R. A. Abbott, M. Cox, H. Markus, and A. Tomkins, Diet, body size and micronutrient status in Parkinson's disease, Eur. J. Clin. Nutr, vol.46, pp.879-884, 1992.

R. D. Abbott, H. Petrovitch, L. R. White, K. H. Masaki, C. M. Tanner et al., Frequency of bowel movements and the future risk of Parkinson's disease, Neurology, vol.57, pp.456-462, 2001.

R. Abdullah, I. Basak, K. S. Patil, G. Alves, J. P. Larsen et al., Parkinson's disease and age: The obvious but largely unexplored link, Exp. Gerontol, vol.68, pp.33-38, 2015.

A. Abizaid, Z. Liu, Z. B. Andrews, M. Shanabrough, E. Borok et al., Ghrelin modulates the activity and synaptic input organization of midbrain dopamine neurons while promoting appetite, J. Clin. Invest, vol.116, pp.3229-3239, 2006.

L. Abramson and M. Seligman, Modeling psychopathology in the laboratory: history and rationale, pp.1-26, 1977.

C. H. Adler, T. G. Beach, J. G. Hentz, H. A. Shill, J. N. Caviness et al., Low clinical diagnostic accuracy of early vs advanced Parkinson disease: clinicopathologic study, Neurology, vol.83, pp.406-412, 2014.

D. Agamonis, Chapter 9 Degenerative diseases

, Neuropathol. Illus. Interact. Course Med. Stud. Resid. URL, vol.16, issue.9

S. L. Alberico, M. D. Cassell, and N. S. Narayanan, The Vulnerable Ventral Tegmental Area in Parkinson's Disease, Basal Ganglia, vol.5, pp.51-55, 2015.

T. Alberio and M. Fasano, Proteomics in Parkinson's disease: An unbiased approach towards peripheral biomarkers and new therapies, J. Biotechnol, vol.156, pp.325-337, 2011.

R. L. Albin, A. B. Young, and J. B. Penney, The functional anatomy of basal ganglia disorders, Trends Neurosci, vol.12, pp.366-375, 1989.

I. Aldecoa, J. Navarro-otano, N. Stefanova, F. S. Sprenger, K. Seppi et al., Alpha-synuclein immunoreactivity patterns in the enteric nervous system, Neurosci. Lett, vol.602, pp.145-149, 2015.

O. Al-massadi, A. B. Crujeiras, R. C. Gonzalez, M. Pardo, C. Dieguez et al., Age, sex, and lactating status regulate ghrelin secretion and GOAT mRNA levels from isolated rat stomach, AJP Endocrinol. Metab, vol.299, 2010.

D. Alvarez-fischer, S. Guerreiro, S. Hunot, F. Saurini, M. Marien et al., Modelling Parkinson-like neurodegeneration via osmotic minipump delivery of MPTP and probenecid, J. Neurochem, vol.107, pp.701-711, 2008.

H. Alzahrani and A. Venneri, Cognitive and neuroanatomical correlates of neuropsychiatric symptoms in Parkinson's disease: A systematic review, J. Neurol. Sci, vol.356, pp.32-44, 2015.

W. R. Amberson, H. Penfield, and . Jasper, Epilepsy and the Functional Anatomy of the Human Brain, Science, vol.896, pp.645-646, 1954.

A. D. Andersen, M. Binzer, E. Stenager, and J. B. Gramsbergen, Cerebrospinal fluid biomarkers for Parkinson's disease -a systematic review, Acta Neurol. Scand, 2016.

Z. B. Andrews, D. Erion, R. Beiler, Z. Liu, A. Abizaid et al., Ghrelin Promotes and Protects Nigrostriatal Dopamine Function via a UCP2-Dependent Mitochondrial Mechanism, J. Neurosci, vol.29, pp.14057-14065, 2009.

Z. B. Andrews, Z. Liu, N. Walllingford, D. M. Erion, E. Borok et al., UCP2 mediates ghrelin's action on NPY/AgRP neurons by lowering free radicals, Nature, vol.454, pp.846-851, 2008.

S. V. Angeloni, N. Glynn, G. Ambrosini, M. J. Garant, J. D. Higley et al., Characterization of the rhesus monkey ghrelin gene and factors influencing ghrelin gene expression and fasting plasma levels, Endocrinology, vol.145, pp.2197-2205, 2004.

P. Anglade, S. Vyas, E. C. Hirsch, and Y. Agid, Apoptosis in dopaminergic neurons of the human substantia nigra during normal aging, Histol. Histopathol, vol.12, pp.603-610, 1997.

E. Angot, J. A. Steiner, C. M. Lema-tomé, P. Ekström, B. Mattsson et al., Alpha-synuclein cell-to-cell transfer and seeding in grafted dopaminergic neurons in vivo, PloS One, vol.7, p.39465, 2012.

S. Appel-cresswell, C. Vilarino-guell, M. Encarnacion, H. Sherman, I. Yu et al., Alpha-synuclein p.H50Q, a novel pathogenic mutation for Parkinson's disease, Mov. Disord. Off. J. Mov. Disord. Soc, vol.28, pp.811-813, 2013.

A. Asakawa, A. Inui, M. Fujimiya, R. Sakamaki, N. Shinfuku et al., Stomach regulates energy balance via acylated ghrelin and desacyl ghrelin, Gut, vol.54, pp.18-24, 2005.

A. Asakawa, A. Inui, T. Kaga, H. Yuzuriha, T. Nagata et al., A role of ghrelin in neuroendocrine and behavioral responses to stress in mice, Neuroendocrinology, vol.74, pp.143-147, 2001.

A. Ascherio, H. Chen, M. A. Schwarzschild, S. M. Zhang, G. A. Colditz et al., Caffeine, postmenopausal estrogen, and risk of Parkinson's disease, Neurology, vol.60, pp.790-795, 2003.

F. Assogna, L. Cravello, M. D. Orfei, N. Cellupica, C. Caltagirone et al., Alexithymia in Parkinson's disease: A systematic review of the literature, Parkinsonism Relat. Disord, vol.28, pp.1-11, 2016.

S. Auli?, T. T. Le, F. Moda, S. Abounit, S. Corvaglia et al., Defined ?-synuclein prion-like molecular assemblies spreading in cell culture, BMC Neurosci, vol.15, p.69, 2014.

P. K. Auluck, G. Caraveo, and S. Lindquist, ?-Synuclein: membrane interactions and toxicity in Parkinson's disease, Annu. Rev. Cell Dev. Biol, vol.26, pp.211-233, 2010.

M. Avoli, J. L. Noebels, M. Avoli, M. A. Rogawski, R. W. Olsen et al., Jasper and the Basic Mechanisms of the Epilepsies, National Center for Biotechnology Information (US), 2012.

H. Bagheri, C. Damase-michel, M. Lapeyre-mestre, S. Cismondo, D. O'connell et al., A study of salivary secretion in Parkinson's disease, Clin. Neuropharmacol, vol.22, pp.213-215, 1999.

L. Bahnassawy, S. Nicklas, T. Palm, I. Menzl, F. Birzele et al., The parkinson's disease-associated LRRK2 mutation R1441G inhibits neuronal differentiation of neural stem cells, Stem Cells Dev, vol.22, pp.2487-2496, 2013.

G. Baldanzi, Ghrelin and des-acyl ghrelin inhibit cell death in cardiomyocytes and endothelial cells through ERK1/2 and PI 3-kinase/AKT, J. Cell Biol, vol.159, pp.1029-1037, 2002.

G. Baldanzi, N. Filigheddu, S. Cutrupi, F. Catapano, S. Bonissoni et al., Ghrelin and des-acyl ghrelin inhibit cell death in cardiomyocytes and endothelial cells through ERK1/2 and PI 3-kinase/AKT, J. Cell Biol, vol.159, pp.1029-1037, 2002.

A. Bali and A. S. Jaggi, An Integrative Review on Role and Mechanisms of Ghrelin in Stress, Anxiety and Depression, Curr. Drug Targets, vol.17, pp.495-507, 2016.

R. Bandopadhyay, A. E. Kingsbury, M. R. Cookson, A. R. Reid, I. M. Evans et al., The expression of DJ-1 (PARK7) in normal human CNS and idiopathic Parkinson's disease, Brain J. Neurol, vol.127, pp.420-430, 2004.

U. Bandyopadhyay, U. Bandhyopadhyay, and A. M. Cuervo, Chaperone-mediated autophagy in aging and neurodegeneration: lessons from alpha-synuclein, Exp. Gerontol, vol.42, pp.120-128, 2007.

W. A. Banks, Extent and Direction of Ghrelin Transport Across the Blood-Brain Barrier Is Determined by Its Unique Primary Structure, J. Pharmacol. Exp. Ther, vol.302, pp.822-827, 2002.

R. Barazzoni, M. Zanetti, C. Ferreira, P. Vinci, A. Pirulli et al., Relationships between desacylated and acylated ghrelin and insulin sensitivity in the metabolic syndrome, J. Clin. Endocrinol. Metab, vol.92, pp.3935-3940, 2007.

A. Barbeau, The pathogenesis of Parkinson's disease: a new hypothesis, Can. Med. Assoc. J, vol.87, pp.802-807, 1962.

T. Barberi, P. Klivenyi, N. Y. Calingasan, H. Lee, H. Kawamata et al., Neural subtype specification of fertilization and nuclear transfer embryonic stem cells and application in parkinsonian mice, Nat. Biotechnol, vol.21, pp.1200-1207, 2003.

A. L. Bartels, A. T. Willemsen, R. Kortekaas, B. M. De-jong, R. De-vries et al., Decreased blood-brain barrier P-glycoprotein function in the progression of Parkinson's disease, PSP and MSA, J. Neural Transm, vol.115, pp.1001-1009, 1996.

P. Baskin and J. Salamone, Vacuous jaw movements in rats induced by acute reserpine administration: interactions with different doses of apomorphine, Pharmacol. Biochem. Behav, vol.46, pp.793-797, 1993.

J. A. Bayliss and Z. B. Andrews, Ghrelin is neuroprotective in Parkinson's disease: molecular mechanisms of metabolic neuroprotection, Ther. Adv. Endocrinol. Metab, vol.4, pp.25-36, 2013.

J. A. Bayliss, M. Lemus, V. V. Santos, M. Deo, J. Elsworth et al., Acylated but not des-acyl ghrelin is neuroprotective in an MPTP mouse model of Parkinson's Disease, J. Neurochem, 2016.

J. A. Bayliss, M. B. Lemus, R. Stark, V. V. Santos, A. Thompson et al., Ghrelin-AMPK Signaling Mediates the Neuroprotective Effects of Calorie Restriction in Parkinson's Disease, J. Neurosci. Off. J. Soc. Neurosci, vol.36, pp.3049-3063, 2016.

D. Bega and C. Zadikoff, Complementary & Alternative Management of Parkinson's Disease: An Evidence-Based Review of Eastern Influenced Practices, J. Mov. Disord, vol.7, pp.57-66, 2014.

C. G. Begley, An unappreciated challenge to oncology drug discovery: pitfalls in preclinical research, Am. Soc. Clin. Oncol. Educ. Book ASCO Am. Soc. Clin. Oncol. Meet, pp.466-468, 2013.

C. G. Begley, Six red flags for suspect work, Nature, vol.497, pp.433-434, 2013.

C. G. Begley and L. M. Ellis, Drug development: Raise standards for preclinical cancer research, Nature, vol.483, pp.531-533, 2012.

M. Bélanger and P. J. Magistretti, The role of astroglia in neuroprotection, Dialogues Clin. Neurosci, vol.11, pp.281-295, 2009.

