N. G. Hattangady, L. O. Olala, W. B. Bollag, and W. E. Rainey, Mol. Cell. Endocrinol, vol.350, pp.151-162, 2012.

M. Ehrhart-bornstein, Endocr. Rev, vol.19, pp.101-143, 1998.

A. B. Gilchrist, A. Leake, and B. G. Charlton, Acta Anat. (Basel), vol.146, pp.31-35, 1993.

M. L. Nichols, Science, vol.286, pp.1558-1561, 1999.

M. S. Steinhoff, Physiol. Rev, vol.94, pp.265-301, 2014.

J. K. Prague, Lancet, vol.389, pp.1809-1820, 2017.

R. Saito, Y. Takano, and H. Kamiya, J. Pharmacol. Sci, vol.91, pp.87-94, 2003.

C. Heym, Histochem. Cell Biol, vol.104, pp.233-243, 1995.

J. P. Hinson, L. A. Cameron, A. Purbrick, and S. Kapas, J. Endocrinol, vol.140, pp.91-96, 1994.

G. Mazzocchi, C. Macchi, L. K. Malendowicz, and G. G. Nussdorfer, Neuropeptides, vol.29, pp.53-58, 1995.

J. G. Widdicombe, Am. J. Respir. Crit. Care Med, vol.158, pp.171-175, 1998.

N. M. Page, Proc. Natl. Acad. Sci. U. S. A, vol.100, pp.6245-6250, 2003.

C. E. Gomez-sanchez, Mol. Cell. Endocrinol, vol.383, pp.111-117, 2014.

V. Coiro, Neuroendocrinology, vol.56, pp.459-463, 1992.

S. Hubers, S. Wei, and N. Brown, Hypertension, vol.68, p.180, 2016.

M. Choi, K+ channel mutations in adrenal aldosterone-producing adenomas and hereditary hypertension, Science, vol.331, pp.768-772, 2011.

M. Zennaro, S. Boulkroun, and F. Fernandes-rosa, Endocr. Rev, vol.38, pp.516-537, 2017.

J. Lai, Proc. Natl. Acad. Sci. U. S. A, vol.105, pp.12605-12610, 2008.

K. Vleugels, Exp. Clin. Endocrinol. Diabetes, vol.119, pp.644-648, 2011.

M. Doi, Nat. Med, vol.16, pp.67-74, 2010.

P. J. Hesketh, J. Clin. Oncol, vol.35, pp.3240-3261, 2017.

G. Bredy-dobreva, Biomed. Biochim. Acta, vol.47, pp.25-30, 1988.

J. P. Hinson, G. P. Vinson, S. Kapas, and R. Teja, J. Steroid Biochem. Mol. Biol, vol.40, pp.381-389, 1991.

F. Leboulenger, Endocrinology, vol.133, 1993.

N. G. Hattangady, L. O. Olala, W. B. Bollag, and W. E. Rainey, Acute and chronic regulation of aldosterone production, Mol. Cell. Endocrinol, vol.350, pp.151-162, 2012.

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.

A. B. Gilchrist, A. Leake, and B. G. Charlton, Innervation of the human adrenal cortex: simultaneous visualisation using acetylcholinesterase histochemistry and dopamine betahydroxylase immunohistochemistry, Acta Anat. (Basel), vol.146, pp.31-35, 1993.

M. L. Nichols, Transmission of chronic nociception by spinal neurons expressing the substance P receptor, Science, vol.286, pp.1558-1561, 1999.

M. S. Steinhoff, B. Mentzer, P. Geppetti, C. Pothoulakis, and N. W. Bunnett, Tachykinins and their receptors: contributions to physiological control and the mechanisms of disease, Physiol. Rev, vol.94, pp.265-301, 2014.

J. K. Prague, Neurokinin 3 receptor antagonism as a novel treatment for menopausal hot flushes: a phase 2, randomised, double-blind, placebo-controlled trial, Lancet, vol.389, pp.1809-1820, 2017.

R. Saito, Y. Takano, and H. Kamiya, Roles of substance P and NK(1) receptor in the brainstem in the development of emesis, J. Pharmacol. Sci, vol.91, pp.87-94, 2003.

C. Heym, B. Braun, Y. Shuyi, L. Klimaschewski, and M. Colombo-benkmann, Immunohistochemical correlation of human adrenal nerve fibres and thoracic dorsal root neurons with special reference to substance P. Histochem, Cell Biol, vol.104, pp.233-243, 1995.

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, Met-enkephalin, Leu-enkephalin and corticotrophin-releasing hormone on aldosterone secretion in the intact perfused rat adrenal, J. Endocrinol, vol.140, pp.91-96, 1994.

G. Mazzocchi, C. Macchi, L. K. Malendowicz, and G. G. Nussdorfer, Evidence that endogenous substance-P (SP) is involved in the maintenance of the growth and steroidogenic capacity of rat adrenal zona glomerulosa, Neuropeptides, vol.29, pp.53-58, 1995.

J. G. Widdicombe, Autonomic regulation. i-NANC/e-NANC, Am. J. Respir. Crit. Care Med, vol.158, pp.171-175, 1998.

N. M. Page, Characterization of the endokinins: human tachykinins with cardiovascular activity, Proc. Natl. Acad. Sci. U. S. A, vol.100, pp.6245-6250, 2003.

C. E. Gomez-sanchez, Development of monoclonal antibodies against human CYP11B1 and CYP11B2, Mol. Cell. Endocrinol, vol.383, pp.111-117, 2014.

V. Coiro, Stimulation of ACTH/cortisol by intravenously infused substance P in normal men: inhibition by sodium valproate, Neuroendocrinology, vol.56, pp.459-463, 1992.

S. Hubers, S. Wei, and N. Brown, Quantification of substance P in human blood by mass spectroscopy, Hypertension, vol.68, p.180, 2016.

M. Choi, K + channel mutations in adrenal aldosterone-producing adenomas and hereditary hypertension, Science, vol.331, pp.768-772, 2011.

M. Zennaro, S. Boulkroun, and F. Fernandes-rosa, Genetic causes of functional adrenocortical adenomas, Endocr. Rev, vol.38, pp.516-537, 2017.

J. Lai, Differences in the length of the carboxyl terminus mediate functional properties of neurokinin-1 receptor, Proc. Natl. Acad. Sci. U. S. A, vol.105, pp.12605-12610, 2008.

K. Vleugels, ERK1/2 MAPKs and Wnt signaling pathways are independently involved in adipocytokine-mediated aldosterone secretion, Exp. Clin. Endocrinol. Diabetes, vol.119, pp.644-648, 2011.

M. Doi, Salt-sensitive hypertension in circadian clock-deficient Cry-null mice involves dysregulated adrenal Hsd3b6, Nat. Med, vol.16, pp.67-74, 2010.

P. J. Hesketh, Antiemetics: American Society of Clinical Oncology Clinical Practice Guideline Update, J. Clin. Oncol, vol.35, pp.3240-3261, 2017.

G. Bredy-dobreva, D. Zafirov, K. Nieber, P. Oehme, and M. Papasova, Effects of substance P on isolated preparations from rabbit and rat renal arteries, Biomed. Biochim. Acta, vol.47, pp.25-30, 1988.

J. P. Hinson, G. P. Vinson, S. Kapas, and R. Teja, The relationship between adrenal vascular events and steroid secretion: the role of mast cells and endothelin, J. Steroid Biochem. Mol. Biol, vol.40, pp.381-389, 1991.

F. Leboulenger, Immunohistochemical distribution, biochemical characterization, and biological action of tachykinins in the frog adrenal gland, Endocrinology, vol.133, 1993.

H. Lefebvre, 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.

A. K. Majumdar, Pharmacokinetics of aprepitant after single and multiple oral doses in healthy volunteers, J. Clin. Pharmacol, vol.46, pp.291-300, 2006.

M. S. Steinhoff, B. Mentzer, P. Geppetti, C. Pothoulakis, and N. W. Bunnett, Tachykinins and their receptors: contributions to physiological control and the mechanisms of disease, Physiol. Rev, vol.94, pp.265-301, 2014.

H. Lefebvre, 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.

C. Duparc, 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

H. Lefebvre, 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, 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.

J. H. Pratt, A. Ganguly, C. A. Parkinson, and M. H. Weinberger, Stimulation of aldosterone secretion by metoclopramide in humans: apparent independence of renal and pituitary mediation, Metabolism, vol.30, pp.129-134, 1981.

E. P. Bouras, M. Camilleri, D. D. Burton, and S. Mckinzie, Selective stimulation of colonic transit by the benzofuran 5HT4 agonist, prucalopride, in healthy humans, Gut, vol.44, pp.682-686, 1999.

J. E. Levine, An Introduction to Neuroendocrine Systems, Handbook of Neuroendocrinology, pp.3-19, 2012.

M. Haase, H. S. Willenberg, and S. R. Bornstein, Update on the corticomedullary interaction in the adrenal gland, Endocr Dev, vol.20, pp.28-37, 2011.

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

G. P. Vinson, Functional Zonation of the Adult Mammalian Adrenal Cortex, Front Neurosci, vol.10, p.238, 2016.

J. W. Funder, Aldosterone in Advancing Age: Don't Shoot the Messenger. Circulation, vol.136, pp.356-364, 2017.

K. Nanba, A. Vaidya, G. H. Williams, I. Zheng, T. Else et al., Age-Related Autonomous Aldosteronism, vol.136, pp.347-55, 2017.

A. M. Capponi, Regulation of cholesterol supply for mineralocorticoid biosynthesis, Trends Endocrinol Metab TEM, vol.13, issue.3, pp.118-139, 2002.

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, issue.5248, pp.518-538, 1996.

Y. Tsai, W. E. Rainey, and W. B. Bollag, Very low-density lipoprotein (VLDL)-induced signals mediating aldosterone production, J Endocrinol, vol.232, issue.2, pp.115-144, 2017.

W. B. Bollag, Regulation of aldosterone synthesis and secretion, Compr Physiol, vol.4, issue.3, pp.1017-55, 2014.

F. B. Kraemer, Adrenal cholesterol utilization, Mol Cell Endocrinol, pp.42-47, 2007.

W. Shen, S. Azhar, and F. B. Kraemer, Lipid droplets and steroidogenic cells, Exp Cell Res, vol.340, issue.2, pp.209-223, 2016.

