, Control of Aldosterone Secretion: A Model for Convergence in Cellular Signaling Pathways, Physiol Rev, vol.84, pp.489-539, 2004.

J. L. Stamos and W. I. Weis, The ?-Catenin Destruction Complex, Cold Spring Harb Perspect Biol, vol.5, p.7898, 2013.

M. St-jean, N. E. Ghorayeb, I. Bourdeau, and A. Lacroix, Aberrant G-protein coupled hormone receptor in adrenal diseases, Best Pract Res Clin Endocrinol Metab, vol.32, pp.165-187, 2018.

D. M. Stocco, StAR Protein and the Regulation of Steroid Hormone Biosynthesis, Annu Rev Physiol, vol.63, pp.193-213, 2001.

C. A. Stratakis and B. Sa, Genetics of adrenal tumors associated with Cushing's syndrome: a new classification for bilateral adrenocortical hyperplasias, Nat Clin Pract Endocrinol Metab, vol.3, pp.748-757, 2007.

C. A. Stratakis, J. A. Carney, L. S. Kirschner, H. S. Willenberg, S. Brauer et al., Synaptophysin immunoreactivity in primary pigmented nodular adrenocortical disease: neuroendocrine properties of tumors associated with Carney complex, J Clin Endocrinol Metab, vol.84, pp.1122-1128, 1999.

M. A. Stull, V. Pai, A. J. Vomachka, A. M. Marshall, G. A. Jacob et al., Mammary gland homeostasis employs serotonergic regulation of epithelial tight junctions, Proc Natl Acad Sci U S A, vol.104, pp.16708-16713, 2007.

A. Teo, S. Garg, H. Shaikh, L. Zhou, J. et al., Pregnancy, Primary Aldosteronism, and Adrenal CTNNB1 Mutations, 2015.

M. Thomas, M. Keramidas, E. Monchaux, and F. , Dual Hormonal Regulation of Endocrine Tissue Mass and Vasculature by Adrenocorticotropin in the Adrenal Cortex, Endocrinology, vol.145, pp.4320-4329, 2004.

F. Tissier, C. Cavard, L. Groussin, K. Perlemoine, G. Fumey et al., Mutations of beta-catenin in adrenocortical tumors: activation of the Wnt signaling pathway is a frequent event in both benign and malignant adrenocortical tumors, Cancer Res, vol.65, pp.7622-7627, 2005.

T. Tominaga, J. Fukata, Y. Naito, T. Usui, N. Murakami et al., Prostaglandin-dependent in vitro stimulation of adrenocortical steroidogenesis by interleukins, Endocrinology, vol.128, pp.526-531, 1991.

A. F. Turcu and A. Rj, Adrenal steroidogenesis and congenital adrenal hyperplasia, Endocrinol Metab Clin North Am, vol.44, pp.275-296, 2015.

B. M. Twarog and P. Ih, Serotonin content of some mammalian tissues and urine and a method for its determination, Am J Physiol, vol.175, pp.157-161, 1953.

S. Uchida-kitajima, T. Yamauchi, Y. Takashina, M. Okada-iwabu, M. Iwabu et al., 5-Hydroxytryptamine 2A receptor signaling cascade modulates adiponectin and plasminogen activator inhibitor 1 expression in adipose tissue, FEBS Lett, vol.582, pp.3037-3044, 2008.

Y. M. Ulrich-lai, H. F. Figueiredo, M. M. Ostrander, D. C. Choi, W. C. Engeland et al., Chronic stress induces adrenal hyperplasia and hypertrophy in a subregion-specific manner, Am J Physiol-Endocrinol Metab, vol.291, pp.965-973, 2006.

P. M. Vanhoutte, Cardiovascular effects of serotonin, J Cardiovasc Pharmacol, vol.10, pp.8-11, 1987.

I. Vermes and A. Beishuizen, The hypothalamic-pituitary-adrenal response to critical illness, Best Pract Res Clin Endocrinol Metab, vol.15, pp.495-511, 2001.

D. Vezzosi, R. Libé, C. Baudry, M. Rizk-rabin, A. Horvath et al., Phosphodiesterase 11A (PDE11A) gene defects in patients with acth-independent macronodular adrenal hyperplasia (AIMAH): functional variants may contribute to genetic susceptibility of bilateral adrenal tumors, J Clin Endocrinol Metab, vol.97, pp.2063-2069, 2012.