C. Belzung and M. Lemoine, Criteria of validity for animal models of psychiatric disorders: focus on anxiety disorders and depression, Biol. Mood Anxiety Disord, vol.1, p.9, 2011.
URL : https://hal.archives-ouvertes.fr/halshs-00792412

A. L. Benabid, P. Pollak, C. Gervason, D. Hoffmann, D. M. Gao et al., Long-term suppression of tremor by chronic stimulation of the ventral intermediate thalamic nucleus, Lancet Lond. Engl, vol.337, pp.403-406, 1991.

A. L. Benabid, P. Pollak, C. Gross, D. Hoffmann, A. Benazzouz et al., Acute and long-term effects of subthalamic nucleus stimulation in Parkinson's disease, Stereotact. Funct. Neurosurg, vol.62, pp.76-84, 1994.

A. L. Benabid, P. Pollak, A. Louveau, S. Henry, and J. De-rougemont, Combined (thalamotomy and stimulation) stereotactic surgery of the VIM thalamic nucleus for bilateral Parkinson disease, Appl. Neurophysiol, vol.50, pp.344-346, 1987.

A. Bender, K. J. Krishnan, C. M. Morris, G. A. Taylor, A. K. Reeve et al., High levels of mitochondrial DNA deletions in substantia nigra neurons in aging and Parkinson disease, Nat. Genet, vol.38, pp.515-517, 2006.

A. Benso, D. H. St-pierre, F. Prodam, E. Gramaglia, R. Granata et al., Metabolic effects of overnight continuous infusion of unacylated ghrelin in humans, Eur. J. Endocrinol. Eur. Fed. Endocr. Soc, vol.166, pp.911-916, 2012.

H. Bernheimer, W. Birkmayer, O. Hornykiewicz, K. Jellinger, and F. Seitelberger, Brain dopamine and the syndromes of Parkinson and Huntington. Clinical, morphological and neurochemical correlations, J. Neurol. Sci, vol.20, pp.415-455, 1973.

A. Bertler and E. Rosengren, Occurrence and distribution of dopamine in brain and other tissues, Experientia, vol.15, pp.10-11, 1959.

R. Betarbet, T. B. Sherer, G. Mackenzie, M. Garcia-osuna, A. V. Panov et al., Chronic systemic pesticide exposure reproduces features of Parkinson's disease, Nat. Neurosci, vol.3, pp.1301-1306, 2000.

J. Bethlem, . Den-hartog, and W. A. Jager, The incidence and characteristics of Lewy bodies in idiopathic paralysis agitans (Parkinson's disease), J. Neurol. Neurosurg. Psychiatry, vol.23, pp.74-80, 1960.

R. Bhidayasiri and D. Tarsy, Parkinson's Disease: Hoehn and Yahr Scale, Movement Disorders: A Video Atlas, Current Clinical Neurology, pp.4-5, 2012.

R. Bhidayasiri and D. D. Truong, Therapeutic strategies for nonmotor symptoms in early Parkinson's disease: the case for a higher priority and stronger evidence, Parkinsonism Relat. Disord, vol.18, issue.1, pp.110-113, 2012.

J. L. Biedler, L. Helson, and B. A. Spengler, Morphology and growth, tumorigenicity, and cytogenetics of human neuroblastoma cells in continuous culture, Cancer Res, vol.33, pp.2643-2652, 1973.

H. Birkmayer, The L-3,4-dioxyphenylalanine (DOPA)-effect in Parkinson-akinesia, Wien Klin Wochenschr, vol.10, pp.787-795, 1961.

F. Blandini and M. Armentero, Dopamine receptor agonists for Parkinson's disease, Expert Opin. Investig. Drugs, vol.23, pp.387-410, 2014.

K. Blennow, L. Biscetti, P. Eusebi, and L. Parnetti, Cerebrospinal fluid biomarkers in Alzheimer's and Parkinson's diseases-From pathophysiology to clinical practice, Mov. Disord. Off. J. Mov. Disord. Soc, vol.31, pp.836-847, 2016.

J. Blesa, S. Phani, V. Jackson-lewis, and S. Przedborski, Classic and New Animal Models of Parkinson's Disease, J. Biomed. Biotechnol, vol.2012, pp.1-10, 2012.

J. Blesa, C. Pifl, M. A. Sánchez-gonzález, C. Juri, M. A. García-cabezas et al., The nigrostriatal system in the presymptomatic and symptomatic stages in the MPTP monkey model: a PET, histological and biochemical study, Neurobiol. Dis, vol.48, pp.79-91, 2012.

J. Blesa and S. Przedborski, Parkinson's disease: animal models and dopaminergic cell vulnerability, Front. Neuroanat, vol.8, p.155, 2014.

B. R. Bloem, N. M. De-vries, and G. Ebersbach, Nonpharmacological treatments for patients with Parkinson's disease, Mov. Disord, vol.30, pp.1504-1520, 2015.

A. M. Bonnet, M. F. Jutras, V. Czernecki, J. C. Corvol, and M. Vidailhet, Nonmotor symptoms in Parkinson's disease in 2012: relevant clinical aspects, 2012.

J. Booij, J. D. Speelman, M. W. Horstink, and E. C. Wolters, The clinical benefit of imaging striatal dopamine transporters with [123I]FP-CIT SPET in differentiating patients with presynaptic parkinsonism from those with other forms of parkinsonism, Eur. J. Nucl. Med, vol.28, pp.266-272, 2001.

H. Braak and E. Braak, Cognitive impairment in Parkinson's disease: amyloid plaques, neurofibrillary tangles, and neuropil threads in the cerebral cortex, J. Neural Transm. Park. Dis. Dement. Sect, vol.2, pp.45-57, 1990.

H. Braak, R. A. De-vos, E. N. Jansen, H. Bratzke, and E. Braak, Neuropathological hallmarks of Alzheimer's and Parkinson's diseases. Prog, Brain Res, vol.117, pp.267-285, 1998.

H. Braak, R. A. De-vos, J. Bohl, and K. Del-tredici, Gastric ?-synuclein immunoreactive inclusions in Meissner's and Auerbach's plexuses in cases staged for Parkinson's disease-related brain pathology, Neurosci. Lett, vol.396, pp.67-72, 2006.

H. Braak, E. Ghebremedhin, U. Rüb, H. Bratzke, and K. Tredici, Stages in the development of Parkinson's disease-related pathology, Cell Tissue Res, vol.318, pp.121-134, 2004.

H. Braak, K. D. Tredici, U. Rüb, R. A. De-vos, E. N. Jansen-steur et al., Staging of brain pathology related to sporadic Parkinson's disease, Neurobiol. Aging, vol.24, pp.197-211, 2003.

J. Bradbury, ?-synuclein gene triplication discovered in Parkinson's disease, Lancet Neurol, vol.2, p.715, 2003.

E. Braungart, M. Gerlach, P. Riederer, R. Baumeister, and M. C. Hoener, Caenorhabditis elegans MPP+ model of Parkinson's disease for high-throughput drug screenings, Neurodegener. Dis, vol.1, pp.175-183, 2004.

S. Breid, M. E. Bernis, J. T. Babila, M. C. Garca, H. Wille et al., Neuroinvasion of ?-Synuclein Prionoids after Intraperitoneal and Intraglossal Inoculation, J. Virol, 2016.

T. Breidert, J. Callebert, M. T. Heneka, G. Landreth, J. M. Launay et al., Protective action of the peroxisome proliferator-activated receptor-gamma agonist pioglitazone in a mouse model of Parkinson's disease, J. Neurochem, vol.82, pp.615-624, 2002.

V. Brochard, B. Combadière, A. Prigent, Y. Laouar, A. Perrin et al., Infiltration of CD4+ lymphocytes into the brain contributes to neurodegeneration in a mouse model of Parkinson disease, J. Clin. Invest, vol.119, pp.182-192, 2009.
URL : https://hal.archives-ouvertes.fr/inserm-00393947

F. Broglio, C. Gottero, F. Prodam, C. Gauna, G. Muccioli et al., Non-acylated ghrelin counteracts the metabolic but not the neuroendocrine response to acylated ghrelin in humans, J. Clin. Endocrinol. Metab, vol.89, pp.3062-3065, 2004.

F. Broglio, C. Gottero, F. Prodam, C. Gauna, G. Muccioli et al., Non-acylated ghrelin counteracts the metabolic but not the neuroendocrine response to acylated ghrelin in humans, J. Clin. Endocrinol. Metab, vol.89, pp.3062-3065, 2004.

A. I. Brooks, C. A. Chadwick, H. A. Gelbard, D. A. Cory-slechta, and H. J. Federoff, Paraquat elicited neurobehavioral syndrome caused by dopaminergic neuron loss, Brain Res, vol.823, pp.1-10, 1999.

E. Broussolle, F. Borson, J. M. Gonzalez-de-suso, J. A. Chayvialle, M. Beylot et al., Rev. Neurol, vol.147, pp.46-51, 1991.

D. Brück, G. K. Wenning, N. Stefanova, and L. Fellner, Glia and alpha-synuclein in neurodegeneration: A complex interaction, Neurobiol. Dis, vol.85, pp.262-274, 2016.

I. Bulgarelli, L. Tamiazzo, E. Bresciani, D. Rapetti, S. Caporali et al., Desacyl-ghrelin and synthetic GH-secretagogues modulate the production of inflammatory cytokines in mouse microglia cells stimulated by beta-amyloid fibrils, J. Neurosci. Res, vol.87, pp.2718-2727, 2009.

D. J. Burn, E. N. Rowan, L. M. Allan, S. Molloy, J. T. O'brien et al., Motor subtype and cognitive decline in Parkinson's disease, Parkinson's disease with dementia, and dementia with Lewy bodies, J. Neurol. Neurosurg. Psychiatry, vol.77, pp.585-589, 2006.

J. Burré, M. Sharma, T. Tsetsenis, V. Buchman, M. R. Etherton et al., Alpha-synuclein promotes SNARE-complex assembly in vivo and in vitro, Science, vol.329, pp.1663-1667, 2010.

Z. I. Cabantchik, A. Munnich, M. B. Youdim, and D. Devos, Regional siderosis: a new challenge for iron chelation therapy, Front. Pharmacol, vol.4, 2013.

A. Cabral, G. Fernandez, and M. Perello, Analysis of brain nuclei accessible to ghrelin present in the cerebrospinal fluid, Neuroscience, vol.253, pp.406-415, 2013.

B. Callaghan and J. B. Furness, Novel and Conventional Receptors for Ghrelin, Desacyl-Ghrelin, and Pharmacologically Related Compounds, Pharmacol. Rev, vol.66, pp.984-1001, 2014.

M. B. Callesen, J. Scheel-krüger, M. L. Kringelbach, and A. Møller, A systematic review of impulse control disorders in Parkinson's disease, J. Park. Dis, vol.3, pp.105-138, 2013.

J. P. Camiña, M. Lodeiro, O. Ischenko, A. C. Martini, and F. F. Casanueva, Stimulation by ghrelin of p42/p44 mitogen-activated protein kinase through the GHS-R1a receptor: role of G-proteins and beta-arrestins, J. Cell. Physiol, vol.213, pp.187-200, 2007.

V. P. Carlini, M. E. Monzón, M. M. Varas, A. B. Cragnolini, H. B. Schiöth et al., Ghrelin increases anxiety-like behavior and memory retention in rats, Biochem. Biophys. Res. Commun, vol.299, pp.739-743, 2002.

A. Carlsson, M. Lindqvist, and T. Magnusson, 3,4-Dihydroxyphenylalanine and 5-hydroxytryptophan as reserpine antagonists, Nature, vol.180, p.1200, 1957.

A. Carlsson, M. Lindqvist, T. Magnusson, and B. Waldeck, On the presence of 3-hydroxytyramine in brain, Science, vol.127, p.471, 1958.