Z. S. Krozowski, S. E. Rundle, C. Wallace, M. J. Castell, J. H. Shen et al., Immunolocalization of renal mineralocorticoid receptors with an antiserum against a peptide deduced from the complementary deoxyribonucleic acid sequence, Endocrinology, vol.125, issue.1, pp.192-200, 1989.

B. C. Rossier, O. Staub, and E. Hummler, Genetic dissection of sodium and potassium transport along the aldosterone-sensitive distal nephron: Importance in the control of blood pressure and hypertension, FEBS Lett, vol.587, issue.13, pp.1929-1970, 2013.

M. Colafella, K. M. Danser, and A. , Recent Advances in Angiotensin Research. Hypertens Dallas Tex 1979, vol.69, pp.994-999, 2017.

J. P. Arroyo, C. Ronzaud, D. Lagnaz, O. Staub, and G. Gamba, Aldosterone Paradox: Differential Regulation of Ion Transport in Distal Nephron, Physiology, vol.26, issue.2, pp.115-138, 2011.

R. A. Studer, E. Person, M. Robinson-rechavi, and B. C. Rossier, Evolution of the epithelial sodium channel and the sodium pump as limiting factors of aldosterone action on sodium transport, Physiol Genomics, vol.43, issue.13, pp.844-54, 2011.

S. Viengchareun, L. Menuet, D. Martinerie, L. Munier, M. et al., The mineralocorticoid receptor: insights into its molecular and (patho)physiological biology, Nucl Recept Signal, vol.5, p.12, 2007.
URL : https://hal.archives-ouvertes.fr/inserm-00193515

C. Hellal-levy, J. Fagart, A. Souque, and M. E. Rafestin-oblin, Mechanistic aspects of mineralocorticoid receptor activation, Kidney Int, vol.57, issue.4, pp.1250-1255, 2000.

P. Ferrari and Z. Krozowski, Role of the 11beta-hydroxysteroid dehydrogenase type 2 in blood pressure regulation, Kidney Int, vol.57, issue.4, pp.1374-81, 2000.

B. C. Rossier, M. Bochud, and O. Devuyst, The Hypertension Pandemic: An Evolutionary Perspective. Physiol Bethesda Md, vol.32, pp.112-137, 2017.

P. Pearce and J. W. Funder, High affinity aldosterone binding sites (type I receptors) in rat heart, Clin Exp Pharmacol Physiol, vol.14, pp.859-66, 1987.

A. Gueret, N. Harouki, J. Favre, G. Galmiche, L. Nicol et al., Vascular Smooth Muscle Mineralocorticoid Receptor Contributes to Coronary and Left Ventricular Dysfunction After Myocardial Infarction. Hypertens Dallas Tex 1979, vol.67, pp.717-740, 2016.
URL : https://hal.archives-ouvertes.fr/inserm-02296624

A. P. Davel, I. J. Anwar, and I. Z. Jaffe, The endothelial mineralocorticoid receptor: mediator of the switch from vascular health to disease, Curr Opin Nephrol Hypertens, vol.26, issue.2, pp.97-104, 2017.

J. Yang and M. J. Young, The mineralocorticoid receptor and its coregulators, J Mol Endocrinol, vol.43, issue.2, pp.53-64, 2009.

J. W. Funder, Mineralocorticoid receptors: distribution and activation, Heart Fail Rev, vol.10, issue.1, pp.15-22, 2005.

E. P. Gomez-sanchez, Mineralocorticoid receptors in the brain and cardiovascular regulation: minority rule?, Trends Endocrinol Metab TEM, vol.22, issue.5, pp.179-87, 2011.

D. Lienhard, M. Lauterburg, G. Escher, F. J. Frey, and B. M. Frey, High salt intake down-regulates colonic mineralocorticoid receptors, epithelial sodium channels and 11?-hydroxysteroid dehydrogenase type 2, PloS One, vol.7, issue.5, p.37898, 2012.

R. V. Richardson, E. J. Batchen, M. A. Denvir, G. A. Gray, and K. E. Chapman, Cardiac GR and MR: From Development to Pathology, Trends Endocrinol Metab TEM, vol.27, issue.1, pp.35-43, 2016.

A. Ouvrard-pascaud, Y. Sainte-marie, J. Bénitah, R. Perrier, C. Soukaseum et al., Conditional mineralocorticoid receptor expression in the heart leads to life-threatening arrhythmias, Circulation, vol.111, issue.23, pp.3025-3058, 2005.

B. Pitt, F. Zannad, W. J. Remme, R. Cody, A. Castaigne et al., The effect of spironolactone on morbidity and mortality in patients with severe heart failure. Randomized Aldactone Evaluation Study Investigators, N Engl J Med, vol.341, issue.10, pp.709-726, 1999.

B. Pitt, W. Remme, F. Zannad, J. Neaton, F. Martinez et al., Eplerenone, a selective aldosterone blocker, in patients with left ventricular dysfunction after myocardial infarction, N Engl J Med, vol.348, issue.14, pp.1309-1330, 2003.

F. Zannad, J. Mcmurray, H. Krum, D. J. Van-veldhuisen, K. Swedberg et al., Eplerenone in patients with systolic heart failure and mild symptoms, N Engl J Med, vol.364, issue.1, pp.11-21, 2011.
URL : https://hal.archives-ouvertes.fr/hal-01742988

A. Mccurley, P. W. Pires, S. B. Bender, M. Aronovitz, M. J. Zhao et al., Direct regulation of blood pressure by smooth muscle cell mineralocorticoid receptors, Nat Med, vol.18, issue.9, pp.1429-1462, 2012.

K. Rombouts, A. Wielant, K. Hellemans, D. Schuppan, and A. Geerts, Influence of aldosterone on collagen synthesis and proliferation of rat cardiac fibroblasts, Br J Pharmacol, vol.134, issue.1, pp.224-256, 2001.

S. Bunda, P. Liu, Y. Wang, K. Liu, and A. Hinek, Aldosterone induces elastin production in cardiac fibroblasts through activation of insulin-like growth factor-I receptors in a mineralocorticoid receptorindependent manner, Am J Pathol, vol.171, issue.3, pp.809-828, 2007.

A. Mccurley and I. Z. Jaffe, Mineralocorticoid receptors in vascular function and disease, Mol Cell Endocrinol, vol.350, issue.2, pp.256-65, 2012.

G. Bernini, F. Galetta, F. Franzoni, M. Bardini, C. Taurino et al., Arterial stiffness, intimamedia thickness and carotid artery fibrosis in patients with primary aldosteronism, J Hypertens, 2008.

M. Caprio, B. G. Newfell, A. La-sala, W. Baur, A. Fabbri et al., Functional mineralocorticoid receptors in human vascular endothelial cells regulate intercellular adhesion molecule-1 expression and promote leukocyte adhesion, Circ Res, vol.102, issue.11, pp.1359-67, 2008.

N. G. Hattangady, L. O. Olala, W. B. Bollag, and W. E. Rainey, Acute and chronic regulation of aldosterone production, Mol Cell Endocrinol, vol.350, issue.2, pp.151-62, 2012.

H. Nishimura, Renin-angiotensin system in vertebrates: phylogenetic view of structure and function, Anat Sci Int, vol.92, issue.2, pp.215-262, 2017.

R. A. Gomez, Fate of Renin Cells During Development and Disease, vol.69, pp.387-95, 2017.

C. N. Young and R. L. Davisson, Angiotensin-II, the Brain, and Hypertension: An Update. Hypertens Dallas Tex 1979, vol.66, pp.920-926, 2015.

M. Feuilloley and H. Vaudry, Role of the cytoskeleton in adrenocortical cells, Endocr Rev, vol.17, issue.3, pp.269-88, 1996.

N. Cherradi, B. Pardo, A. S. Greenberg, F. B. Kraemer, and A. M. Capponi, Angiotensin II Activates Cholesterol Ester Hydrolase in Bovine Adrenal Glomerulosa Cells through Phosphorylation Mediated by p42/p44
URL : https://hal.archives-ouvertes.fr/hal-02459083

, Mitogen-Activated Protein Kinase. Endocrinology, vol.144, issue.11, pp.4905-4920, 2003.

A. Fleury, A. P. Mathieu, L. Ducharme, D. B. Hales, and J. Lehoux, Phosphorylation and function of the hamster adrenal steroidogenic acute regulatory protein (StAR), J Steroid Biochem Mol Biol, 2004.

W. B. Bollag, Regulation of Aldosterone Synthesis and Secretion

N. J. Hoboken and . Usa, , pp.1017-55, 2014.

L. Brizuela, M. Rábano, P. Gangoiti, N. Narbona, J. M. Macarulla et al., Sphingosine-1-phosphate stimulates aldosterone secretion through a mechanism involving the PI3K/PKB and MEK/ERK 1/2 pathways, J Lipid Res, vol.48, issue.10, pp.2264-74, 2007.

M. F. Rossier, T channels and steroid biosynthesis: in search of a link with mitochondria, Cell Calcium, vol.40, issue.2, pp.155-64, 2006.

A. Spät, L. Fülöp, and G. Szanda, The role of mitochondrial Ca(2+) and NAD(P)H in the control of aldosterone secretion, Cell Calcium, vol.52, issue.1, pp.64-72, 2012.

N. Cherradi, Y. Brandenburger, and A. M. Capponi, Mitochondrial regulation of mineralocorticoid biosynthesis by calcium and the StAR protein, Eur J Endocrinol, vol.139, issue.3, pp.249-256, 1998.
URL : https://hal.archives-ouvertes.fr/hal-02459010

T. Ota, M. Doi, F. Yamazaki, D. Yarimizu, K. Okada et al., Angiotensin II Triggers Expression of the Adrenal Gland Zona Glomerulosa-Specific 3?-Hydroxysteroid Dehydrogenase Isoenzyme through De Novo Protein Synthesis of the Orphan Nuclear Receptors NGFIB and NURR1, Mol Cell Biol, vol.34, issue.20, pp.3880-94, 2014.