G. Vitellius, S. Trabado, C. Hoeffel, J. Bouligand, A. Bennet et al., Significant prevalence of NR3C1 mutations in incidentally discovered bilateral adrenal hyperplasia: results of the French MUTA-GR Study, Eur J Endocrinol, vol.178, pp.411-423, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01822592

J. Vouillarmet, F. Fernandes-rosa, J. Graeppi-dulac, P. Lantelme, M. Decaussin-petrucci et al., Aldosterone-Producing Adenoma With a Somatic KCNJ5 Mutation Revealing APC-Dependent Familial Adenomatous Polyposis, J Clin Endocrinol Metab, vol.101, pp.3874-3878, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01850033

I. Vrezas, H. S. Willenberg, G. Mansmann, N. Hiroi, R. Fritzen et al., Ectopic adrenocorticotropin (ACTH) and corticotropin-releasing hormone (CRH) production in the adrenal gland: Basic and clinical aspects, Microsc Res Tech, vol.61, pp.308-314, 2003.

S. Vuorenoja, A. Rivero-muller, S. Kiiveri, M. Bielinska, M. Heikinheimo et al., Adrenocortical tumorigenesis, luteinizing hormone receptor and transcription factors GATA-4 and GATA-6, Mol Cell Endocrinol, vol.269, pp.38-45, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00531903

E. M. Walczak and G. D. Hammer, Regulation of the adrenocortical stem cell niche: implications for disease, Nat Rev Endocrinol, vol.11, pp.14-28, 2015.

E. M. Walczak, R. Kuick, I. Finco, N. Bohin, S. M. Hrycaj et al., Wnt Signaling Inhibits Adrenal Steroidogenesis by Cell-Autonomous and Non-Cell-Autonomous Mechanisms, Mol Endocrinol, vol.28, pp.1471-1486, 2014.

S. W. Walker, E. R. Lightly, S. W. Milner, and W. Bc, Catecholamine stimulation of cortisol secretion by 3-day primary cultures of purified zona fasciculata/reticularis cells isolated from bovine adrenal cortex, Mol Cell Endocrinol, vol.57, pp.139-147, 1988.

D. J. Walther and M. Bader, A unique central tryptophan hydroxylase isoform, Biochem Pharmacol, vol.66, pp.1673-1680, 2003.

W. Wang and G. Giebisch, Regulation of potassium (K) handling in the renal collecting duct, Pflugers Arch, vol.458, pp.157-168, 2009.

R. Waterston, K. Lindblad-toh, E. Birney, J. Rogers, J. F. Abril et al., Nature, vol.420, pp.520-562, 2002.

S. Watson, C. Serrate, and S. Vignot, , 2010.

, Bull Cancer (Paris), vol.97, pp.1477-1483

I. Weigand, C. L. Ronchi, M. Rizk-rabin, G. D. Dalmazi, V. Wild et al., Differential expression of the protein kinase A subunits in normal adrenal glands and adrenocortical adenomas, Sci Rep, vol.7, p.49, 2017.

L. S. Weinstein, A. Shenker, P. V. Gejman, M. J. Merino, and E. Friedman, Activating Mutations of the Stimulatory G Protein in the McCune-Albright Syndrome, N Engl J Med, vol.325, pp.1688-1695, 1991.

P. Werminghaus, M. Haase, P. J. Hornsby, S. Schinner, M. Schott et al., Hedgehog-signaling is upregulated in non-producing human adrenal adenomas and antagonism of hedgehog-signaling inhibits proliferation of NCI-H295R cells and an immortalized primary human adrenal cell line, J Steroid Biochem Mol Biol, vol.139, pp.7-15, 2014.

W. Weyler, Y. P. Hsu, and B. Xo, Biochemistry and genetics of monoamine oxidase, Pharmacol Ther, vol.47, pp.391-417, 1990.

C. Wilke, S. Sheriff, and M. Soleimani, Vasopressin-independent regulation of collecting duct aquaporin-2 in food deprivation, Kidney Int, vol.67, pp.201-216, 2005.

G. H. Williams, Aldosterone Biosynthesis, Regulation, and Classical Mechanism of Action, Heart Fail Rev, vol.10, pp.7-13, 2005.

S. F. Witchel, Congenital Adrenal Hyperplasia, J Pediatr Adolesc Gynecol, vol.30, pp.520-534, 2017.

E. H. Wong, G. P. Reynolds, D. W. Bonhaus, S. Hsu, and R. M. Eglen, Characterization of [3H]GR 113808 binding to 5-HT4 receptors in brain tissues from patients with neurodegenerative disorders, Behav Brain Res, vol.73, pp.249-252, 1996.