M. Carta, T. Carlsson, A. Muñoz, D. Kirik, and A. Björklund, Serotonin-dopamine interaction in the induction and maintenance of L-DOPA-induced dyskinesias, Prog. Brain Res, vol.172, pp.465-478, 2008.

W. M. Caudle, R. E. Colebrooke, P. C. Emson, and G. W. Miller, Altered vesicular dopamine storage in Parkinson's disease: a premature demise, Trends Neurosci, vol.31, pp.303-308, 2008.

M. Cazorla, F. D. De-carvalho, M. O. Chohan, M. Shegda, N. Chuhma et al., Dopamine D2 receptors regulate the anatomical and functional balance of basal ganglia circuitry, Neuron, vol.81, pp.153-164, 2014.
URL : https://hal.archives-ouvertes.fr/inserm-02438087

J. S. Chandran, X. Lin, A. Zapata, A. Höke, M. Shimoji et al., Progressive behavioral deficits in DJ-1-deficient mice are associated with normal nigrostriatal function, Neurobiol. Dis, vol.29, pp.505-514, 2008.

M. Chartier-harlin, J. C. Dachsel, C. Vilariño-güell, S. J. Lincoln, F. Leprêtre et al., Translation initiator EIF4G1 mutations in familial Parkinson disease, Am. J. Hum. Genet, vol.89, pp.398-406, 2011.

M. Chartier-harlin, J. Kachergus, C. Roumier, V. Mouroux, X. Douay et al., ?-synuclein locus duplication as a cause of familial Parkinson's disease, The Lancet, vol.364, pp.1167-1169, 2004.

K. Chaudhuri and A. H. Schapira, Non-motor symptoms of Parkinson's disease: dopaminergic pathophysiology and treatment, Lancet Neurol, vol.8, pp.464-474, 2009.

K. R. Chaudhuri, D. G. Healy, and A. H. Schapira, Nonmotor symptoms of Parkinson's disease: diagnosis and management, Lancet Neurol, vol.5, pp.235-245, 2006.

C. Chen, Y. Weng, K. Chien, K. Lin, T. Yeh et al., G2019S) LRRK2 activates MKK4-JNK pathway and causes degeneration of SN dopaminergic neurons in a transgenic mouse model of PD, Cell Death Differ, vol.19, pp.1623-1633, 2012.

H. Chen, S. M. Zhang, M. A. Hernán, M. A. Schwarzschild, W. C. Willett et al., Nonsteroidal anti-inflammatory drugs and the risk of Parkinson disease, Arch. Neurol, vol.60, pp.1059-1064, 2003.

M. Chesselet, In vivo alpha-synuclein overexpression in rodents: a useful model of Parkinson's disease?, Exp. Neurol, vol.209, pp.22-27, 2008.

M. Chesselet, F. Richter, C. Zhu, I. Magen, M. B. Watson et al., A Progressive Mouse Model of Parkinson's Disease: The Thy1-aSyn, Neurotherapeutics, vol.9, pp.297-314, 2012.

Z. H. Cheung and N. Y. Ip, The emerging role of autophagy in Parkinson's disease, Mol. Brain, vol.2, p.29, 2009.

K. Chiba, L. A. Peterson, K. P. Castagnoli, A. J. Trevor, and N. Castagnoli, Studies on the molecular mechanism of bioactivation of the selective nigrostriatal toxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, Drug Metab. Dispos. Biol. Fate Chem, vol.13, pp.342-347, 1985.

K. Chiba, A. Trevor, and N. Castagnoli, Metabolism of the neurotoxic tertiary amine, MPTP, by brain monoamine oxidase, Biochem. Biophys. Res. Commun, vol.120, pp.574-578, 1984.

C. C. Chiueh, S. P. Markey, R. S. Burns, J. N. Johannessen, A. Pert et al., Neurochemical and behavioral effects of systemic and intranigral administration of N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine in the rat, Eur. J. Pharmacol, vol.100, pp.189-194, 1984.

K. B. Chow, J. Sun, K. Chu, W. Tai-cheung, C. H. Cheng et al., The truncated ghrelin receptor polypeptide (GHS-R1b) is localized in the endoplasmic reticulum where it forms heterodimers with ghrelin receptors (GHS-R1a) to attenuate their cell surface expression, Mol. Cell. Endocrinol, vol.348, pp.247-254, 2012.

L. J. Cloud and J. G. Greene, Gastrointestinal Features of Parkinson's Disease, Curr. Neurol. Neurosci. Rep, vol.11, pp.379-384, 2011.

F. C. Colpaert, Pharmacological characteristics of tremor, rigidity and hypokinesia induced by reserpine in rat, Neuropharmacology, vol.26, pp.1431-1440, 1987.

B. Connolly and S. H. Fox, Treatment of cognitive, psychiatric, and affective disorders associated with Parkinson's disease, Neurother. J. Am. Soc. Exp. Neurother, vol.11, pp.78-91, 2014.

J. Corcuff, E. Krim, F. Tison, A. Foubert-sanier, D. Guehl et al., Subthalamic nucleus stimulation in patients with Parkinson's disease does not increase serum ghrelin levels, Br. J. Nutr, vol.95, pp.1028-1029, 2006.

J. L. Costa, D. Naot, J. Lin, M. Watson, K. E. Callon et al., Ghrelin is an Osteoblast Mitogen and Increases Osteoclastic Bone Resorption In Vitro, Int. J. Pept, p.605193, 2011.

G. C. Cotzias, M. H. Van-woert, and L. M. Schiffer, Aromatic amino acids and modification of parkinsonism, N. Engl. J. Med, vol.276, pp.374-379, 1967.

M. A. Cowley, R. G. Smith, S. Diano, M. Tschöp, N. Pronchuk et al., The distribution and mechanism of action of ghrelin in the CNS demonstrates a novel hypothalamic circuit regulating energy homeostasis, Neuron, vol.37, pp.649-661, 2003.

A. M. Cuervo, L. Stefanis, R. Fredenburg, P. T. Lansbury, and D. Sulzer, Impaired degradation of mutant alpha-synuclein by chaperone-mediated autophagy, Science, vol.305, pp.1292-1295, 2004.

D. E. Cummings, J. Q. Purnell, R. S. Frayo, K. Schmidova, B. E. Wisse et al., A preprandial rise in plasma ghrelin levels suggests a role in meal initiation in humans, Diabetes, vol.50, pp.1714-1719, 2001.

P. Damier, E. C. Hirsch, P. Zhang, Y. Agid, and F. Javoy-agid, Glutathione peroxidase, glial cells and Parkinson's disease, Neuroscience, vol.52, pp.1-6, 1993.

Y. Date, M. Nakazato, N. Murakami, M. Kojima, K. Kangawa et al., Ghrelin Acts in the Central Nervous System to Stimulate Gastric Acid Secretion, Biochem. Biophys. Res. Commun, vol.280, pp.904-907, 2001.

W. Dauer and S. Przedborski, Parkinson's disease: mechanisms and models, Neuron, vol.39, pp.889-909, 2003.

K. D. Dave, S. De-silva, N. P. Sheth, S. Ramboz, M. J. Beck et al., Phenotypic characterization of recessive gene knockout rat models of Parkinson's disease, Neurobiol. Dis, vol.70, pp.190-203, 2014.

G. P. Davey, K. F. Tipton, and M. P. Murphy, Uptake and accumulation of 1-methyl-4-phenylpyridinium by rat liver mitochondria measured using an ion-selective electrode, Biochem. J, vol.288, pp.439-443, 1992.

G. C. Davis, A. C. Williams, S. P. Markey, M. H. Ebert, E. D. Caine et al., Chronic Parkinsonism secondary to intravenous injection of meperidine analogues, Psychiatry Res, vol.1, pp.249-254, 1979.

B. J. Day, M. Patel, L. Calavetta, L. Y. Chang, and J. S. Stamler, A mechanism of paraquat toxicity involving nitric oxide synthase, Proc. Natl. Acad. Sci. U. S. A, vol.96, pp.12760-12765, 1999.

M. De-rijk, C. Tzourio, M. M. Breteler, J. F. Dartigues, L. Amaducci et al., Prevalence of parkinsonism and Parkinson's disease in Europe: the EUROPARKINSON Collaborative Study. European Community Concerted Action on the Epidemiology of Parkinson's disease, J. Neurol. Neurosurg. Psychiatry, vol.62, pp.10-15, 1997.

B. Dehay, J. Bové, N. Rodríguez-muela, C. Perier, A. Recasens et al., Pathogenic Lysosomal Depletion in Parkinson's Disease, J. Neurosci, vol.30, pp.12535-12544, 2010.

P. J. Delhanty, S. J. Neggers, and A. J. Van-der-lely, Des-acyl ghrelin: a metabolically active peptide, Endocr. Dev, vol.25, pp.112-121, 2013.

P. J. Delhanty, Ghrelin and unacylated ghrelin stimulate human osteoblast growth via mitogen-activated protein kinase (MAPK)/phosphoinositide 3-kinase (PI3K) pathways in the absence of GHS-R1a, J. Endocrinol, vol.188, pp.37-47, 2006.

P. J. Delhanty, M. Huisman, M. Julien, K. Mouchain, P. Brune et al., The acylated (AG) to unacylated (UAG) ghrelin ratio in esterase inhibitortreated blood is higher than previously described, Clin. Endocrinol. (Oxf.), vol.82, pp.142-146, 2015.

P. J. Delhanty, S. J. Neggers, and A. J. Van-der-lely, Should we consider des-acyl ghrelin as a separate hormone and if so, what does it do?, Front. Horm. Res, vol.42, pp.163-174, 2014.

M. R. Delong, Primate models of movement disorders of basal ganglia origin, Trends Neurosci, vol.13, pp.281-285, 1990.

C. Delporte, Structure and Physiological Actions of Ghrelin. Scientifica, vol.2013, pp.1-25, 2013.

P. Derkinderen, T. Rouaud, T. Lebouvier, S. B. Des-varannes, M. Neunlist et al., Parkinson disease The enteric nervous system spills its guts, Neurology, vol.77, pp.1761-1767, 2011.

S. Desagher, J. Glowinski, and J. Premont, Astrocytes protect neurons from hydrogen peroxide toxicity, J. Neurosci, vol.16, pp.2553-2562, 1996.

P. Desplats, H. Lee, E. Bae, C. Patrick, E. Rockenstein et al., Inclusion formation and neuronal cell death through neuron-to-neuron transmission of alpha-synuclein, Proc. Natl. Acad. Sci. U. S. A, vol.106, pp.13010-13015, 2009.

U. Dettmer, A. J. Newman, V. E. Von-saucken, T. Bartels, and D. Selkoe, KTKEGV repeat motifs are key mediators of normal ?-synuclein tetramerization: Their mutation causes excess monomers and neurotoxicity, Proc. Natl. Acad. Sci. U. S. A, vol.112, pp.9596-9601, 2015.

D. Devos, C. Moreau, J. C. Devedjian, J. Kluza, M. Petrault et al., Targeting chelatable iron as a therapeutic modality in Parkinson's disease, Antioxid. Redox Signal, vol.21, pp.195-210, 2014.

S. Diano, S. Farr, S. .. Benoit, E. Mcnay, I. Da-silva et al., Ghrelin controls hippocampal spine synapse density and memory prformance, Nat. Neurosci, vol.9, pp.381-388, 2006.

V. Dias, E. Junn, and M. M. Mouradian, The Role of Oxidative Stress in Parkinson's Disease, J. Park. Dis, vol.3, pp.461-491, 2013.

D. W. Dickson, Parkinson's Disease and Parkinsonism: Neuropathology. Cold Spring Harb, Perspect. Med, vol.2, p.9258, 2012.