J. O. Davis, J. Urquhart, and J. T. Higgins, The effects of alteration of plasma sodium and potassium concentration on aldosterone secretion, J Clin Invest, vol.42, pp.597-609, 1963.

E. Mulkerrin, F. H. Epstein, and B. A. Clark, Aldosterone responses to hyperkalemia in healthy elderly humans, J Am Soc Nephrol JASN, vol.6, issue.5, pp.1459-62, 1995.

V. N. Uebele, C. E. Nuss, J. J. Renger, and T. M. Connolly, Role of voltage-gated calcium channels in potassiumstimulated aldosterone secretion from rat adrenal zona glomerulosa cells, J Steroid Biochem Mol Biol, vol.92, issue.3, pp.209-227, 2004.

A. Spät and L. Hunyady, Control of aldosterone secretion: a model for convergence in cellular signaling pathways, Physiol Rev, vol.84, issue.2, pp.489-539, 2004.

M. Choi, U. I. Scholl, P. Yue, P. Björklund, B. Zhao et al., K+ channel mutations in adrenal aldosterone-producing adenomas and hereditary hypertension, Science, vol.331, issue.6018, pp.768-72, 2011.

C. R. Jefcoate, J. Lee, N. Cherradi, H. Takemori, and H. Duan, cAMP stimulation of STAR expression and cholesterol metabolism is modulated by co-expression of labile suppressors of transcription and mRNA turnover, Mol Cell Endocrinol, vol.336, issue.1-2, pp.53-62, 2011.
URL : https://hal.archives-ouvertes.fr/hal-02459115

P. R. Manna, D. W. Eubank, E. Lalli, P. Sassone-corsi, and D. M. Stocco, Transcriptional regulation of the mouse steroidogenic acute regulatory protein gene by the cAMP response-element binding protein and steroidogenic factor 1, J Mol Endocrinol, vol.30, issue.3, pp.381-397, 2003.

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, issue.1-2, pp.87-94, 2001.

A. Nehme, C. Cerutti, N. Dhaouadi, M. P. Gustin, P. Courand et al., Atlas of tissue reninangiotensin-aldosterone system in human: A transcriptomic meta-analysis. Sci Rep, vol.5, p.10035, 2015.

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, issue.2, pp.101-144, 1998.

E. Lalli, J. Barhanin, M. Zennaro, and R. Warth, Local Control of Aldosterone Production and Primary Aldosteronism, Trends Endocrinol Metab TEM, vol.27, issue.3, pp.123-154, 2016.

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, issue.2, pp.101-144, 1998.

M. Bader, R. W. Holz, K. Kumakura, and N. Vitale, Exocytosis: the chromaffin cell as a model system, Ann N Y Acad Sci, vol.971, pp.178-83, 2002.

M. B. Khan, B. R. Lee, and T. Kamitani, A simple and sensitive method for the demonstration of norepinephrine-storing adrenomedullary chromaffin cells, Histochem Cell Biol, vol.138, issue.1, pp.155-65, 2012.

, Tietz fundamentals of clinical chemistry, vol.6

. St and . Louis, Miss: Saunders Elsevier, vol.952, 2008.

S. Nussey and S. Whitehead, The adrenal gland

, BIOS Scientific Publishers, 2001.

S. R. Bornstein, M. Ehrhart-bornstein, H. Usadel, M. Böckmann, and W. A. Scherbaum, Morphological evidence for a close interaction of chromaffin cells with cortical cells within the adrenal gland, Cell Tissue Res, vol.265, issue.1, pp.1-9, 1991.

S. R. Bornstein, M. Ehrhart-bornstein, W. A. Scherbaum, E. F. Pfeiffer, and J. J. Holst, Effects of splanchnic nerve stimulation on the adrenal cortex may be mediated by chromaffin cells in a paracrine manner, Endocrinology, vol.127, issue.2, pp.900-906, 1990.

H. Lefebvre, M. Thomas, C. Duparc, J. Bertherat, and E. Louiset, Role of ACTH in the Interactive/Paracrine Regulation of Adrenal Steroid Secretion in Physiological and Pathophysiological Conditions, Front Endocrinol, vol.7, p.98, 2016.
URL : https://hal.archives-ouvertes.fr/inserm-01354821

S. Schinner and S. R. Bornstein, Cortical-chromaffin cell interactions in the adrenal gland, Endocr Pathol, vol.16, issue.2, pp.91-99, 2005.

G. G. Nussdorfer, Paracrine control of adrenal cortical function by medullary chromaffin cells, Pharmacol Rev, vol.48, issue.4, pp.495-530, 1996.

H. Lefebvre, G. Prévost, and E. Louiset, Autocrine/paracrine regulatory mechanisms in adrenocortical neoplasms responsible for primary adrenal hypercorticism, Eur J Endocrinol, vol.169, issue.5, pp.115-138, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01946266

A. Evrard, C. Hober, A. Racadot, J. Lefebvre, and M. C. Vantyghem,

, Ann Biol Clin, vol.57, issue.2, pp.149-55, 1999.

A. Ganguly, Atrial natriuretic peptide-induced inhibition of aldosterone secretion: a quest for mediator(s), Am J Physiol, vol.263, issue.2, pp.181-194, 1992.

R. Kvetnanský, K. Pacák, K. Fukuhara, E. Viskupic, B. Hiremagalur et al., Sympathoadrenal system in stress. Interaction with the hypothalamic-pituitary-adrenocortical system, Ann N Y Acad Sci, vol.771, pp.131-58, 1995.

D. Cat, A. N. Friederich-persson, M. White, A. Touyz, and R. M. , Adipocytes, aldosterone and obesity-related hypertension, J Mol Endocrinol, vol.57, issue.1, pp.7-21, 2016.

S. Schinner, H. S. Willenberg, D. Krause, M. Schott, V. Lamounier-zepter et al., Adipocytederived 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, issue.5, pp.864-70, 2005.

M. Ehrhart-bornstein, K. Arakelyan, A. W. Krug, W. A. Scherbaum, and S. R. Bornstein, Fat cells may be the obesity-hypertension link: human adipogenic factors stimulate aldosterone secretion from adrenocortical cells, Endocr Res, vol.30, issue.4, pp.865-70, 2004.

V. Lamounier-zepter, M. Ehrhart-bornstein, and S. R. Bornstein, Mineralocorticoid-stimulating activity of adipose tissue, Best Pract Res Clin Endocrinol Metab, vol.19, issue.4, pp.567-75, 2005.

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, vol.100, issue.24, pp.14211-14217, 2003.

B. Vogt, M. Bochud, and M. Burnier, The association of aldosterone with obesity-related hypertension and the metabolic syndrome, Semin Nephrol, vol.27, issue.5, pp.529-566, 2007.

S. R. Bornstein, H. Rutkowski, and I. Vrezas, Cytokines and steroidogenesis, Mol Cell Endocrinol, vol.215, issue.1-2, pp.135-176, 2004.

C. Marx, S. R. Bornstein, G. W. Wolkersdörfer, M. Peter, W. G. Sippell et al., Relevance of major histocompatibility complex class II expression as a hallmark for the cellular differentiation in the human adrenal cortex, J Clin Endocrinol Metab, vol.82, issue.9, pp.3136-3176, 1997.

Y. Hayashi, T. Hiyoshi, T. Takemura, C. Kurashima, and K. Hirokawa, Focal lymphocytic infiltration in the adrenal cortex of the elderly: immunohistological analysis of infiltrating lymphocytes, Clin Exp Immunol, vol.77, issue.1, pp.101-106, 1989.

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, issue.1, pp.137-178, 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, issue.5, pp.1492-1499, 1994.

J. A. González-hernández, M. Ehrhart-bornstein, E. Späth-schwalbe, W. A. Scherbaum, and S. R. Bornstein, Human adrenal cells express tumor necrosis factor-alpha messenger ribonucleic acid: evidence for paracrine control of adrenal function, J Clin Endocrinol Metab, vol.81, issue.2, pp.807-820, 1996.

G. Päth, S. R. Bornstein, M. Ehrhart-bornstein, and W. A. Scherbaum, Interleukin-6 and the interleukin-6 receptor in the human adrenal gland: expression and effects on steroidogenesis, J Clin Endocrinol Metab, vol.82, issue.7, pp.2343-2352, 1997.

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, issue.6, pp.3084-3093, 1989.

A. M. Gilfillan and C. Tkaczyk, Integrated signalling pathways for mast-cell activation, Nat Rev Immunol, vol.6, issue.3, pp.218-248, 2006.

T. C. Theoharides, P. Valent, and C. Akin, Mast Cells, Mastocytosis, and Related Disorders, N Engl J Med, vol.373, issue.2, pp.163-72, 2009.

J. P. Hinson, G. P. Vinson, J. Pudney, and B. J. Whitehouse, Adrenal mast cells modulate vascular and secretory responses in the intact adrenal gland of the rat, J Endocrinol, vol.121, issue.2, pp.253-60, 1989.

S. Wernersson and G. Pejler, Mast cell secretory granules: armed for battle, Nat Rev Immunol, vol.14, issue.7, pp.478-94, 2014.

T. C. Theoharides, D. Kempuraj, M. Tagen, P. Conti, and D. Kalogeromitros, Differential release of mast cell mediators and the pathogenesis of inflammation, Immunol Rev, vol.217, pp.65-78, 2007.

L. Tang, T. A. Jennings, and J. W. Eaton, Mast cells mediate acute inflammatory responses to implanted biomaterials, Proc Natl Acad Sci, vol.95, issue.15, pp.8841-8847, 1998.

U. Lippert, M. Artuc, A. Grützkau, M. Babina, S. Guhl et al., Human skin mast cells express H2 and H4, but not H3 receptors, J Invest Dermatol, vol.123, issue.1, pp.116-139, 2004.

A. R. Johnson and E. G. Erdös, Release of histamine from mast cells by vasoactive peptides, Proc Soc Exp Biol Med Soc Exp Biol Med N Y N, vol.142, issue.4, pp.1252-1258, 1973.

W. Li, T. Guo, D. Liang, Y. Sun, W. S. Kingery et al., Substance P signaling controls mast cell activation, degranulation, and nociceptive sensitization in a rat fracture model of complex regional pain syndrome, Anesthesiology, vol.116, issue.4, pp.882-95, 2012.