V. Wu, S. Wang, S. Chueh, Y. Huang, K. Lin et al., The prevalence of CTNNB1 mutations in primary aldosteronism and consequences for clinical outcomes, vol.7, p.39121, 2017.

Y. Xing, A. Lerario, W. Rainey, and G. D. Hammer, Development of Adrenal Cortex Zonation, Endocrinol Metab Clin North Am, vol.44, pp.243-274, 2015.

Y. Xing, C. R. Parker, M. Edwards, and R. We, Adrenocorticotrophic hormone is a potent regulator of gene expression in human adrenal cells, J Mol Endocrinol, vol.45, pp.59-68, 2010.

Y. Xing, W. E. Rainey, J. W. Apolzan, O. L. Francone, R. Harris et al., Adrenal Cell Aldosterone Production Is Stimulated by Very-Low-Density Lipoprotein (VLDL), Endocrinology, vol.153, pp.721-731, 2012.

L. Ye, L. Xiaoying, X. Kong, W. Wang, Y. Bi et al., Hypomethylation in the promoter region of POMC gene correlates with ectopic overexpression in thymic carcinoids, J Endocrinol, vol.185, pp.337-343, 2005.

P. Ye, B. Mariniello, F. Mantero, H. Shibata, and R. We, G-protein-coupled receptors in aldosterone-producing adenomas: a potential cause of hyperaldosteronism, J Endocrinol, vol.195, pp.39-48, 2007.

J. Young, C. Deneux, M. Grino, C. Oliver, P. Chanson et al., Pitfall of Petrosal Sinus Sampling in a Cushing's Syndrome Secondary to Ectopic Adrenocorticotropin-Corticotropin Releasing Hormone (ACTH-CRH) Secretion, J Clin Endocrinol Metab, vol.83, pp.305-308, 1998.

B. Yu, B. Ragazzon, M. Rizk-rabin, and J. Bertherat, Protein kinase A alterations in endocrine tumors, Horm Metab Res Horm Stoffwechselforschung Horm Metab, vol.44, pp.741-748, 2012.

M. Zennaro, F. L. Fernandes-rosa, and S. Boulkroun, Overview of aldosterone-related genetic syndromes and recent advances, Curr Opin Endocrinol Diabetes Obes, 2018.

M. Zubair, S. Ishihara, S. Oka, K. Okumura, and K. Morohashi, Two-step regulation of Ad4BP/SF-1 gene transcription during fetal adrenal development: initiation by a Hox-Pbx1-Prep1 complex and maintenance via autoregulation by Ad4BP/SF-1, Mol Cell Biol, vol.26, pp.4111-4121, 2006.

O. Zwermann, Y. Suttmann, M. Bidlingmaier, F. Beuschlein, and M. Reincke, Screening for membrane hormone receptor expression in primary aldosteronism, Eur J Endocrinol, vol.160, pp.443-451, 2009.

, Abstract In most cases, cortisol hypersecretion (Cushing's syndrome

, ACTH-independent" owing to suppressed plasma ACTH levels. The molecular mechanisms underlying the maintenance of cortisol hypersecretion by adrenocortical adenomas and BMAHs in the absence of circulating ACTH has long remained unknown. However, major advances have been made during the past recent years in the comprehension of the pathophysiology of primary adrenal CS. Two main types of molecular defects have been shown to favor cortisol hypersecretion by adrenocortical neoplasms: somatic mutations responsible for activation of intracellular signaling pathways and abnormally expressed (or illegitimate) membrane receptors by tumor cells. In the human adrenal gland, serotonin (5-HT), released by subcapsular mast cells stimulates corticosteroid secretion through activation of its type 4 receptor (5-HT4R). The 5-HT4R is principally expressed in zona glomerulosa cells but weakly expressed in zona fasciculata cells explaining why 5-HT strongly stimulates aldosterone production. Interestingly, in primary pigmented nodular adrenocortical disease (PPNAD) cells, activation of the cAMP/PKA pathway by PRKAR1A mutations triggers upregulation of the 5-HT synthesizing enzyme tryptophan hydroxylase (TPH) type 2 together with the 5-HT4, 5-HT6 and 5-HT7 receptors, positively coupled to cAMP/PKA signaling pathway. 5-HT strongly stimulates cortisol production and inhibition of TPH reduced corticosteroidogenesis in cultured PPNAD cells. In human, cortisol secretion is normally stimulated by ACTH also through activation of the cAMP/PKA signaling pathway. Patients suffering from Cushing's disease, paraneoplastic Cushing's syndrome (paraCS), 21-hydroxylase deficiency or BMAH display high plasma or intraadrenal ACTH levels. In these patients, we show that chronic stimulation of cAMP/PKA pathway by ACTH originating from a patient with paraCS, 5-HT and 5-HT4/7 receptors agonists were able to activate cortisol secretion. On the other hand, the role of Wnt/?-catenin signaling pathway in the emergence of illegitimate receptors is still debated, CS) results from ACTH-producing pituitary adenoma (Cushing's disease). Occasionally, CS is the consequence of a unilateral adrenal adenoma or a bilateral macronodular adrenal hyperplasia (BMAH) producing cortisol. In these conditions, hypercortisolism is referred to as