D. W. Dickson, H. Braak, J. E. Duda, C. Duyckaerts, T. Gasser et al., Neuropathological assessment of Parkinson's disease: refining the diagnostic criteria, Lancet Neurol, vol.8, pp.70238-70246, 2009.

N. N. Dissanayaka, A. Sellbach, S. Matheson, J. D. O'sullivan, P. A. Silburn et al., Anxiety disorders in Parkinson's disease: prevalence and risk factors, Mov. Disord. Off. J. Mov. Disord. Soc, vol.25, pp.838-845, 2010.

C. B. Do, J. Y. Tung, E. Dorfman, A. K. Kiefer, E. M. Drabant et al., Web-based genome-wide association study identifies two novel loci and a substantial genetic component for Parkinson's disease, PLoS Genet, vol.7, p.1002141, 2011.

J. Dong, N. Song, J. Xie, and H. Jiang, Ghrelin Antagonized 1-Methyl-4-Phenylpyridinium (MPP+)-Induced Apoptosis in MES23.5 Cells, J. Mol. Neurosci, vol.37, pp.182-189, 2009.

E. R. Dorsey, R. Constantinescu, J. P. Thompson, K. M. Biglan, R. G. Holloway et al., Projected number of people with Parkinson disease in the most populous nations, Neurology, vol.68, pp.384-386, 2005.

E. R. Dorsey, B. P. George, B. Leff, and A. W. Willis, The coming crisis, Neurology, vol.80, 1989.

R. L. Doty, Olfactory dysfunction in Parkinson disease, Nat. Rev. Neurol, vol.8, pp.329-339, 2012.

W. Duan and M. P. Mattson, Dietary restriction and 2-deoxyglucose administration improve behavioral outcome and reduce degeneration of dopaminergic neurons in models of Parkinson's disease, J. Neurosci. Res, vol.57, pp.195-206, 1999.

S. B. Dunnett and M. Lelos, Behavioral analysis of motor and non-motor symptoms in rodent models of Parkinson's disease. Prog, Brain Res, vol.184, issue.10, pp.84003-84011, 2010.

C. J. Dunning, J. F. Reyes, J. A. Steiner, and P. Brundin, Can Parkinson's disease pathology be propagated from one neuron to another?, Prog. Neurobiol, vol.97, pp.205-219, 2012.

J. Dusonchet, O. Kochubey, K. Stafa, S. M. Young, R. Zufferey et al., A rat model of progressive nigral neurodegeneration induced by the Parkinson's disease-associated G2019S mutation in LRRK2, J. Neurosci. Off. J. Soc. Neurosci, vol.31, pp.907-912, 2011.

S. Duty and P. Jenner, Animal models of Parkinson's disease: a source of novel treatments and clues to the cause of the disease: Animal models of Parkinson's disease, Br. J. Pharmacol, vol.164, pp.1357-1391, 2011.

R. C. Duvoisin, R. Eldridge, A. Williams, J. Nutt, and D. Calne, Twin study of Parkinson disease, Neurology, vol.31, pp.77-77, 1981.

M. J. Eadie and J. H. Tyrer, ALIMENTARY DISORDER IN PARKINSONISM, Australas. Ann. Med, vol.14, pp.13-22, 1965.

S. Edvardson, Y. Cinnamon, A. Ta-shma, A. Shaag, Y. Yim et al., A deleterious mutation in DNAJC6 encoding the neuronal-specific clathrin-uncoating co-chaperone auxilin, is associated with juvenile parkinsonism, PloS One, vol.7, 2012.

L. L. Edwards, R. F. Pfeiffer, E. M. Quigley, R. Hofman, and M. Balluff, Gastrointestinal symptoms in Parkinson's disease, Mov. Disord. Off. J. Mov. Disord. Soc, vol.6, pp.151-156, 1991.

T. L. Edwards, W. K. Scott, C. Almonte, A. Burt, E. H. Powell et al., Genome-wide association study confirms SNPs in SNCA and the MAPT region as common risk factors for Parkinson disease, Ann. Hum. Genet, vol.74, pp.97-109, 2010.

H. Ehringer and O. Hornykiewicz, Distribution of noradrenaline and dopamine (3-hydroxytyramine) in the human brain and their behavior in diseases of the extrapyramidal system, 1960.

, Klin. Wochenschr, vol.38, pp.1236-1239

T. Eichner and S. E. Radford, A diversity of assembly mechanisms of a generic amyloid fold, Mol. Cell, vol.43, pp.8-18, 2011.

E. Eidelberg, B. A. Brooks, W. W. Morgan, J. G. Walden, and R. H. Kokemoor, Variability and functional recovery in the N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine model of parkinsonism in monkeys, Neuroscience, vol.18, pp.817-822, 1986.

P. Ejlerskov, I. Rasmussen, T. T. Nielsen, A. Bergström, Y. Tohyama et al., Tubulin polymerization-promoting protein (TPPP/p25?) promotes unconventional secretion of ?-synuclein through exophagy by impairing autophagosome-lysosome fusion, J. Biol. Chem, vol.288, pp.17313-17335, 2013.

N. Ejskjaer, G. Dimcevski, J. Wo, P. M. Hellström, L. C. Gormsen et al., Safety and efficacy of ghrelin agonist TZP-101 in relieving symptoms in patients with diabetic gastroparesis: a randomized, placebo-controlled study, Neurogastroenterol. Motil. Off. J. Eur. Gastrointest. Motil. Soc, vol.22, pp.1069-281, 2010.

N. Ejskjaer, J. M. Wo, T. Esfandyari, M. Jamal, G. Dimcevski et al., A phase 2a, randomized, double-blind 28-day study of TZP-102 a ghrelin receptor agonist for diabetic gastroparesis, Neurogastroenterol. Motil. Off. J. Eur. Gastrointest. Motil. Soc, vol.25, pp.140-150, 2013.

A. Elbaz, L. Carcaillon, S. Kab, and F. Moisan, Epidemiology of Parkinson's disease, Rev. Neurol, vol.172, pp.14-26, 2016.

R. Eldridge and S. E. Ince, The low concordance rate for Parkinson's disease in twins A possible explanation, Neurology, vol.34, pp.1354-1354, 1984.

J. D. Erickson, L. E. Eiden, M. K. Schafer, and E. Weihe, Reserpine-and tetrabenazine-sensitive transport of (3)H-histamine by the neuronal isoform of the vesicular monoamine transporter, J. Mol. Neurosci. MN, vol.6, pp.277-287, 1995.

A. H. Evans, A. D. Lawrence, J. Potts, L. Macgregor, R. Katzenschlager et al., Relationship between impulsive sensation seeking traits, smoking, alcohol and caffeine intake, and Parkinson's disease, J. Neurol. Neurosurg. Psychiatry, vol.77, pp.317-321, 2006.

G. Fabbrini, J. M. Brotchie, F. Grandas, M. Nomoto, and C. G. Goetz, Levodopa-induced dyskinesias, Mov. Disord. Off. J. Mov. Disord. Soc, vol.22, pp.1379-1389, 2007.

S. Fahn, The medical treatment of Parkinson disease from James Parkinson to George Cotzias, Mov. Disord. Off. J. Mov. Disord. Soc, vol.30, pp.4-18, 2015.

B. H. Falkenburger, T. Saridaki, and E. Dinter, Cellular models for Parkinson's disease, J. Neurochem, 2016.

B. H. Falkenburger, P. D. Schulz, P. D. Riederer, P. D. Reichmann, P. D. Youdim et al., Limitations of cellular models in Parkinson's disease research, Journal of Neural Transmission. Supplementa, pp.261-268, 2006.

M. Farrer, K. Gwinn-hardy, M. Muenter, F. W. Devrieze, R. Crook et al., A chromosome 4p haplotype segregating with Parkinson's disease and postural tremor, Hum. Mol. Genet, vol.8, pp.81-85, 1999.

M. B. Feany and W. W. Bender, A Drosophila model of Parkinson's disease, Nature, vol.404, pp.394-398, 2000.

F. Fenelon, Neurosurgery of the ansa lenticularis in dyskinesia and Parkinson's disease, 1955.

. Sem and . Hôp, Organe Fondé Par Assoc. Enseign. Méd. Hôp. Paris, vol.31, pp.1835-1837

F. Fenelon, Subpallidal extrapyramidotomy by electrocoagulation in the treatment of dyskinesia, Rev. Esp. Oto-Neuro-Oftalmol. Neurocir, vol.14, pp.99-104, 1955.

R. Ferese, N. Modugno, R. Campopiano, M. Santilli, S. Zampatti et al., Four Copies of SNCA Responsible for Autosomal Dominant Parkinson's Disease in Two Italian Siblings, p.546462, 2015.

V. S. Fernandes, J. R. Santos, A. H. Leão, A. M. Medeiros, T. G. Melo et al., Repeated treatment with a low dose of reserpine as a progressive model of Parkinson's disease, Behav. Brain Res, vol.231, pp.154-163, 2012.

R. J. Ferrante, J. B. Schulz, N. W. Kowall, and M. F. Beal, Systemic administration of rotenone produces selective damage in the striatum and globus pallidus, but not in the substantia nigra, Brain Res, vol.753, pp.157-162, 1997.

U. Fiszer, M. Micha?owska, B. Baranowska, E. Woli?ska-witort, W. Jeske et al., Leptin and ghrelin concentrations and weight loss in Parkinson's disease, Acta Neurol. Scand, vol.121, pp.230-236, 2010.

S. M. Fleming, Early and Progressive Sensorimotor Anomalies in Mice Overexpressing Wild-Type Human -Synuclein, J. Neurosci, vol.24, pp.9434-9440, 2004.

S. M. Fleming, J. Salcedo, P. Fernagut, E. Rockenstein, E. Masliah et al., Early and progressive sensorimotor anomalies in mice overexpressing wild-type human alpha-synuclein, J. Neurosci. Off. J. Soc. Neurosci, vol.24, pp.9434-9440, 2004.

M. S. Forman, J. Q. Trojanowski, V. M. Lee, and .. , Neurodegenerative diseases: a decade of discoveries paves the way for therapeutic breakthroughs, Nat. Med, vol.10, pp.1055-1063, 2004.

J. S. Fowler, N. D. Volkow, G. J. Wang, J. Logan, N. Pappas et al., Agerelated increases in brain monoamine oxidase B in living healthy human subjects, Neurobiol. Aging, vol.18, pp.431-435, 1997.

L. Frago, Neuroprotective actions of ghrelin and growth hormone secretagogues, Front. Mol. Neurosci, vol.4, 2011.

M. François, S. Barde, N. Achamrah, J. Breton, J. Do-rego et al., The number of preproghrelin mRNA expressing cells is increased in mice with activity-based anorexia, Neuropeptides, vol.51, pp.17-23, 2015.

N. E. Fritz, D. A. Kegelmeyer, A. D. Kloos, S. Linder, A. Park et al., Motor performance differentiates individuals with Lewy body dementia, Parkinson's and Alzheimer's disease, Gait Posture, vol.50, pp.1-7, 2016.

M. Funayama, K. Hasegawa, H. Kowa, M. Saito, S. Tsuji et al., A new locus for Parkinson's disease (PARK8) maps to chromosome 12p11.2-q13.1, Ann. Neurol, vol.51, pp.296-301, 2002.

M. Funayama, K. Ohe, T. Amo, N. Furuya, J. Yamaguchi et al., CHCHD2 mutations in autosomal dominant late-onset Parkinson's disease: a genomewide linkage and sequencing study, Lancet Neurol, vol.14, issue.14, pp.70266-70268, 2015.