C. A. Maggi, The effects of tachykinins on inflammatory and immune cells, Regul Pept, vol.70, issue.2-3, pp.75-90, 1997.

T. C. Theoharides, Neuroendocrinology of mast cells: Challenges and controversies, Exp Dermatol, 2017.

D. Ribatti, The development of human mast cells. An historical reappraisal, Exp Cell Res, vol.342, issue.2, pp.210-215, 2016.

M. L. Linenberger, F. W. Jacobson, L. G. Bennett, V. C. Broudy, F. H. Martin et al., Stem cell factor production by human marrow stromal fibroblasts, Exp Hematol, vol.23, issue.10, pp.1104-1118, 1995.

V. C. Broudy, N. L. Kovach, L. G. Bennett, N. Lin, F. W. Jacobsen et al., Human umbilical vein endothelial cells display high-affinity c-kit receptors and produce a soluble form of the c-kit receptor, Blood, 1994.

, Apr, vol.15, issue.8, pp.2145-52

J. S. Kim, H. Kubota, S. Y. Nam, K. Doi, and J. Saegusa, Expression of cytokines and proteases in mast cells in the lesion of subcapsular cell hyperplasia in mouse adrenal glands, Toxicol Pathol, vol.28, issue.2, pp.297-303, 2000.

M. Welle, Development, significance, and heterogeneity of mast cells with particular regard to the mast cell-specific proteases chymase and tryptase, J Leukoc Biol, vol.61, issue.3, pp.233-278, 1997.

G. H. Caughey, W. W. Raymond, and P. J. Wolters, Angiotensin II generation by mast cell alpha-and betachymases, Biochim Biophys Acta, vol.1480, issue.1-2, pp.245-57, 2000.

C. J. Mackins, S. Kano, N. Seyedi, U. Schäfer, A. C. Reid et al., Cardiac mast cell-derived renin promotes local angiotensin formation, norepinephrine release, and arrhythmias in ischemia/reperfusion, J Clin Invest, vol.116, issue.4, pp.1063-70, 2006.

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, issue.4, pp.550-560, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01544140

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.5001-5008, 2001.

H. Lefebvre, D. Cartier, C. Duparc, V. Contesse, I. Lihrmann et al., Effect of serotonin4 (5-HT4) receptor agonists on aldosterone secretion in idiopathic hyperaldosteronism, Endocr Res, vol.26, issue.4, pp.583-590, 2000.
URL : https://hal.archives-ouvertes.fr/hal-01928526

, , 2017.

G. P. Rossi, A. Sacchetto, M. Cesari, and A. C. Pessina, Interactions between endothelin-1 and the reninangiotensin-aldosterone system, Cardiovasc Res, vol.43, issue.2, pp.300-307, 1999.

G. Mazzocchi, L. K. Malendowicz, F. G. Musajo, G. Gottardo, A. Markowska et al., Role of endothelins in regulation of vascular tone in the in situ perfused rat adrenals, Am J Physiol, 1998.

G. P. Rossi, M. Cavallin, G. G. Nussdorfer, and A. C. Pessina, The endothelin-aldosterone axis and cardiovascular diseases, J Cardiovasc Pharmacol, vol.38, issue.2, pp.49-52, 2001.

P. G. Kopf, K. M. Gauthier, D. X. Zhang, J. R. Falck, and W. B. Campbell, Angiotensin II regulates adrenal vascular tone through zona glomerulosa cell-derived EETs and DHETs, Hypertens Dallas Tex, 1979.

, Feb, vol.57, issue.2, pp.323-332

D. X. Zhang, K. M. Gauthier, J. R. Falck, A. Siddam, and W. B. Campbell, Steroid-producing cells regulate arterial tone of adrenal cortical arteries, Endocrinology, vol.148, issue.8, pp.3569-76, 2007.

D. Curro, V. Ipavec, and P. Preziosi, Neurotransmitters of the non-adrenergic non-cholinergic relaxation of proximal stomach, Eur Rev Med Pharmacol Sci, vol.12, issue.1, pp.53-62, 2008.

T. W. Mackay, G. Hulks, and N. J. Douglas, Non-adrenergic, non-cholinergic function in the human airway, Respir Med, vol.92, issue.3, pp.461-467, 1998.

G. Burnstock, Autonomic Neurotransmission: 60 Years Since Sir Henry Dale, Annu Rev Pharmacol Toxicol, vol.49, issue.1, pp.1-30, 2009.

P. J. Barnes, The third nervous system in the lung : physiology and clinical perspectives, 2017.

B. G. Katzung, Basic & clinical pharmacology, 2012.

G. F. Joos, P. R. Germonpré, and R. A. Pauwels, Neural mechanisms in asthma, Clin Exp Allergy J Br Soc Allergy Clin Immunol, vol.30, issue.1, pp.60-65, 2000.

J. G. Widdicombe, Autonomic Regulation, Am J Respir Crit Care Med, vol.158, issue.supplement_2, pp.171-176, 1998.

S. Offermanns and W. Rosenthal, Encyclopedia of molecular pharmacology, 2008.

R. Tomita, Y. Kurosu, and K. Munakata, Relationship between nitric oxide and non-adrenergic noncholinergic inhibitory nerves in human lower esophageal sphincter, J Gastroenterol, 1997.

G. Burnstock, Physiology and pathophysiology of purinergic neurotransmission, Physiol Rev, vol.87, issue.2, pp.659-797, 2007.

J. M. Lundberg, Pharmacology of cotransmission in the autonomic nervous system: integrative aspects on amines, neuropeptides, adenosine triphosphate, amino acids and nitric oxide, Pharmacol Rev, vol.48, issue.1, pp.113-78, 1996.

P. Huidobro-toro, J. , V. Donoso, and M. , Sympathetic co-transmission: the coordinated action of ATP and noradrenaline and their modulation by neuropeptide Y in human vascular neuroeffector junctions, Eur J Pharmacol, vol.500, issue.1-3, pp.27-35, 2004.

J. M. Lundberg, A. Änggärd, J. Fahrenkrug, T. Hökfelt, and V. Mutt, Vasoactive intestinal polypeptide in cholinergic neurons of exocrine glands: Functional significance of coexisting transmitters for vasodilation and secretion, Proc Natl Acad Sci, vol.77, issue.3, pp.1651-1656, 1980.

T. L. Parker, W. K. Kesse, A. A. Mohamed, and M. Afework, The innervation of the mammalian adrenal gland, J Anat, vol.183, issue.2, pp.265-76, 1993.

K. Unsicker, On the innervation of the rat and pig adrenal cortex. Z Für Zellforsch Mikrosk Anat, vol.116, pp.151-157, 1971.

P. M. Robinson, R. A. Perry, K. J. Hardy, J. P. Coghlan, and B. A. Scoggins, The innervation of the adrenal cortex in the sheep, Ovis ovis, J Anat, vol.124, pp.117-146, 1977.

T. L. Parker, A. A. Mohamed, and R. E. Coupland, The innervation of the adrenal gland. IV. The source of preand postganglionic nerve fibres to the guinea-pig adrenal gland, J Anat, vol.172, pp.17-24, 1990.

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, issue.2, pp.101-143, 1998.

A. M. Mcnicol, J. Richmond, and B. G. Charlton, A study of general innervation of the human adrenal cortex using PGP 9.5 immunohistochemistry, Acta Anat (Basel), vol.151, issue.2, pp.120-123, 1994.

B. G. Charlton, J. Mcgadey, D. Russell, and D. E. Neal, Noradrenergic innervation of the human adrenal cortex as revealed by dopamine-beta-hydroxylase immunohistochemistry, J Anat, vol.180, pp.501-507, 1992.

B. G. Charlton, O. F. Nkomazana, J. Mcgadey, and D. E. Neal, A preliminary study of acetylcholinesterasepositive innervation in the human adrenal cortex, J Anat, vol.176, pp.99-104, 1991.

A. B. Gilchrist, A. Leake, and B. G. Charlton, Innervation of the human adrenal cortex: simultaneous visualisation using acetylcholinesterase histochemistry and dopamine beta-hydroxylase immunohistochemistry, Acta Anat (Basel), vol.146, issue.1, pp.31-36, 1993.

H. Kondo, Immunohistochemical analysis of the localization of neuropeptides in the adrenal gland, Arch Histol Jpn Nihon Soshikigaku Kiroku, vol.48, issue.5, pp.453-81, 1985.

A. B. Gilchrist, A. Leake, and B. G. Charlton, Innervation of the human adrenal cortex: simultaneous visualisation using acetylcholinesterase histochemistry and dopamine beta-hydroxylase immunohistochemistry, Acta Anat (Basel), vol.146, issue.1, pp.31-36, 1993.

A. Souvatzoglou, The Sympathoadrenal System: Integrative Regulation of the Cortical and the

, Adrenal Glands: Diagnostic Aspects and Surgical Therapy

. Springer, , 2005.

, , pp.33-39

W. C. Engeland, Functional innervation of the adrenal cortex by the splanchnic nerve, Horm Metab Res, vol.30, pp.311-314, 1998.

B. G. Charlton, J. Mcgadey, D. Russell, and D. E. Neal, Noradrenergic innervation of the human adrenal cortex as revealed by dopamine-beta-hydroxylase immunohistochemistry, J Anat, vol.180, pp.501-507, 1992.

I. E. Tóth, E. S. Vizi, J. P. Hinson, and G. P. Vinson, Innervation of the adrenal cortex, its physiological relevance, with primary focus on the noradrenergic transmission, Microsc Res Tech, vol.36, issue.6, pp.534-579, 1997.

W. K. Kesse, T. L. Parker, and R. E. Coupland, The innervation of the adrenal gland. I. The source of pre-and postganglionic nerve fibres to the rat adrenal gland, J Anat, vol.157, pp.33-41, 1988.

B. G. Charlton, O. F. Nkomazana, J. Mcgadey, and D. E. Neal, A preliminary study of acetylcholinesterasepositive innervation in the human adrenal cortex, J Anat, vol.176, pp.99-104, 1991.

W. C. Engeland, Functional innervation of the adrenal cortex by the splanchnic nerve. Horm Metab Res Horm Stoffwechselforschung Horm Métabolisme, vol.30, pp.311-315, 1998.