, Our results also demonstrate that intraadrenal 5-HT is involved in corticosteroids hypersecretion related to different diseases including Cushing's disease, paraneoplastic Cushing's syndrome, 21-hydroxylase deficiency and BMAH. TPH inhibitors may thus represent a new therapeutic approach of corticosteroid excess in patients suffering from these disorders

, Dans la surrénale humaine normale, la sérotonine (5-HT), synthétisée et libérée par les mastocytes sous-capsulaires, stimule la sécrétion des corticostéroïdes via son récepteur 5-HT4. Ce dernier est principalement localisé à la surface des cellules de la zone glomérulée mais faiblement exprimé au niveau des cellules de la zone fasciculée, expliquant l'action stimulante prédominante de la 5-HT sur la sécrétion d'aldostérone. Dans la dysplasie micronodulaire pigmentée des surrénales, l'activation de la voie AMPc/PKA par une mutation du gène PRKAR1A est responsable d'une surexpression de la tryptophane hydroxylase (TPH) de type 2, enzyme limitante de la synthèse de 5-HT, Plus rarement, le syndrome de Cushing est la conséquence d'un adénome corticosurrénalien unilatéral ou d'une hyperplasie bilatérale des surrénales (HMBS) sécrétant du cortisol. Ces deux pathologies appartiennent à la catégorie des hypercortisolismes dits ACTH-indépendants en raison des taux plasmatiques effondrés d'adrénocorticotrophine (ACTH)

, (paraCS), d'un déficit en 21-hydroxylase ou d'HMBS présentent des taux d'ACTH plasmatique ou intrasurrénalienne élevés. Chez ces patients, nous montrons que la stimulation chronique de la voie AMPc/PKA par l'ACTH provoque une surexpression de la TPH de type 1

, Par ailleurs, le rôle de la voie Wnt/?-caténine dans l'apparition des récepteurs illégitimes reste controversé. Nous avons donc évalué l'expression des récepteurs 5-HT4, 5-HT6, 5-HT7, du LH-R et du GIP-R dans une tumeur corticosurrénalienne avec mutation germinale du gène APC et deux modèles d'activation constitutive de la voie Wnt/?-caténine dans le cortex surrénalien, incluant des souris génétiquement modifiées et des cellules corticosurrénaliennes humaines en culture primaire. Nos résultats indiquent que l'activation de la voie Wnt/?-caténine favorise une surexpression significative du LH-R dans les 3 modèles étudiés, dans les cellules stéroïdogènes. Pour l'un des patients avec paraCS, nous avons pu montrer que les cellules corticosurrénaliennes en culture sécrètent du cortisol en réponse à la 5-HT ou à des agonistes des récepteurs 5-HT4 ou 5-HT7

/. Ampc, . Pka, and . Wnt, Ils indiquent en outre que la 5-HT intrasurrénalienne est impliquée dans l'hypersécrétion de corticostéroïdes associée à différentes pathologies incluant la maladie de Cushing et le syndrome de Cushing paranéoplasique, le bloc en 21-hydroxylase et l'HMBS. Le recours à des inhibiteurs sélectifs de la tryptophane hydroxylase pourrait donc permettre de réduire l, caténine par des mutations génétiques, favorise l'émergence de systèmes de régulation surrénaliens aberrants

, Mots clés : surrénale, cortisol, ACTH, sérotonine, Wnt/?-caténine