J. B. Furness, B. Hunne, N. Matsuda, L. Yin, D. Russo et al., Investigation of the presence of ghrelin in the central nervous system of the rat and mouse, Neuroscience, vol.193, pp.1-9, 2011.

X. Gao, E. R. Martin, Y. Liu, G. Mayhew, J. M. Vance et al., Genome-wide linkage screen in familial Parkinson disease identifies loci on chromosomes 3 and 18, Am. J. Hum. Genet, vol.84, pp.499-504, 2009.

R. C. Gardner, J. F. Burke, J. Nettiksimmons, S. Goldman, C. M. Tanner et al., Traumatic brain injury in later life increases risk for Parkinson disease, Ann. Neurol, vol.77, pp.987-995, 2015.

T. Gasser, B. Müller-myhsok, Z. K. Wszolek, R. Oehlmann, D. B. Calne et al., A susceptibility locus for Parkinson's disease maps to chromosome 2p13, Nat. Genet, vol.18, pp.262-265, 1998.

C. Gauna, P. J. Delhanty, L. J. Hofland, J. A. Janssen, F. Broglio et al., Ghrelin stimulates, whereas des-octanoyl ghrelin inhibits, glucose output by primary hepatocytes, J. Clin. Endocrinol. Metab, vol.90, pp.1055-1060, 2005.

C. Gauna, B. Van-de-zande, A. Van-kerkwijk, A. P. Themmen, A. J. Van-der-lely et al., Unacylated ghrelin is not a functional antagonist but a full agonist of the type 1a growth hormone secretagogue receptor (GHS-R), Mol. Cell. Endocrinol, vol.274, pp.30-34, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00531929

F. Gaven, P. Marin, and S. Claeysen, Primary culture of mouse dopaminergic neurons, J. Vis. Exp. JoVE e51751, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01943338

J. F. Geddes, A. J. Hughes, A. J. Lees, and S. E. Daniel, Pathological overlap in cases of parkinsonism associated with neurofibrillary tangles. A study of recent cases of postencephalitic parkinsonism and comparison with progressive supranuclear palsy and Guamanian parkinsonism-dementia complex, Brain J. Neurol, vol.116, pp.281-302, 1993.

N. Germain, B. Galusca, D. Grouselle, D. Frere, S. Billard et al., Ghrelin and obestatin circadian levels differentiate bingeing-purging from restrictive anorexia nervosa, J. Clin. Endocrinol. Metab, vol.95, pp.3057-3062, 2010.

N. Germain, B. Galusca, D. Grouselle, D. Frere, V. Tolle et al., Ghrelin/obestatin ratio in two populations with low bodyweight: constitutional thinness and anorexia nervosa, Psychoneuroendocrinology, vol.34, pp.413-419, 2009.

E. Gershon and W. W. Vale, CRF type 2 receptors mediate the metabolic effects of ghrelin in C2C12 cells, Obes. Silver Spring Md, vol.22, pp.380-389, 2014.

E. Ghigo, F. Broglio, E. Arvat, M. Maccario, M. Papotti et al., Ghrelin: more than a natural GH secretagogue and/or an orexigenic factor, Clin. Endocrinol. (Oxf.), vol.62, pp.1-17, 2005.

P. L. Gildenberg, Evolution of basal ganglia surgery for movement disorders, Stereotact. Funct. Neurosurg, vol.84, pp.131-135, 2006.

Y. Glinka, M. Gassen, and M. B. Youdim, Mechanism of 6-hydroxydopamine neurotoxicity, J. Neural Transm. Suppl, vol.50, pp.55-66, 1997.

S. Gnanapavan, B. Kola, S. A. Bustin, D. G. Morris, P. Mcgee et al., The tissue distribution of the mRNA of the ghrelin and subtypes of its receptor, GHS-R, in humans, J. Clin. Endocrinol. Metab, vol.87, pp.2988-2991, 2002.

J. Godau, A. Hussl, P. Lolekha, A. J. Stoessl, and K. Seppi, Neuroimaging: Current role in detecting pre-motor Parkinson's disease, Mov. Disord, vol.27, pp.634-643, 2012.

M. Goedert, M. G. Spillantini, K. Del-tredici, and H. Braak, 100 years of Lewy pathology, Nat. Rev. Neurol, vol.9, pp.13-24, 2013.

C. G. Goetz, The History of Parkinson's Disease: Early Clinical Descriptions and Neurological Therapies. Cold Spring Harb, Perspect. Med, vol.1, pp.8862-008862, 2011.

C. G. Goetz, S. Fahn, P. Martinez-martin, W. Poewe, C. Sampaio et al., Movement Disorder Society-sponsored revision of the Unified Parkinson's Disease Rating Scale (MDS-UPDRS): Process, format, and clinimetric testing plan, Mov. Disord. Off. J. Mov. Disord. Soc, vol.22, pp.41-47, 2007.

M. S. Goldberg, Parkin-deficient Mice Exhibit Nigrostriatal Deficits but Not Loss of Dopaminergic Neurons, J. Biol. Chem, vol.278, pp.43628-43635, 2003.

J. E. Goldman, S. H. Yen, F. C. Chiu, and N. S. Peress, Lewy bodies of Parkinson's disease contain neurofilament antigens, Science, vol.221, pp.1082-1084, 1983.

J. M. Goldstein, A. Barnett, and J. B. Malick, The evaluation of anti-parkinson drugs on reserpineinduced rigidity in rats, Eur. J. Pharmacol, vol.33, pp.183-188, 1975.

R. A. González-polo, M. Niso-santano, M. A. Ortíz-ortíz, A. Gómez-martín, J. M. Morán et al., Relationship between autophagy and apoptotic cell death in human neuroblastoma cells treated with paraquat: could autophagy be a "brake, Autophagy, vol.3, pp.366-367, 2007.

R. A. González-polo, M. Niso-santano, M. A. Ortíz-ortíz, A. Gómez-martín, J. M. Morán et al., Inhibition of paraquat-induced autophagy accelerates the apoptotic cell death in neuroblastoma SH-SY5Y cells, Toxicol. Sci. Off. J. Soc. Toxicol, vol.97, pp.448-458, 2007.

R. Goodman and H. R. Herschman, Nerve growth factor-mediated induction of tyrosine hydroxylase in a clonal pheochromocytoma cell line, Proc. Natl. Acad. Sci. U. S. A, vol.75, pp.4587-4590, 1978.

A. M. Gotham, R. G. Brown, and C. D. Marsden, Depression in Parkinson's disease: a quantitative and qualitative analysis, J. Neurol. Neurosurg. Psychiatry, vol.49, pp.381-389, 1986.

R. Granata, F. Settanni, L. Biancone, L. Trovato, R. Nano et al., Acylated and unacylated ghrelin promote proliferation and inhibit apoptosis of pancreatic beta-cells and human islets: involvement of 3',5'-cyclic adenosine monophosphate/protein kinase A, extracellular signal-regulated kinase 1/2, and phosphatidyl inositol 3-Kinase/Akt signaling, Endocrinology, vol.148, pp.512-529, 2007.

C. M. Grau and L. A. Greene, Use of PC12 Cells and Rat Superior Cervical Ganglion Sympathetic Neurons as Models for Neuroprotective Assays Relevant to Parkinson's Disease, Methods Mol. Biol. Clifton NJ, vol.846, pp.201-211, 2012.

M. T. Gray and J. M. Woulfe, Striatal blood-brain barrier permeability in Parkinson's disease, J. Cereb. Blood Flow Metab. Off. J. Int. Soc. Cereb. Blood Flow Metab, vol.35, pp.747-750, 2015.

J. T. Greenamyre, T. B. Sherer, R. Betarbet, and A. V. Panov, Complex I and Parkinson's Disease, IUBMB Life, vol.52, pp.135-141, 2001.

J. G. Greenfield and F. D. Bosanquet, The brain-stem lesions in Parkinsonism, J. Neurol. Neurosurg. Psychiatry, vol.16, pp.213-226, 1953.

S. Greffard, M. Verny, A. Bonnet, D. Seilhean, J. Hauw et al., A stable proportion of Lewy body bearing neurons in the substantia nigra suggests a model in which the Lewy body causes neuronal death, Neurobiol. Aging, vol.31, pp.99-103, 2010.

R. C. Griggs, B. Fontaine, and G. Sobue, Global imperatives and challenges facing the practice of neurology, Neurol. Clin. Pract, vol.1, pp.49-54, 2011.

C. E. Gross, T. Boraud, D. Guehl, B. Bioulac, and E. Bezard, From experimentation to the surgical treatment of Parkinson's disease: prelude or suite in basal ganglia research?, Prog. Neurobiol, vol.59, pp.509-532, 1999.

X. Guan, H. Yu, O. C. Palyha, K. K. Mckee, S. D. Feighner et al., Distribution of mRNA encoding the growth hormone secretagogue receptor in brain and peripheral tissues, Mol. Brain Res, vol.48, pp.23-29, 1997.

G. Guiot and S. Brion, , 1953.

, Rev. Neurol, vol.89, pp.578-580

J. A. Gutierrez, P. J. Solenberg, D. R. Perkins, J. A. Willency, M. D. Knierman et al., Ghrelin octanoylation mediated by an orphan lipid transferase, Proc. Natl. Acad. Sci. 105, pp.6320-6325, 2008.

C. A. Haaxma, B. R. Bloem, G. F. Borm, W. J. Oyen, K. L. Leenders et al., Gender differences in Parkinson's disease, J. Neurol. Neurosurg. Psychiatry, vol.78, pp.819-824, 2007.

A. Haehner, S. Boesveldt, H. W. Berendse, A. Mackay-sim, J. Fleischmann et al., Prevalence of smell loss in Parkinson's disease--a multicenter study, Parkinsonism Relat. Disord, vol.15, pp.490-494, 2009.

S. Halbgebauer, P. Öckl, K. Wirth, P. Steinacker, and M. Otto, Protein biomarkers in Parkinson's disease: Focus on cerebrospinal fluid markers and synaptic proteins, Mov. Disord. Off. J. Mov. Disord. Soc, vol.31, pp.848-860, 2016.

M. Hallett, Parkinson's disease tremor: pathophysiology, Parkinsonism Relat. Disord, vol.18, issue.1, pp.85-86, 2012.

G. Halliday, M. T. Herrero, K. Murphy, H. Mccann, F. Ros-bernal et al., No Lewy pathology in monkeys with over 10 years of severe MPTP Parkinsonism, Mov. Disord. Off. J. Mov. Disord. Soc, vol.24, pp.1519-1523, 2009.

G. M. Halliday, Thalamic changes in Parkinson's disease, Proceedings of WFN XVIII World Congress on Parkinson's Disease and, vol.15, pp.152-155, 2009.

D. J. Hampshire, E. Roberts, Y. Crow, J. Bond, A. Mubaidin et al., Kufor-Rakeb syndrome, pallido-pyramidal degeneration with supranuclear upgaze paresis and dementia, maps to 1p36, J. Med. Genet, vol.38, pp.680-682, 2001.

T. Hasegawa, M. Matsuzaki, A. Takeda, A. Kikuchi, H. Akita et al., Accelerated alpha-synuclein aggregation after differentiation of SH-SY5Y neuroblastoma cells, Brain Res, vol.1013, pp.51-59, 2004.

Z. S. Heetun and E. M. Quigley, Gastroparesis and Parkinson's disease: a systematic review, Parkinsonism Relat. Disord, vol.18, pp.433-440, 2012.

M. A. Hely, J. G. Morris, W. G. Reid, and R. Trafficante, Sydney multicenter study of Parkinson's disease: Non-L-dopa-responsive problems dominate at 15 years, Mov. Disord, vol.20, pp.190-199, 2005.