Y. M. Ulrich-lai and W. C. Engeland, Hyperinnervation during adrenal regeneration influences the rate of functional recovery, Neuroendocrinology, vol.71, issue.2, pp.107-130, 2000.

S. R. Bornstein, W. C. Engeland, M. Ehrhart-bornstein, and J. P. Herman, Dissociation of ACTH and glucocorticoids, Trends Endocrinol Metab TEM, vol.19, issue.5, pp.175-80, 2008.

R. M. Buijs, J. Wortel, J. J. Van-heerikhuize, M. G. Feenstra, T. Horst et al., Anatomical and functional demonstration of a multisynaptic suprachiasmatic nucleus adrenal (cortex) pathway, Eur J Neurosci, vol.11, issue.5, pp.1535-1579, 1999.

A. Saria, S. P. Wilson, A. Molnar, O. H. Viveros, and F. Lembeck, Substance P and opiate-like peptides in human adrenal medulla, Neurosci Lett, vol.20, issue.2, pp.195-200, 1980.

R. I. Linnoila, R. P. Diaugustine, A. Hervonen, and R. J. Miller, Distribution of [Met5]-and [Leu5]-enkephalin-, vasoactive intestinal polypeptide-and substance P-like immunoreactivities in human adrenal glands, Neuroscience, vol.5, issue.12, pp.2247-59, 1980.

C. Heym, B. Braun, Y. Shuyi, L. Klimaschewski, and M. Colombo-benkmann, Immunohistochemical correlation of human adrenal nerve fibres and thoracic dorsal root neurons with special reference to substance P, Histochem Cell Biol, vol.104, issue.3, pp.233-276, 1995.

M. Colombo-benkmann, L. Klimaschewski, and C. Heym, Immunohistochemical heterogeneity of nerve cells in the human adrenal gland with special reference to substance P, J Histochem Cytochem, vol.44, issue.4, pp.369-75, 1996.

-. Basic-neurochemistry and . Bookshelf, What are neuropeptides?, Methods Mol Biol Clifton NJ, vol.789, pp.1-36, 2011.

H. Satake, M. Ogasawara, T. Kawada, K. Masuda, M. Aoyama et al., Tachykinin and tachykinin receptor of an ascidian, Ciona intestinalis: evolutionary origin of the vertebrate tachykinin family, J Biol Chem, vol.279, issue.51, pp.53798-805, 2004.

H. Satake, T. Kawada, K. Nomoto, and H. Minakata, Insight into tachykinin-related peptides, their receptors, and invertebrate tachykinins: a review, Zoolog Sci, vol.20, issue.5, pp.533-582, 2003.

T. Van-loy, H. P. Vandersmissen, J. Poels, M. B. Van-hiel, H. Verlinden et al., Tachykinin-related peptides and their receptors in invertebrates: a current view, Peptides, vol.31, issue.3, pp.520-524, 2010.

H. Satake, M. Aoyama, T. Sekiguchi, and T. Kawada, Insight into molecular and functional diversity of tachykinins and their receptors, Protein Pept Lett, vol.20, issue.6, pp.615-642, 2013.

W. Zhou, S. Li, Y. Liu, X. Qi, H. Chen et al., The evolution of tachykinin/tachykinin receptor (TAC/TACR) in vertebrates and molecular identification of the TAC3/TACR3 system in zebrafish (Danio rerio), Mol Cell Endocrinol, vol.361, issue.1-2, pp.202-214, 2012.

H. Nawa, H. Kotani, and S. Nakanishi, Tissue-specific generation of two preprotachykinin mRNAs from one gene by alternative RNA splicing, Nature, vol.312, issue.5996, pp.729-763, 1984.

A. J. Harmar, A. Armstrong, J. C. Pascall, K. Chapman, R. R. Curtis et al., cDNA sequence of human beta-preprotachykinin, the common precursor to substance P and neurokinin A, FEBS Lett, vol.208, issue.1, pp.67-72, 1986.

J. E. Krause, J. M. Chirgwin, M. S. Carter, Z. S. Xu, and A. D. Hershey, Three rat preprotachykinin mRNAs encode the neuropeptides substance P and neurokinin A, Proc Natl Acad Sci, vol.84, issue.3, pp.881-886, 1987.

M. G. Butler, T. A. Nelson, D. J. Driscoll, A. M. Manzardo, . Evaluation et al., , vol.3, 2015.

A. H. Amin, T. 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, issue.3, pp.596-618, 1954.

M. M. Chang, S. E. Leeman, and H. D. Niall, Amino-acid sequence of substance P, Nature New Biol, vol.232, issue.29, pp.86-93, 1971.

T. M. Fong, S. A. Anderson, H. Yu, R. R. Huang, and C. D. Strader, Differential activation of intracellular effector by two isoforms of human neurokinin-1 receptor, Mol Pharmacol, vol.41, issue.1, pp.24-30, 1992.

R. M. Snider, J. W. Constantine, J. A. Lowe, K. P. Longo, W. S. Lebel et al., A potent nonpeptide antagonist of the substance P (NK1) receptor. Science, vol.251, pp.435-442, 1991.

M. Otsuka and K. Yoshioka, Neurotransmitter functions of mammalian tachykinins, Physiol Rev, vol.73, issue.2, pp.229-308, 1993.

M. Okumura, R. Arakawa, H. Ito, C. Seki, H. Takahashi et al., Quantitative analysis of NK1 receptor in the human brain using PET with 18F-FE-SPA-RQ, J Nucl Med Off Publ Soc Nucl Med, 2008.

R. D. Helme and K. Thomas, Substance P in human hypothalamus, Clin Exp Neurol, vol.22, pp.97-101, 1986.

C. Fergani, L. Mazzella, L. M. Coolen, R. B. Mccosh, S. L. Hardy et al., Do Substance P and Neurokinin A Play Important Roles in the Control of LH Secretion in Ewes? Endocrinology, 2016.

V. Coiro, L. Capretti, R. Volpi, C. Davoli, A. Marcato et al., Stimulation of ACTH/cortisol by intravenously infused substance P in normal men: inhibition by sodium valproate, Neuroendocrinology, vol.56, issue.4, pp.459-63, 1992.

E. Mistrova, P. Kruzliak, C. Dvorakova, and M. , Role of substance P in the cardiovascular system, Neuropeptides, vol.58, pp.41-51, 2016.

M. Reinecke, E. Weihe, and W. G. Forssmann, Substance P-immunoreactive nerve fibers in the heart, Neurosci Lett, vol.20, issue.3, pp.265-274, 1980.

D. E. Newby, D. G. Sciberras, C. J. Ferro, B. J. Gertz, D. Sommerville et al., Substance P-induced vasodilatation is mediated by the neurokinin type 1 receptor but does not contribute to basal vascular tone in man, Br J Clin Pharmacol, vol.48, issue.3, pp.336-380, 1999.

D. E. Newby, D. G. Sciberras, C. M. Mendel, B. J. Gertz, N. A. Boon et al., Intra-arterial substance P mediated vasodilatation in the human forearm: pharmacology, reproducibility and tolerability, Br J Clin Pharmacol, vol.43, issue.5, pp.493-502, 1997.

D. C. Crossman, S. W. Larkin, R. W. Fuller, G. J. Davies, and A. Maseri, Substance P dilates epicardial coronary arteries and increases coronary blood flow in humans, Circulation, vol.80, issue.3, pp.475-84, 1989.

M. Okumura, R. Arakawa, H. Ito, C. Seki, H. Takahashi et al., Quantitative Analysis of NK1 Receptor in the Human Brain Using PET with 18F-FE-SPA-RQ, J Nucl Med, vol.49, issue.11, pp.1749-55, 2008.

P. Ducrotté, Constipation sous opioïdes : physiopathologie et prise en charge actuelle. Hépato-Gastro Oncol Dig, vol.17, pp.48-56, 2010.

M. S. Steinhoff, V. Mentzer, B. Geppetti, P. Pothoulakis, C. Bunnett et al., Tachykinins and their receptors: contributions to physiological control and the mechanisms of disease, Physiol Rev, 2014.

P. Holzer and U. Holzer-petsche, Roles in neural excitation, secretion and inflammation, Tachykinins in the gut. Part II, vol.73, pp.219-63, 1997.

D. A. Groneberg, S. Harrison, Q. T. Dinh, P. Geppetti, and A. Fischer, Tachykinins in the respiratory tract, Curr Drug Targets, vol.7, issue.8, pp.1005-1015, 2006.

S. Amadesi, J. Moreau, M. Tognetto, J. Springer, M. Trevisani et al., NK1 receptor stimulation causes contraction and inositol phosphate increase in medium-size human isolated bronchi, Am J Respir Crit Care Med, vol.163, issue.5, pp.1206-1217, 2001.

O. Lesouhaitier, M. Esneu, M. K. Kodjo, C. Hamel, V. Contesse et al., Neuroendocrine communication in the frog adrenal gland, Zoolog Sci, vol.12, issue.3, pp.255-64, 1995.
URL : https://hal.archives-ouvertes.fr/hal-02334690

F. Leboulenger, L. Vaglini, J. M. Conlon, F. Homo-delarche, Y. Wang et al.,

, Immunohistochemical distribution, biochemical characterization, and biological action of tachykinins in the frog adrenal gland, Endocrinology, vol.133, issue.5, pp.1999-2008, 1993.

H. Murabayashi, H. Kuramoto, H. Kawano, M. Sasaki, N. Kitamura et al., Immunohistochemical features of substance P-immunoreactive chromaffin cells and nerve fibers in the rat adrenal gland, Arch Histol Cytol, vol.70, issue.3, pp.183-96, 2007.

E. J. Whitworth, O. Kosti, D. Renshaw, and J. P. Hinson, Adrenal neuropeptides: regulation and interaction with ACTH and other adrenal regulators, Microsc Res Tech, vol.61, issue.3, pp.259-67, 2003.

F. Leboulenger, L. Vaglini, J. M. Conlon, F. Homo-delarche, Y. Wang et al.,

, Immunohistochemical distribution, biochemical characterization, and biological action of tachykinins in the frog adrenal gland, Endocrinology, vol.133, issue.5, pp.1999-2008, 1993.

G. G. Nussdorfer, L. K. Malendowicz, A. S. Belloni, G. Mazzocchi, and P. Rebuffat, Effects of substance P on the rat adrenal zona glomerulosa in vivo, Peptides, vol.9, issue.5, pp.1145-1154, 1988.

A. J. Mitchell, A. M. Lone, A. D. Tinoco, and A. Saghatelian, Proteolysis controls endogenous substance P levels, PloS One, vol.8, issue.7, p.68638, 2013.

M. A. Cascieri, H. G. Bull, R. A. Mumford, A. A. Patchett, N. A. Thornberry et al., Carboxyl-terminal tripeptidyl hydrolysis of substance P by purified rabbit lung angiotensin-converting enzyme and the potentiation of substance P activity in vivo by captopril and MK-422, Mol Pharmacol, vol.25, issue.2, pp.287-93, 1984.

R. A. Skidgel and E. G. Erdös, Angiotensin converting enzyme (ACE) and neprilysin hydrolyze neuropeptides: a brief history, the beginning and follow-ups to early studies, Peptides, vol.25, issue.3, pp.521-526, 2004.

F. Pailleux, J. Lemoine, and F. Beaudry, Investigation of the metabolic biotransformation of substance P in liver microsomes by liquid chromatography quadrupole ion trap mass spectrometry, Biomed Chromatogr BMC, vol.27, issue.1, pp.39-47, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00875709

M. Hallberg, Neuropeptides: metabolism to bioactive fragments and the pharmacology of their receptors, Med Res Rev, vol.35, issue.3, pp.464-519, 2015.

, The role of neurokinin B signalling in reproductive neuroendocrinology

N. M. Page, R. J. Woods, and P. J. Lowry, A regulatory role for neurokinin B in placental physiology and preeclampsia, Regul Pept, vol.98, issue.3, pp.97-104, 2001.

F. M. Pinto, T. A. Almeida, M. Hernandez, P. Devillier, C. Advenier et al., mRNA expression of tachykinins and tachykinin receptors in different human tissues, Eur J Pharmacol, vol.494, issue.2-3, pp.233-242, 2004.

, Characterization of the endokinins: human tachykinins with cardiovascular activity. -PubMed -NCBI, 2017.

Y. Zhang, L. Lu, C. Furlonger, G. E. Wu, and C. J. Paige, Hemokinin is a hematopoietic-specific tachykinin that regulates B lymphopoiesis, Nat Immunol, vol.1, issue.5, pp.392-399, 2000.

N. M. Page, Characterization of the gene structures, precursor processing and pharmacology of the endokinin peptides, Vascul Pharmacol, vol.45, issue.4, pp.200-208, 2006.

N. M. Page, Hemokinins and endokinins, Cell Mol Life Sci CMLS, vol.61, issue.13, pp.1652-63, 2004.

S. P. Alexander, A. P. Davenport, E. Kelly, N. Marrion, J. A. Peters et al., The Concise Guide to PHARMACOLOGY 2015/16: G protein-coupled receptors, Br J Pharmacol, vol.172, issue.24, pp.5744-869, 2015.

S. Garcia-recio and P. Gascón, Biological and Pharmacological Aspects of the NK1-Receptor, BioMed Res Int, p.495704, 2015.

R. Kage, S. E. Leeman, and N. D. Boyd, Biochemical characterization of two different forms of the substance P receptor in rat submaxillary gland, J Neurochem, vol.60, issue.1, pp.347-51, 1993.

L. Caberlotto, Y. L. Hurd, P. Murdock, J. P. Wahlin, S. Melotto et al., Neurokinin 1 receptor and relative abundance of the short and long isoforms in the human brain, Eur J Neurosci, 2003.

M. Bensaid, B. A. Faucheux, E. Hirsch, Y. Agid, P. Soubrié et al., Expression of tachykinin NK2 receptor mRNA in human brain, Neurosci Lett, vol.303, issue.1, pp.25-33, 2001.

N. P. Gerard, R. L. Eddy, T. B. Shows, and C. Gerard, The human neurokinin A (substance K) receptor. Molecular cloning of the gene, chromosome localization, and isolation of cDNA from tracheal and gastric tissues, J Biol Chem, vol.265, issue.33, pp.20455-62, 1990.

D. B. Hoover, Y. Chang, J. C. Hancock, and L. Zhang, Actions of tachykinins within the heart and their relevance to cardiovascular disease, Jpn J Pharmacol, vol.84, issue.4, pp.367-73, 2000.

H. Mechiche, S. Grassin-delyle, F. M. Pinto, A. Buenestado, L. Candenas et al., Smooth muscle neurokinin-2 receptors mediate contraction in human saphenous veins, Pharmacol Res, 2011.

M. N. Lehman, L. M. Coolen, and R. L. Goodman, Minireview: kisspeptin/neurokinin B/dynorphin (KNDy) cells of the arcuate nucleus: a central node in the control of gonadotropin-releasing hormone secretion, Endocrinology, vol.151, issue.8, pp.3479-89, 2010.

N. E. Rance, S. J. Krajewski, M. A. Smith, M. Cholanian, and P. A. Dacks, Neurokinin B and the Hypothalamic Regulation of Reproduction, Brain Res, vol.1364, pp.116-144, 2010.

E. Hrabovszky, B. Á. Borsay, K. Rácz, L. Herczeg, P. Ciofi et al., Substance P immunoreactivity exhibits frequent colocalization with kisspeptin and neurokinin B in the human infundibular region, PloS One, vol.8, issue.8, p.72369, 2013.

N. M. Page, N. J. Bell, S. M. Gardiner, I. T. Manyonda, K. J. Brayley et al., Characterization of the endokinins: human tachykinins with cardiovascular activity, Proc Natl Acad Sci, vol.100, issue.10, pp.6245-50, 2003.

L. Valentin-hansen, T. M. Frimurer, J. Mokrosinski, N. D. Holliday, and T. W. Schwartz, Biased Gs versus Gq proteins and ?-arrestin signaling in the NK1 receptor determined by interactions in the water hydrogen bond network, J Biol Chem, vol.290, issue.40, pp.24495-508, 2015.

L. Quartara and C. A. Maggi, The tachykinin NK1 receptor. Part I: ligands and mechanisms of cellular activation, Neuropeptides, vol.31, issue.6, pp.537-63, 1997.

L. Quartara and C. A. Maggi, The tachykinin NK1 receptor. Part II: Distribution and pathophysiological roles, Neuropeptides, vol.32, issue.1, pp.1-49, 1998.

K. Mcconalogue, C. U. Corvera, P. D. Gamp, E. F. Grady, and N. W. Bunnett, Desensitization of the neurokinin-1 receptor (NK1-R) in neurons: effects of substance P on the distribution of NK1-R, Galphaq/11, Gprotein receptor kinase-2/3, and beta-arrestin-1/2, Mol Biol Cell, vol.9, issue.8, pp.2305-2329, 1998.

M. S. Steinhoff, V. Mentzer, B. Geppetti, P. Pothoulakis, C. Bunnett et al., Tachykinins and Their Receptors: Contributions to Physiological Control and the Mechanisms of Disease, Physiol Rev, 2014.

P. J. Skerrett, Substance P causes pain--but also heals, Science, vol.249, issue.4969, p.625, 1990.

S. D. Brain and H. M. Cox, Neuropeptides and their receptors: innovative science providing novel therapeutic targets, Br J Pharmacol, vol.147, issue.1, pp.202-213, 2006.

R. Coveñas and M. Muñoz, Cancer progression and substance P, Histol Histopathol, vol.29, issue.7, pp.881-90, 2014.

I. M. Hennig, J. A. Laissue, U. Horisberger, and J. C. Reubi, Substance-P receptors in human primary neoplasms: tumoral and vascular localization, Int J Cancer, vol.61, issue.6, pp.786-92, 1995.

F. Esteban, M. Muñoz, M. A. González-moles, and M. Rosso, A role for substance P in cancer promotion and progression: a mechanism to counteract intracellular death signals following oncogene activation or DNA damage, Cancer Metastasis Rev, vol.25, issue.1, pp.137-182, 2006.

A. I. Vinik, J. Gonin, B. G. England, T. Jackson, M. K. Mcleod et al., Plasma substance-P in neuroendocrine tumors and idiopathic flushing: the value of pentagastrin stimulation tests and the effects of somatostatin analog, J Clin Endocrinol Metab, vol.70, issue.6, pp.1702-1711, 1990.

I. M. Modlin, K. Oberg, A. Taylor, I. Drozdov, L. Bodei et al., Neuroendocrine tumor biomarkers: current status and perspectives, Neuroendocrinology, vol.100, issue.4, pp.265-77, 2014.

J. M. Feldman and T. M. O'dorisio, Role of neuropeptides and serotonin in the diagnosis of carcinoid tumors, Am J Med, vol.81, issue.6B, pp.41-49, 1986.

Y. Wang and D. H. Wang, Role of substance P in renal injury during DOCA-salt hypertension, Endocrinology, vol.153, issue.12, pp.5972-5981, 2012.

H. M. Dehlin, E. J. Manteufel, A. L. Monroe, M. H. Reimer, and S. P. Levick, Substance P acting via the neurokinin-1 receptor regulates adverse myocardial remodeling in a rat model of hypertension, Int J Cardiol, 2013.

, Oct, vol.12, issue.5, pp.4643-51

A. Brattström and T. Seidenbecher, Central substance P increased blood pressure, heart rate and splanchnic nerve activity in anaesthetized rats without impairment of the baroreflex regulation, Neuropeptides, vol.23, issue.2, pp.81-87, 1992.

A. J. Moyes, S. C. Stanford, P. S. Hosford, A. J. Hobbs, and A. G. Ramage, Raised arterial blood pressure in neurokinin-1 receptor-deficient mice (NK1R(-/-) ): evidence for a neural rather than a vascular mechanism, Exp Physiol, vol.101, issue.5, pp.588-98, 2016.

H. D. Faulhaber, R. Rathsack, G. Rostock, V. Homuth, D. Pfeiffer et al., Evidence of decreased plasma substance P levels in human essential hypertension and influence of prazosin treatment

, Biomed Biochim Acta, vol.42, issue.7-8, pp.1019-1044, 1983.