M. A. Hernán, B. Takkouche, F. Caamaño-isorna, and J. J. Gestal-otero, A meta-analysis of coffee drinking, cigarette smoking, and the risk of Parkinson's disease, Ann. Neurol, vol.52, pp.276-284, 2002.

A. A. Hicks, H. Pétursson, T. Jónsson, H. Stefánsson, H. S. Jóhannsdóttir et al., A susceptibility gene for lateonset idiopathic Parkinson's disease, Ann. Neurol, vol.52, pp.549-555, 2002.

T. Hideshima, J. E. Bradner, D. Chauhan, and K. C. Anderson, Intracellular protein degradation and its therapeutic implications, Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res, vol.11, pp.8530-8533, 2005.

R. Hinde, Animal models in human psychobiology, The Uses of Similarities and Differences in Comparative Psychopathology, pp.187-202, 1976.

Y. Hirata and T. Nagatsu, Rotenone and CCCP inhibit tyrosine hydroxylation in rat striatal tissue slices, Toxicology, vol.216, pp.9-14, 2005.

E. C. Hirsch, S. Hunot, and A. Hartmann, Neuroinflammatory processes in Parkinson's disease, Parkinsonism Relat. Disord, vol.11, 2005.

H. Hoepken, S. Gispert, M. Azizov, M. Klinkenberg, F. Ricciardi et al., Parkinson patient fibroblasts show increased alpha-synuclein expression, Exp. Neurol, vol.212, pp.307-313, 2008.

B. Holdorff, Friedrich Heinrich Lewy (1885-1950) and his work, J. Hist. Neurosci, vol.11, pp.19-28, 2002.

R. G. Holloway, I. Shoulson, S. Fahn, K. Kieburtz, A. Lang et al., Arch. Neurol, vol.61, pp.1044-1053, 2004.

P. Holmans, V. Moskvina, L. Jones, M. Sharma, . Parkinson's-disease-genomics-consortium et al., A pathway-based analysis provides additional support for an immune-related genetic susceptibility to Parkinson's disease, Hum. Mol. Genet, vol.22, pp.1039-1049, 2013.

S. Holmqvist, O. Chutna, L. Bousset, P. Aldrin-kirk, W. Li et al., Direct evidence of Parkinson pathology spread from the gastrointestinal tract to the brain in rats, Acta Neuropathol. (Berl, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01155404

B. Holst, A. Cygankiewicz, T. H. Jensen, M. Ankersen, and T. W. Schwartz, High Constitutive Signaling of the Ghrelin Receptor-Identification of a Potent Inverse Agonist, Mol. Endocrinol, vol.17, pp.2201-2210, 2003.

C. L. Hoppel, D. Grinblatt, H. C. Kwok, P. K. Arora, M. P. Singh et al., Inhibition of mitochondrial respiration by analogs of 4-phenylpyridine and 1-methyl-4-phenylpyridinium cation (MPP+), the neurotoxic metabolite of MPTP, Biochem. Biophys. Res. Commun, vol.148, pp.684-693, 1987.

H. Hosoda and K. Kangawa, Standard sample collections for blood ghrelin measurements, Methods Enzymol, vol.514, pp.113-126, 2012.

A. Howard, S. Feighner, D. Cully, J. Arena, P. Liberator et al., A receptor in pituitary and hypothalamus that functions in growth hormone release, vol.16, pp.974-977, 1996.

K. Hoyles and J. C. Sharma, Olfactory loss as a supporting feature in the diagnosis of Parkinson's disease: a pragmatic approach, J. Neurol, vol.260, pp.2951-2958, 2013.

S. Hwang, M. Moon, S. Kim, L. Hwang, K. J. Ahn et al., Neuroprotective effect of ghrelin is associated with decreased expression of prostate apoptosis response-4, Endocr. J, vol.56, pp.609-617, 2009.

P. Ibáñez, S. Lesage, S. Janin, E. Lohmann, F. Durif et al., Alpha-synuclein gene rearrangements in dominantly inherited parkinsonism: frequency, phenotype, and mechanisms, Arch. Neurol, vol.66, pp.102-108, 2009.

A. Ieraci, A. Mallei, and M. Popoli, Social Isolation Stress Induces Anxious-Depressive-Like Behavior and Alterations of Neuroplasticity-Related Genes in Adult Male Mice, Neural Plast, p.6212983, 2016.

K. Ikemoto, I. Nagatsu, A. Nishimura, K. Nishi, and R. Arai, Do all of human midbrain tyrosine hydroxylase neurons synthesize dopamine?, Brain Res, vol.805, pp.255-258, 1998.

M. Iljina, G. A. Garcia, M. H. Horrocks, L. Tosatto, M. L. Choi et al., Kinetic model of the aggregation of alpha-synuclein provides insights into prion-like spreading, Proc. Natl. Acad. Sci. U. S. A, vol.113, pp.1206-1215, 2016.

A. Inui, Ghrelin: An orexigenic and somatotrophic signal from the stomach, Nat. Rev. Neurosci, vol.2, pp.551-560, 2001.

D. J. Irwin, V. M. Lee, .. Trojanowski, and J. Q. , Parkinson's disease dementia: convergence of ?synuclein, tau and amyloid-? pathologies, Nat. Rev. Neurosci, vol.14, pp.626-636, 2013.

V. Jackson-lewis, M. Jakowec, R. E. Burke, and S. Przedborski, Time course and morphology of dopaminergic neuronal death caused by the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, Neurodegener. J. Neurodegener. Disord. Neuroprotection Neuroregeneration, vol.4, pp.257-269, 1995.

F. Jacob, Souris, la Mouche et l'Homme, 1996.

S. A. Jagmag, N. Tripathi, S. D. Shukla, S. Maiti, and S. Khurana, Evaluation of Models of Parkinson's Disease, Front. Neurosci, vol.9, 2016.

S. Jain, Y. Park, S. Comer, D. Rudoy, D. Ivanco et al., Identifying drug-naïve Parkinsonism with Non-motor features & CSF biomarkers: A case-control study, 2015.

W. Jang, H. J. Kim, H. Li, K. D. Jo, M. K. Lee et al., The Neuroprotective Effect of Erythropoietin on Rotenone-Induced Neurotoxicity in SH-SY5Y Cells Through the Induction of Autophagy, Mol. Neurobiol, vol.53, pp.3812-3821, 2015.

L. M. Jaremka, M. A. Belury, R. R. Andridge, W. B. Malarkey, R. Glaser et al., Interpersonal stressors predict ghrelin and leptin levels in women, Psychoneuroendocrinology, vol.48, pp.178-188, 2014.

H. Jiang, L. Betancourt, and R. G. Smith, Ghrelin Amplifies Dopamine Signaling by Cross Talk Involving Formation of Growth Hormone Secretagogue Receptor/Dopamine Receptor Subtype 1 Heterodimers, Mol. Endocrinol, vol.20, pp.1772-1785, 2006.

H. Jiang, L. Li, J. Wang, and J. Xie, Ghrelin antagonizes MPTP-induced neurotoxicity to the dopaminergic neurons in mouse substantia nigra, Exp. Neurol, vol.212, pp.532-537, 2008.

I. P. Johnson, Age-related neurodegenerative disease research needs aging models. Front, Aging Neurosci, vol.7, p.168, 2015.

J. P. Johnston, Some observations upon a new inhibitor of monoamine oxidase in brain tissue, Biochem. Pharmacol, vol.17, pp.1285-1297, 1968.

T. M. Kääriäinen, M. Käenmäki, M. M. Forsberg, N. Oinas, A. Tammimäki et al., Unpredictable rotational responses to L-dopa in the rat model of Parkinson's disease: the role of L-dopa pharmacokinetics and striatal dopamine depletion, Basic Clin. Pharmacol. Toxicol, vol.110, pp.162-170, 2012.

G. Kadas, M. Klotz, K. Schäfer, K. Fassbender, D. Thomi et al., Do different assays for human acylated ghrelin concentrations provide comparable results?, Neuro Endocrinol. Lett, vol.35, pp.37-41, 2014.

M. E. Kalaitzakis, M. B. Graeber, S. M. Gentleman, and R. K. Pearce, Striatal beta-amyloid deposition in Parkinson disease with dementia, J. Neuropathol. Exp. Neurol, vol.67, pp.155-161, 2008.

L. V. Kalia, S. K. Kalia, P. J. Mclean, A. M. Lozano, and A. E. Lang, ?-Synuclein oligomers and clinical implications for Parkinson disease, Ann. Neurol, vol.73, pp.155-169, 2013.

L. V. Kalia and A. E. Lang, Parkinson's disease, Lancet Lond. Engl, vol.386, issue.14, pp.61393-61396, 2015.

K. Kalinderi, S. Bostantjopoulou, and L. Fidani, The genetic background of Parkinson's disease: current progress and future prospects, Acta Neurol. Scand, 2016.

J. Kamegai, I. Wakabayashi, R. D. Kineman, and L. A. Frohman, Growth hormone-releasing hormone receptor (GHRH-R) and growth hormone secretagogue receptor (GHS-R) mRNA levels during postnatal development in male and female rats, J. Neuroendocrinol, vol.11, pp.299-306, 1999.

J. Kang, D. J. Irwin, A. S. Chen-plotkin, A. Siderowf, C. Caspell et al., Association of cerebrospinal fluid ?-amyloid 1-42, T-tau, P-tau181, and ?-synuclein levels with clinical features of drug-naive patients with early Parkinson disease, JAMA Neurol, vol.70, pp.1277-1287, 2013.

H. Karasawa, C. Pietra, C. Giuliano, S. Garcia-rubio, X. Xu et al., New ghrelin agonist, HM01 alleviates constipation and L-dopa-delayed gastric emptying in 6-hydroxydopamine rat model of Parkinson's disease, Neurogastroenterol. Motil, 2014.

A. Karch, F. Llorens, M. Schmitz, A. S. Arora, S. Zafar et al., Stratification by Genetic and Demographic Characteristics Improves Diagnostic Accuracy of Cerebrospinal Fluid Biomarkers in Rapidly Progressive Dementia, J. Alzheimers Dis. JAD, vol.54, pp.1385-1393, 2016.

S. Kay, M. Josefine, S. Siswanto, K. Reijko, H. Helmut et al., The brainstem pathologies of Parkinson's disease and dementia with Lewy bodies, Brain Pathol. Zurich Switz, vol.25, pp.121-135, 2015.

P. C. Keane, M. Kurzawa, P. G. Blain, and C. M. Morris, Mitochondrial Dysfunction in Parkinson's Disease. Park. Dis, 2011.

R. Kenny, G. Cai, J. A. Bayliss, M. Clarke, Y. L. Choo et al., Endogenous ghrelin's role in hippocampal neuroprotection after global cerebral ischemia: does endogenous ghrelin protect against global stroke?, AJP Regul. Integr. Comp. Physiol, vol.304, 2013.

A. Kern, R. Albarran-zeckler, H. E. Walsh, and R. G. Smith, Apo-Ghrelin Receptor Forms Heteromers with DRD2 in Hypothalamic Neurons and Is Essential for Anorexigenic Effects of DRD2 Agonism, Neuron, vol.73, pp.317-332, 2012.

C. Kim, E. Rockenstein, B. Spencer, H. Kim, A. Adame et al., Antagonizing Neuronal Toll-like Receptor 2 Prevents Synucleinopathy by Activating Autophagy, Cell Rep, vol.13, pp.771-782, 2015.

D. Kim, J. H. Paik, D. Shin, H. Kim, C. Park et al., What is the Clinical Significance of Cerebrospinal Fluid Biomarkers in Parkinson's disease? Is the Significance Diagnostic or Prognostic?, Exp. Neurobiol, vol.23, pp.352-364, 2014.