O. Kohlmann, M. L. Cesaretti, M. Ginoza, A. Tavares, M. T. Zanella et al., Role of substance P in blood pressure regulation in salt-dependent experimental hypertension. Hypertens Dallas Tex 1979, vol.29, pp.506-515, 1997.

H. M. Dehlin and S. P. Levick, Substance P in heart failure: the good and the bad, Int J Cardiol, vol.170, issue.3, pp.270-277, 2014.

L. Quartara and M. Altamura, Tachykinin receptors antagonists: from research to clinic, Curr Drug Targets, vol.7, issue.8, pp.975-92, 2006.

R. Ramalho, R. Soares, N. Couto, and A. Moreira, Tachykinin receptors antagonism for asthma: a systematic review, BMC Pulm Med, vol.11, p.41, 2011.

J. K. Prague, R. E. Roberts, A. N. Comninos, S. Clarke, C. N. Jayasena et al., Neurokinin 3 receptor antagonism as a novel treatment for menopausal hot flushes: a phase 2, randomised, double-blind, placebo-controlled trial, Lancet Lond Engl, 2017.

S. Huang and V. L. Korlipara, Neurokinin-1 receptor antagonists: a comprehensive patent survey, Expert Opin Ther Pat, vol.20, issue.8, pp.1019-1064, 2010.

L. Patel and C. Lindley, Aprepitant -a novel NK1-receptor antagonist, Expert Opin Pharmacother, vol.4, issue.12, pp.2279-96, 2003.

R. Hargreaves, J. Ferreira, D. Hughes, J. Brands, J. Hale et al., Development of aprepitant, the first neurokinin-1 receptor antagonist for the prevention of chemotherapy-induced nausea and vomiting, Ann N Y Acad Sci, vol.1222, issue.1, pp.40-48, 2011.

R. Hargreaves, J. Ferreira, D. Hughes, J. Brands, J. Hale et al., Development of aprepitant, the first neurokinin-1 receptor antagonist for the prevention of chemotherapy-induced nausea and vomiting, Ann N Y Acad Sci, vol.1222, issue.1, pp.40-48, 2011.

A. D. Miller and R. A. Leslie, The area postrema and vomiting. Front Neuroendocrinol, vol.15, pp.301-321, 1994.

, The oral NK1 antagonist, aprepitant, given with standard antiemetics provides protection against nausea and vomiting over multiple cycles of cisplatin-based chemotherapy: a combined analysis of two randomised, placebo-controlled phase III clinical trials, 2017.

A. K. Majumdar, L. Howard, M. R. Goldberg, L. Hickey, M. Constanzer et al., Pharmacokinetics of Aprepitant After Single and Multiple Oral Doses in Healthy Volunteers, J Clin Pharmacol, vol.46, issue.3, pp.291-300, 2006.

S. Huskey, B. J. Dean, G. A. Doss, Z. Wang, C. Hop et al., The metabolic disposition of aprepitant, a substance P receptor antagonist, in rats and dogs, Drug Metab Dispos Biol Fate Chem, vol.32, issue.2, pp.246-58, 2004.

L. House, J. Ramirez, M. Seminerio, S. Mirkov, and M. J. Ratain, In vitro glucuronidation of aprepitant: a moderate inhibitor of UGT2B7, Xenobiotica Fate Foreign Compd Biol Syst, vol.45, issue.11, pp.990-998, 2015.

L. Patel and C. Lindley, Aprepitant--a novel NK1-receptor antagonist, Expert Opin Pharmacother, vol.4, issue.12, pp.2279-96, 2003.

P. Patel, J. S. Leeder, M. Piquette-miller, and L. L. Dupuis, Aprepitant and fosaprepitant drug interactions: a systematic review, Br J Clin Pharmacol, 2017.

W. A. Ball, D. B. Snavely, R. J. Hargreaves, A. Szegedi, C. Lines et al., Addition of an NK1 receptor antagonist to an SSRI did not enhance the antidepressant effects of SSRI monotherapy: results from a randomized clinical trial in patients with major depressive disorder, Hum Psychopharmacol, vol.29, issue.6, pp.568-77, 2014.

G. P. Vinson, J. A. Pudney, and B. J. Whitehouse, The mammalian adrenal circulation and the relationship between adrenal blood flow and steroidogenesis, J Endocrinol, vol.105, issue.2, pp.285-94, 1985.

S. J. Bacon and A. D. Smith, Preganglionic sympathetic neurones innervating the rat adrenal medulla: immunocytochemical evidence of synaptic input from nerve terminals containing substance P, GABA or 5-hydroxytryptamine, J Auton Nerv Syst, vol.24, issue.1-2, pp.97-122, 1988.

C. E. Gomez-sanchez, X. Qi, C. Velarde-miranda, M. W. Plonczynski, C. R. Parker et al., Development of monoclonal antibodies against human CYP11B1 and CYP11B2, Mol Cell Endocrinol, vol.383, issue.1-2, pp.111-118, 2014.

D. E. Campbell, N. Raftery, R. Tustin, N. B. Tustin, M. L. Desilvio et al., Measurement of plasmaderived substance P: biological, methodological, and statistical considerations, Clin Vaccine Immunol CVI, vol.13, issue.11, pp.1197-203, 2006.

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, Met-enkephalin, Leu-enkephalin and corticotrophin-releasing hormone on aldosterone secretion in the intact perfused rat adrenal, J Endocrinol, vol.140, issue.1, pp.91-97, 1994.

M. K. Kodjo, L. Desrues, L. Lavagno, A. Fasolo, J. M. Conlon et al., Ranakinin, a naturally occurring tachykinin, stimulates phospholipase C activity in the frog adrenal gland, Endocrinology, vol.139, issue.2, pp.505-517, 1998.

J. Lai, S. Lai, F. Tuluc, M. F. Tansky, L. E. Kilpatrick et al., Differences in the length of the carboxyl terminus mediate functional properties of neurokinin-1 receptor, Proc Natl Acad Sci, vol.105, issue.34, pp.12605-12615, 2008.

K. Vleugels, S. Schinner, D. Krause, H. Morawietz, S. R. Bornstein et al.,

, MAPKs and Wnt signaling pathways are independently involved in adipocytokine-mediated aldosterone secretion, Exp Clin Endocrinol Diabetes Off J Ger Soc Endocrinol Ger Diabetes Assoc, vol.119, issue.10, pp.644-652, 2011.

M. Doi, Y. Takahashi, R. Komatsu, F. Yamazaki, H. Yamada et al., Salt-sensitive hypertension in circadian clock-deficient Cry-null mice involves dysregulated adrenal Hsd3b6, Nat Med, 2010.

G. G. Nussdorfer, Paracrine control of adrenal cortical function by medullary chromaffin cells, Pharmacol Rev, vol.48, issue.4, pp.495-530, 1996.

M. V. Dzurik, A. Diedrich, B. Black, S. Y. Paranjape, S. R. Raj et al., Endogenous substance P modulates human cardiovascular regulation at rest and during orthostatic load, J Appl Physiol Bethesda Md, vol.102, issue.6, pp.2092-2099, 1985.

D. Jacob, I. Busciglio, D. Burton, H. Halawi, I. Oduyebo et al., Effects of NK1 receptors on gastric motor functions and satiation in healthy humans: results from a controlled trial with the NK1 antagonist aprepitant, Am J Physiol Gastrointest Liver Physiol, vol.313, issue.5, pp.505-515, 2017.

H. G. Gullner, W. B. Campbell, and W. A. Pettinger, Effects of substance P on renin release and renal function in anesthetized dogs, Life Sci, vol.24, issue.3, pp.237-282, 1979.

F. Pinet, J. Mizrahi, I. Laboulandine, J. Menard, and P. Corvol, Regulation of prorenin secretion in cultured human transfected juxtaglomerular cells, J Clin Invest, vol.80, issue.3, pp.724-755, 1987.

A. J. Shah, T. Kriska, K. M. Gauthier, J. R. Falck, and W. B. Campbell, Effect of Angiotensin II and ACTH on Adrenal Blood Flow in the Male Rat Adrenal Gland In Vivo, Endocrinology, vol.159, issue.1, pp.217-243, 201801.

F. Bayard, C. R. Cooke, D. J. Tiller, I. Z. Beitins, A. Kowarski et al., The regulation of aldosterone secretion in anephric man, J Clin Invest, vol.50, issue.8, pp.1585-95, 1971.

K. T. Weber, Aldosterone in congestive heart failure, N Engl J Med, vol.345, issue.23, pp.1689-97, 2001.

I. Castagliuolo, L. Valenick, J. Liu, and C. Pothoulakis, Epidermal growth factor receptor transactivation mediates substance P-induced mitogenic responses in U-373 MG cells, J Biol Chem, vol.275, issue.34, pp.26545-50, 2000.

H. Koon, D. Zhao, X. Na, M. P. Moyer, and C. Pothoulakis, Metalloproteinases and transforming growth factor-alpha mediate substance P-induced mitogen-activated protein kinase activation and proliferation in human colonocytes, J Biol Chem, vol.279, issue.44, pp.45519-45546, 2004.

I. Castagliuolo, O. Morteau, A. C. Keates, L. Valenick, C. Wang et al., Protective effects of neurokinin-1 receptor during colitis in mice: role of the epidermal growth factor receptor, Br J Pharmacol, vol.136, issue.2, pp.271-280, 2002.

M. J. Dubon and K. Park, The mechanisms of substance P-mediated migration of bone marrow-derived mesenchymal stem cell-like ST2 cells, Int J Mol Med, vol.37, issue.4, pp.1105-1116, 2016.

S. Amadesi, C. Reni, R. Katare, M. Meloni, A. Oikawa et al., Role for substance p-based nociceptive signaling in progenitor cell activation and angiogenesis during ischemia in mice and in human subjects, Circulation, vol.125, issue.14, pp.1-19, 2012.

A. M. Lerario, I. Finco, C. Lapensee, and G. D. Hammer, Molecular Mechanisms of Stem/Progenitor Cell Maintenance in the Adrenal Cortex, Front Endocrinol, vol.8, p.52, 2017.

S. Suvas, Role of Substance P Neuropeptide in Inflammation, Wound Healing, and Tissue Homeostasis, J Immunol Baltim Md, vol.199, issue.5, pp.1543-52, 1950.

P. Skrabanek, D. Cannon, J. Kirrane, D. Legge, and D. Powell, Circulating immunoreactive substance P in man, Ir J Med Sci, vol.145, issue.12, pp.399-408, 1976.

A. Charloux, C. Gronfier, E. Lonsdorfer-wolf, F. Piquard, and G. Brandenberger, Aldosterone release during the sleep-wake cycle in humans, Am J Physiol, vol.276, issue.1, pp.43-49, 1999.

T. M. Lovell, R. J. Woods, D. J. Butlin, K. J. Brayley, I. T. Manyonda et al., Identification of a novel mammalian post-translational modification, phosphocholine, on placental secretory polypeptides, J Mol Endocrinol, vol.39, issue.3, pp.189-98, 2007.

D. Marzioni, G. Fiore, A. Giordano, M. Nabissi, P. Florio et al., Placental expression of substance P and vasoactive intestinal peptide: evidence for a local effect on hormone release, J Clin Endocrinol Metab, vol.90, issue.4, pp.2378-83, 2005.

P. Lowry, Morning sickness is just the side effect of a new tachykinin that the placenta secretes to improve local blood flow, J Mol Endocrinol, vol.60, issue.1, pp.1-2, 2018.

A. Elsheikh, G. Creatsas, G. Mastorakos, S. Milingos, D. Loutradis et al., The renin-aldosterone system during normal and hypertensive pregnancy, Arch Gynecol Obstet, vol.264, issue.4, pp.182-187, 2001.

C. Delarue, V. Contesse, H. Lefebvre, S. Lenglet, L. Grumolato et al., Pharmacological profile of serotonergic receptors in the adrenal gland, Endocr Res, vol.24, issue.3-4, pp.687-94, 1998.

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.5001-5008, 2001.

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, issue.6, pp.1662-1668, 1993.

H. Lefebvre, K. N. Gonzalez, V. Contesse, C. Delarue, H. Vaudry et al., Effect of prolonged administration of the serotonin4 (5-HT4) receptor agonist cisapride on aldosterone secretion in healthy volunteers, Endocr Res, vol.24, issue.3-4, pp.749-52, 1998.

V. Dimitriadou, A. Rouleau, D. Trung-tuong, M. Newlands, G. J. Miller et al., Functional relationship between mast cells and C-sensitive nerve fibres evidenced by histamine H3-receptor modulation in rat lung and spleen, Clin Sci Lond Engl, vol.87, issue.2, pp.151-63, 1979.

F. Giancola, A. M. Rambaldi, F. Bianco, S. Iusco, N. Romagnoli et al., Localization of the 5-hydroxytryptamine 4 receptor in equine enteric neurons and extrinsic sensory fibers

, Neurogastroenterol Motil Off J Eur Gastrointest Motil Soc, vol.29, issue.7, 2017.

H. Kawabe, K. Kondo, M. Hayashi, M. Fujimaki, S. Nagahama et al., Mechanism of the central effects of dopamine and metoclopramide on aldosterone regulation in the rat, Endocrinol Jpn, 1983.

M. B. Gordon, T. J. Moore, R. G. Dluhy, and G. H. Williams, Dopaminergic blockade of the renin-angiotensinaldosterone system: effect of high and low sodium intakes, Clin Endocrinol, 1983.

M. Toyoda, T. Makino, M. Kagoura, and M. Morohashi, Immunolocalization of substance P in human skin mast cells, Arch Dermatol Res, vol.292, issue.8, pp.418-439, 2000.

H. Lefebvre, V. Contesse, C. Delarue, H. Vaudry, and J. M. Kuhn, Serotonergic regulation of adrenocortical function, Horm Metab Res Horm Stoffwechselforschung Horm Metab, vol.30, issue.6-7, pp.398-403, 1998.

F. Tore and N. Tuncel, Mast cells: target and source of neuropeptides, Curr Pharm Des, vol.15, issue.29, pp.3433-3478, 2009.

J. C. Melby, Angiotensin-converting enzyme in cardiovascular and adrenal tissues and implications for successful blood pressure management, Am J Cardiol, vol.69, issue.10, pp.2-7, 1992.

C. Seifarth, S. Trenkel, H. Schobel, E. G. Hahn, and J. Hensen, Influence of antihypertensive medication on aldosterone and renin concentration in the differential diagnosis of essential hypertension and primary aldosteronism, Clin Endocrinol (Oxf), vol.57, issue.4, pp.457-65, 2002.

P. Mulatero, A. Verhovez, F. Morello, and F. Veglio, Diagnosis and treatment of low-renin hypertension, Clin Endocrinol (Oxf), vol.67, issue.3, pp.324-358, 2007.

E. V. Adlin, L. E. Braitman, and R. S. Vasan, Bimodal aldosterone distribution in low-renin hypertension, Am J Hypertens, vol.26, issue.9, pp.1076-85, 2013.

J. W. Funder, Primary aldosteronism and low-renin hypertension: a continuum? Nephrol Dial Transplant Off Publ Eur Dial Transpl Assoc -Eur Ren Assoc, vol.28, pp.1625-1632, 2013.

R. Baudrand, F. J. Guarda, C. Fardella, G. Hundemer, J. Brown et al., Continuum of Renin-Independent Aldosteronism in Normotension. Hypertens Dallas Tex, vol.69, pp.950-956, 1979.

J. M. Brown, C. Robinson-cohen, M. A. Luque-fernandez, M. A. Allison, R. Baudrand et al., The Spectrum of Subclinical Primary Aldosteronism and Incident Hypertension: A Cohort Study, Ann Intern Med, vol.167, issue.9, pp.630-671, 2017.

T. L. Goodfriend and D. A. Calhoun, Resistant hypertension, obesity, sleep apnea, and aldosterone: theory and therapy. Hypertens Dallas Tex 1979, vol.43, pp.518-542, 2004.

J. Wei and Y. , Meta-analysis of effects of obstructive sleep apnea on the renin-angiotensinaldosterone system, J Geriatr Cardiol JGC, vol.13, issue.4, pp.333-376, 2016.

A. Glasow and S. R. Bornstein, Leptin and the adrenal gland, Eur J Clin Invest, vol.30, issue.3, pp.39-45, 2000.

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, 2005.

A. Huby, G. Antonova, J. Groenendyk, C. E. Gomez-sanchez, W. B. Bollag et al., Adipocyte-Derived Hormone Leptin Is a Direct Regulator of Aldosterone Secretion, Which Promotes Endothelial Dysfunction and Cardiac Fibrosis, Circulation, vol.132, issue.22, pp.2134-2179, 2015.

J. L. Faulkner, T. Bruder-nascimento, B. De-chantemèle, and E. J. , The regulation of aldosterone secretion by leptin: implications in obesity-related cardiovascular disease, Curr Opin Nephrol Hypertens, vol.27, issue.2, pp.63-72, 2018.

G. Colussi, C. Catena, R. Lapenna, E. Nadalini, A. Chiuch et al., Insulin resistance and hyperinsulinemia are related to plasma aldosterone levels in hypertensive patients. Diabetes Care, vol.30, pp.2349-54, 2007.

I. Biaggioni, F. Garcia, T. Inagami, and V. Haile, Hyporeninemic normoaldosteronism in severe autonomic failure, J Clin Endocrinol Metab, vol.76, issue.3, pp.580-586, 1993.

K. Swedberg, P. Eneroth, J. Kjekshus, and L. Wilhelmsen, Hormones regulating cardiovascular function in patients with severe congestive heart failure and their relation to mortality, CONSENSUS Trial Study Group. Circulation, vol.82, issue.5, pp.1730-1736, 1990.

S. Ubaid-girioli, S. E. Ferreira-melo, L. A. Souza, E. A. Nogueira, J. C. Yugar-toledo et al., Aldosterone escape with diuretic or angiotensin-converting enzyme inhibitor/angiotensin II receptor blocker combination therapy in patients with mild to moderate hypertension, J Clin Hypertens Greenwich Conn, vol.9, issue.10, pp.770-774, 2007.

N. Vodovar, M. Séronde, S. Laribi, E. Gayat, J. Lassus et al., Elevated Plasma B-Type Natriuretic Peptide Concentrations Directly Inhibit Circulating Neprilysin Activity in Heart Failure, JACC Heart Fail, vol.3, issue.8, pp.629-665, 2015.

T. Dudenbostel and D. A. Calhoun, Resistant hypertension, obstructive sleep apnoea and aldosterone, J Hum Hypertens, vol.26, issue.5, pp.281-288, 2012.

M. N. Pratt-ubunama, M. K. Nishizaka, R. L. Boedefeld, S. S. Cofield, S. M. Harding et al., Plasma aldosterone is related to severity of obstructive sleep apnea in subjects with resistant hypertension, Chest, vol.131, issue.2, pp.453-462, 2007.

V. K. Somers, D. P. White, R. Amin, W. T. Abraham, F. Costa et al., Sleep apnea and cardiovascular disease: an American Heart Association/american College Of Cardiology Foundation Scientific Statement from the American Heart Association Council for High Blood Pressure Research Professional Education Committee, Council on Clinical Cardiology, Stroke Council, and Council On Cardiovascular Nursing, collaboration with the National Heart, Lung, and Blood Institute National Center on Sleep Disorders Research, vol.118, pp.1080-111, 2008.

D. A. Calhoun, M. K. Nishizaka, M. A. Zaman, and S. M. Harding, Aldosterone excretion among subjects with resistant hypertension and symptoms of sleep apnea, Chest, vol.125, issue.1, pp.112-119, 2004.

K. Gaddam, E. Pimenta, S. J. Thomas, S. S. Cofield, S. Oparil et al., Spironolactone reduces severity of obstructive sleep apnoea in patients with resistant hypertension: a preliminary report, J Hum Hypertens, vol.24, issue.8, pp.532-539, 2010.

A. Ursavas, Upregulating substance P levels to treat obstructive sleep apnea, Expert Opin Ther Targets, vol.12, issue.5, pp.583-591, 2008.