G. J. Kim, S. Y. Park, H. Kim, Y. H. Chun, and S. H. Park, Chromosomal aberrations in neuroblastoma cell lines identified by cross species color banding and chromosome painting, Cancer Genet. Cytogenet, vol.129, pp.10-16, 2001.

J. Kim and H. Sung, Gastrointestinal Autonomic Dysfunction in Patients with Parkinson's Disease, J. Mov. Disord, vol.8, pp.76-82, 2015.

J. Kim and B. S. Jeon, Complementary and alternative medicine in Parkinson's disease patients in Korea, Curr. Neurol. Neurosci. Rep, vol.12, pp.631-632, 2012.

M. S. Kim, C. Y. Yoon, P. G. Jang, Y. J. Park, C. S. Shin et al., The mitogenic and antiapoptotic actions of ghrelin in 3T3-L1 adipocytes, Mol. Endocrinol. Baltim. Md, vol.18, pp.2291-2301, 2004.

R. H. Kim, P. D. Smith, H. Aleyasin, S. Hayley, M. P. Mount et al., Hypersensitivity of DJ-1-deficient mice to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyrindine (MPTP) and oxidative stress, Proc. Natl. Acad. Sci. U. S. A, vol.102, pp.5215-5220, 2005.

A. King, S. Maekawa, I. Bodi, C. Troakes, O. Curran et al., Simulated surgical-type cerebral biopsies from post-mortem brains allows accurate neuropathological diagnoses in the majority of neurodegenerative disease groups, Acta Neuropathol. Commun, vol.1, p.53, 2013.

D. Kirik, C. Rosenblad, C. Burger, C. Lundberg, T. E. Johansen et al., Parkinson-like neurodegeneration induced by targeted overexpression of ?-synuclein in the nigrostriatal system, J. Neurosci, vol.22, pp.2780-2791, 2002.

T. Kitada, S. Asakawa, N. Hattori, H. Matsumine, Y. Yamamura et al., Mutations in the parkin gene cause autosomal recessive juvenile parkinsonism, Nature, vol.392, pp.605-608, 1998.

T. Kitada, A. Pisani, D. R. Porter, H. Yamaguchi, A. Tscherter et al., Impaired dopamine release and synaptic plasticity in the striatum of PINK1-deficient mice, Proc. Natl. Acad. Sci. U. S. A, vol.104, pp.11441-11446, 2007.

D. J. Klionsky and S. D. Emr, Autophagy as a regulated pathway of cellular degradation, Science, vol.290, pp.1717-1721, 2000.

P. Klivenyi, D. Siwek, G. Gardian, L. Yang, A. Starkov et al., Mice lacking alpha-synuclein are resistant to mitochondrial toxins, Neurobiol. Dis, vol.21, pp.541-548, 2006.

T. P. Knowles, M. Vendruscolo, and C. M. Dobson, The amyloid state and its association with protein misfolding diseases, Nat. Rev. Mol. Cell Biol, vol.15, pp.384-396, 2014.

J. C. Koch, F. Bitow, J. Haack, Z. Hedouville, J. Zhang et al., Alpha-Synuclein affects neurite morphology, autophagy, vesicle transport and axonal degeneration in CNS neurons, Cell Death Dis, vol.6, p.1811, 2015.

S. Koga, N. Aoki, R. J. Uitti, J. A. Van-gerpen, W. P. Cheshire et al., When DLB, PD, and PSP masquerade as MSA: an autopsy study of 134 patients, Neurology, vol.85, pp.404-412, 2015.

J. Koikkalainen, H. Rhodius-meester, A. Tolonen, F. Barkhof, B. Tijms et al., Differential diagnosis of neurodegenerative diseases using structural MRI data, NeuroImage Clin, vol.11, pp.435-449, 2016.

M. Kojima, H. Hosoda, Y. Date, M. Nakazato, and H. Matsuo, Ghelin is a gowth-hormonereleasing acylated peptide from stomach, Nature, vol.402, pp.656-660, 1999.

M. Kojima and K. Kangawa, Ghrelin: structure and function, Physiol. Rev, vol.85, pp.495-522, 2005.

J. H. Kordower, Y. Chu, R. A. Hauser, T. B. Freeman, and C. W. Olanow, Lewy body-like pathology in long-term embryonic nigral transplants in Parkinson's disease, Nat. Med, vol.14, pp.504-506, 2008.

J. H. Kordower, T. B. Freeman, B. J. Snow, F. J. Vingerhoets, E. J. Mufson et al., Neuropathological evidence of graft survival and striatal reinnervation after the transplantation of fetal mesencephalic tissue in a patient with Parkinson's disease, N. Engl. J. Med, vol.332, pp.1118-1124, 1995.

J. A. Korecka, R. E. Van-kesteren, E. Blaas, S. O. Spitzer, J. H. Kamstra et al., Phenotypic Characterization of Retinoic Acid Differentiated SH-SY5Y Cells by Transcriptional Profiling, PLoS ONE, vol.8, 2013.

R. Kortekaas, K. L. Leenders, J. C. Van-oostrom, W. Vaalburg, J. Bart et al., Blood-brain barrier dysfunction in parkinsonian midbrain in vivo, Ann. Neurol, vol.57, pp.176-179, 2005.

P. T. Kotzbauer, N. J. Cairns, M. C. Campbell, A. W. Willis, B. A. Racette et al., Pathologic accumulation of ?-synuclein and A? in Parkinson disease patients with dementia, Arch. Neurol, vol.69, pp.1326-1331, 2012.

T. Kovács, N. J. Cairns, and P. L. Lantos, Olfactory centres in Alzheimer's disease: olfactory bulb is involved in early Braak's stages, Neuroreport, vol.12, pp.285-288, 2001.

P. Krack, A. Batir, N. Van-blercom, S. Chabardes, V. Fraix et al., Five-year follow-up of bilateral stimulation of the subthalamic nucleus in advanced Parkinson's disease, N. Engl. J. Med, vol.349, pp.1925-1934, 2003.

S. M. Kranick and J. E. Duda, Olfactory dysfunction in Parkinson's disease, Neurosignals, vol.16, pp.35-40, 2008.

T. Lebouvier, M. Tasselli, S. Paillusson, H. Pouclet, M. Neunlist et al., Biopsable Neural Tissues: Toward New Biomarkers for Parkinson's Disease? Front, Psychiatry, vol.1, 2010.

C. S. Lee, H. Sauer, and A. Bjorklund, Dopaminergic neuronal degeneration and motor impairments following axon terminal lesion by instrastriatal 6-hydroxydopamine in the rat, Neuroscience, vol.72, pp.641-653, 1996.

H. Lee, J. Suk, C. Patrick, E. Bae, J. Cho et al., Direct transfer of alpha-synuclein from neuron to astroglia causes inflammatory responses in synucleinopathies, J. Biol. Chem, vol.285, pp.9262-9272, 2010.

M. K. Lee, W. Stirling, Y. Xu, X. Xu, D. Qui et al., Human alpha-synuclein-harboring familial Parkinson's disease-linked Ala-53 --> Thr mutation causes neurodegenerative disease with alpha-synuclein aggregation in transgenic mice, Proc. Natl. Acad. Sci. U. S. A, vol.99, pp.8968-8973, 2002.

A. F. Leentjens, K. Dujardin, L. Marsh, P. Martinez-martin, I. H. Richard et al., Anxiety rating scales in Parkinson's disease: Critique and recommendations, Mov. Disord, vol.23, 2008.

A. J. Lees, J. Hardy, T. Revesz, A. J. Lees, M. Selikhova et al., The black stuff and Konstantin Nikolaevich Tretiakoff, Mov. Disord. Off. J. Mov. Disord. Soc, vol.373, pp.777-783, 2008.

F. Lemarié, E. Beauchamp, P. Legrand, and V. Rioux, Revisiting the metabolism and physiological functions of caprylic acid (C8:0) with special focus on ghrelin octanoylation, Biochimie, vol.120, pp.40-48, 2016.

L. R. Lemkau, G. Comellas, K. D. Kloepper, W. S. Woods, J. M. George et al., Mutant protein A30P ?-synuclein adopts wild-type fibril structure, despite slower fibrillation kinetics, J. Biol. Chem, vol.287, pp.11526-11532, 2012.

L. R. Lemkau, G. Comellas, S. W. Lee, L. K. Rikardsen, W. S. Woods et al., Site-specific perturbations of alpha-synuclein fibril structure by the Parkinson's disease associated mutations A53T and E46K, PloS One, vol.8, p.49750, 2013.

E. Leroy, D. Anastasopoulos, S. Konitsiotis, C. Lavedan, and M. H. Polymeropoulos, Deletions in the Parkin gene and genetic heterogeneity in a Greek family with early onset Parkinson's disease, Hum. Genet, vol.103, pp.424-427, 1998.

S. Lesage, M. Anheim, F. Letournel, L. Bousset, A. Honoré et al., French Parkinson's Disease Genetics Study Group, 2013. G51D ?-synuclein mutation causes a novel parkinsonian-pyramidal syndrome, Ann. Neurol, vol.73, pp.459-471

S. Lesage, V. Drouet, E. Majounie, V. Deramecourt, M. Jacoupy et al., Loss of VPS13C Function in Autosomal-Recessive Parkinsonism Causes Mitochondrial Dysfunction and Increases PINK1/Parkin-Dependent Mitophagy, Am. J. Hum. Genet, vol.98, pp.500-513, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01289266

S. Levi, M. Cox, M. Lugon, M. Hodkinson, and A. Tomkins, Increased energy expenditure in Parkinson's disease, BMJ, vol.301, pp.1256-1257, 1990.

F. Levin, T. Edholm, P. T. Schmidt, P. Grybäck, H. Jacobsson et al., Ghrelin Stimulates Gastric Emptying and Hunger in Normal-Weight Humans, J. Clin. Endocrinol. Metab, vol.91, pp.3296-3302, 2006.

J. Levin, A. Kurz, T. Arzberger, A. Giese, and G. U. Höglinger, The Differential Diagnosis and Treatment of Atypical Parkinsonism, Dtsch. Ärztebl. Int, vol.113, pp.61-69, 2016.

B. Levine and D. J. Klionsky, Development by self-digestion: molecular mechanisms and biological functions of autophagy, Dev. Cell, vol.6, pp.463-477, 2004.

P. A. Lewitt, Levodopa therapy for Parkinson's disease: Pharmacokinetics and pharmacodynamics, Mov. Disord. Off. J. Mov. Disord. Soc, vol.30, pp.64-72, 2015.

F. Lewy, Paralysis agitans. I. Pathologische Anatomie, Handbuch der Neurologie. Lewandowski M, pp.920-933, 1912.

F. H. Lewy, Zur pathologisch-anatomischen Differentialdiagnose der Paralysis agitans und der Huntingtonschen Chorea, Z. Für Gesamte Neurol. Psychiatr, vol.73, pp.170-187, 1921.

F. Li, H. Ji, and L. Shen, A Meta-Analysis of Tea Drinking and Risk of Parkinson's, Disease. Sci. World J, 2012.

J. Li, E. Englund, H. Widner, S. Rehncrona, A. Björklund et al., Characterization of Lewy body pathology in 12-and 16-year-old intrastriatal mesencephalic grafts surviving in a patient with Parkinson's disease, Mov. Disord. Off. J. Mov. Disord. Soc, vol.25, pp.1091-1096, 2010.

N. Li, Mitochondrial Complex I Inhibitor Rotenone Induces Apoptosis through Enhancing Mitochondrial Reactive Oxygen Species Production, J. Biol. Chem, vol.278, pp.8516-8525, 2003.

Y. Li, W. Liu, T. F. Oo, L. Wang, Y. Tang et al., Mutant LRRK2(R1441G) BAC transgenic mice recapitulate cardinal features of Parkinson's disease, Nat. Neurosci, vol.12, pp.826-828, 2009.

P. Limousin, P. Krack, P. Pollak, A. Benazzouz, C. Ardouin et al., Electrical stimulation of the subthalamic nucleus in advanced Parkinson's disease, N. Engl. J. Med, vol.339, pp.1105-1111, 1998.

P. Limousin, P. Pollak, A. Benazzouz, D. Hoffmann, E. Broussolle et al., Bilateral subthalamic nucleus stimulation for severe Parkinson's disease, Mov. Disord. Off. J. Mov. Disord. Soc, vol.10, pp.672-674, 1995.

C. Lin, J. Lin, Y. Liu, C. Chang, and R. Wu, Risk of Parkinson's disease following anxiety disorders: a nationwide population-based cohort study, Eur. J. Neurol, vol.22, pp.1280-1287, 2015.

O. Lindvall and A. Björklund, Cell therapy in Parkinson's disease, NeuroRx J. Am. Soc. Exp. Neurother, vol.1, pp.382-393, 2004.

L. Liu, J. Song, J. Lu, Y. Huang, Y. Zeng et al., Tianma Gouteng Yin, a Traditional Chinese Medicine decoction, exerts neuroprotective effects in animal and cellular models of Parkinson's disease, Sci. Rep, vol.5, p.16862, 2015.

Y. Liu, D. Peter, A. Roghani, S. Schuldiner, G. G. Privé et al., A cDNA that suppresses MPP+ toxicity encodes a vesicular amine transporter, Cell, vol.70, pp.539-551, 1992.

K. J. Livak and T. D. Schmittgen, Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method, Methods San Diego Calif, vol.25, pp.402-408, 2001.

F. Llorens, M. Schmitz, D. Varges, N. Kruse, N. Gotzmann et al., Cerebrospinal ?-synuclein in ?-synuclein aggregation disorders: tau/?-synuclein ratio as potential biomarker for dementia with Lewy bodies, J. Neurol, vol.263, pp.2271-2277, 2016.

C. Lo-bianco, J. Ridet, B. L. Schneider, N. Deglon, and P. Aebischer, alpha -Synucleinopathy and selective dopaminergic neuron loss in a rat lentiviral-based model of Parkinson's disease, Proc. Natl. Acad. Sci. U. S. A, vol.99, pp.10813-10818, 2002.

C. M. Long-smith, A. M. Sullivan, and Y. M. Nolan, The influence of microglia on the pathogenesis of Parkinson's disease, Prog. Neurobiol, vol.89, pp.277-287, 2009.

F. M. Lopes, R. Schröder, M. L. Júnior, F. Da, A. Zanotto-filho et al., Comparison between proliferative and neuron-like SH-SY5Y cells as an in vitro model for Parkinson disease studies, Brain Res, vol.1337, pp.85-94, 2010.

N. Lorenzen, S. B. Nielsen, A. K. Buell, J. D. Kaspersen, P. Arosio et al., The role of stable ?-synuclein oligomers in the molecular events underlying amyloid formation, J. Am. Chem. Soc, vol.136, pp.3859-3868, 2014.

J. Lotharius, S. Barg, P. Wiekop, C. Lundberg, H. K. Raymon et al., Effect of mutant alpha-synuclein on dopamine homeostasis in a new human mesencephalic cell line, J. Biol. Chem, vol.277, pp.38884-38894, 2002.

X. Lu, S. M. Fleming, B. Meurers, L. C. Ackerson, F. Mortazavi et al., Bacterial artificial chromosome transgenic mice expressing a truncated mutant parkin exhibit age-dependent hypokinetic motor deficits, dopaminergic neuron degeneration, and accumulation of proteinase K-resistant alpha-synuclein, J. Neurosci. Off. J. Soc. Neurosci, vol.29, pp.1962-1976, 2009.

P. Luoma, A. Melberg, J. O. Rinne, J. A. Kaukonen, N. N. Nupponen et al., Parkinsonism, premature menopause, and mitochondrial DNA polymerase gamma mutations: clinical and molecular genetic study, Lancet Lond. Engl, vol.364, pp.16983-16986, 2004.

E. S. Luth, I. G. Stavrovskaya, T. Bartels, B. S. Kristal, and D. J. Selkoe, Soluble, Prefibrillar ?-Synuclein Oligomers Promote Complex I-dependent, Ca2+-induced Mitochondrial Dysfunction, J. Biol. Chem, vol.289, pp.21490-21507, 2014.

J. Luthman, A. Fredriksson, E. Sundström, G. Jonsson, and T. Archer, Selective lesion of central dopamine or noradrenaline neuron systems in the neonatal rat: motor behavior and monoamine alterations at adult stage, Behav. Brain Res, vol.33, pp.267-277, 1989.

I. Mackenzie, The pathology of Parkinson's disease. Br. Columbia Med, J, vol.43, pp.142-147, 2001.

J. Makovey, V. Naganathan, M. Seibel, and P. Sambrook, Gender differences in plasma ghrelin and its relations to body composition and bone -an opposite-sex twin study, Clin. Endocrinol. (Oxf.), vol.66, pp.530-537, 2007.

B. K. Mani, A. K. Walker, E. J. Lopez-soto, J. Raingo, C. E. Lee et al., Neuroanatomical characterization of a growth hormone secretagogue receptorgreen fluorescent protein reporter mouse: Characterization of Ghsr-egfp reporter mouse, J. Comp. Neurol, vol.522, pp.3644-3666, 2014.

A. B. Manning-bo?, W. M. Caudle, X. A. Perez, S. H. Reaney, R. Paletzki et al., Increased vulnerability of nigrostriatal terminals in DJ-1-deficient mice is mediated by the dopamine transporter, Neurobiol. Dis, vol.27, pp.141-150, 2007.

A. B. Manning-bog, A. L. Mccormack, J. Li, V. N. Uversky, A. L. Fink et al., The herbicide paraquat causes up-regulation and aggregation of alpha-synuclein in mice: paraquat and alpha-synuclein, J. Biol. Chem, vol.277, pp.1641-1644, 2002.

K. Marder, M. Tang, H. Mejia, B. Alfaro, L. Cote et al., Risk of Parkinson's disease among first-degree relatives A community-based study, Neurology, vol.47, pp.155-160, 1996.

C. Marin and E. Aguilar, In vivo 6-OHDA-induced neurodegeneration and nigral autophagic markers expression, Neurochem. Int, vol.58, pp.521-526, 2011.

E. Markaki, J. Ellul, Z. Kefalopoulou, E. Trachani, A. Theodoropoulou et al., The Role of Ghrelin, Neuropeptide Y and Leptin Peptides in Weight Gain after Deep Brain Stimulation for Parkinson's Disease, Stereotact. Funct. Neurosurg, vol.90, pp.104-112, 2012.

H. S. Markus, M. Cox, and A. M. Tomkins, Raised resting energy expenditure in Parkinson's disease and its relationship to muscle rigidity, Clin. Sci, vol.83, pp.199-204, 1992.

M. Martinez-vicente, Autophagy in neurodegenerative diseases: From pathogenic dysfunction to therapeutic modulation, Semin. Cell Dev. Biol, vol.40, pp.115-126, 2015.

M. Martinez-vicente and A. M. Cuervo, Autophagy and neurodegeneration: when the cleaning crew goes on strike, Lancet Neurol, vol.6, pp.352-361, 2007.

L. Martins, D. Fernández-mallo, M. G. Novelle, M. J. Vázquez, M. Tena-sempere et al., Hypothalamic mTOR Signaling Mediates the Orexigenic Action of Ghrelin, PLoS ONE, vol.7, 2012.

S. Mary, J. Fehrentz, M. Damian, G. Gaibelet, H. Orcel et al., Heterodimerization with Its Splice Variant Blocks the Ghrelin Receptor 1a in a Non-signaling Conformation, J. Biol. Chem, vol.288, pp.24656-24665, 2013.

E. Masliah, E. Rockenstein, M. Mante, L. Crews, B. Spencer et al., Passive immunization reduces behavioral and neuropathological deficits in an alpha-synuclein transgenic model of Lewy body disease, PloS One, vol.6, 2011.

E. Masliah, E. Rockenstein, I. Veinbergs, M. Mallory, M. Hashimoto et al., Dopaminergic loss and inclusion body formation in alpha-synuclein mice: implications for neurodegenerative disorders, Science, vol.287, pp.1265-1269, 2000.

N. Maswood, J. Young, E. Tilmont, Z. Zhang, D. M. Gash et al., Caloric restriction increases neurotrophic factor levels and attenuates neurochemical and behavioral deficits in a primate model of Parkinson's disease, Proc. Natl. Acad. Sci. U. S. A, vol.101, pp.18171-18176, 2004.

H. Matsumine, A Parkin gene (PARK 2) and Parkinson's disease, 1999.

, Rinsh? Shinkeigaku Clin. Neurol, vol.39, pp.9-12

H. Matsumine, M. Saito, S. Shimoda-matsubayashi, H. Tanaka, A. Ishikawa et al., Localization of a gene for an autosomal recessive form of juvenile Parkinsonism to chromosome 6q25.2-27, Am. J. Hum. Genet, vol.60, pp.588-596, 1997.

R. Mayeux, J. Denaro, N. Hemenegildo, K. Marder, M. X. Tang et al., A population-based investigation of Parkinson's disease with and without dementia. Relationship to age and gender, Arch. Neurol, vol.49, pp.492-497, 1992.

R. W. Mccallum, A. Lembo, T. Esfandyari, B. R. Bhandari, N. Ejskjaer et al., Phase 2b, randomized, double-blind 12-week studies of TZP-102, a ghrelin receptor agonist for diabetic gastroparesis, Neurogastroenterol. Motil, vol.25, pp.705-717, 2013.

A. L. Mccormack and D. A. Di-monte, Effects of L-dopa and other amino acids against paraquatinduced nigrostriatal degeneration, J. Neurochem, vol.85, pp.82-86, 2003.

W. T. Mckinney, Animal models of depression: an overview, Psychiatr. Dev, vol.2, pp.77-96, 1984.

D. A. Mcritchie, H. R. Cartwright, and G. M. Halliday, Specific A10 dopaminergic nuclei in the midbrain degenerate in Parkinson's disease, Exp. Neurol, vol.144, pp.202-213, 1997.

Y. Mear, A. Enjalbert, and S. Thirion, GHS-R1a constitutive activity and its physiological relevance, Front. Neurosci, vol.7, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00966209

M. Méquinion, E. Caron, S. Zgheib, A. Stievenard, P. Zizzari et al., Physical activity: benefit or weakness in metabolic adaptations in a mouse model of chronic food restriction?, Am. J. Physiol. -Endocrinol, 2015.

. Metab, , vol.308, pp.241-255

M. Méquinion, F. Langlet, S. Zgheib, S. Dickson, B. Dehouck et al., Ghrelin: Central and Peripheral Implications in Anorexia Nervosa, 2013.

G. E. Meredith, S. Totterdell, E. Petroske, K. Santa-cruz, R. C. Callison et al., Lysosomal malfunction accompanies alpha-synuclein aggregation in a progressive mouse model of Parkinson's disease, Brain Res, vol.956, pp.156-165, 2002.

M. Meulener, A. J. Whitworth, C. E. Armstrong-gold, P. Rizzu, P. Heutink et al., Drosophila DJ-1 mutants are selectively sensitive to environmental toxins associated with Parkinson's disease, Curr. Biol. CB, vol.15, pp.1572-1577, 2005.

E. Mezey, A. M. Dehejia, G. Harta, N. Tresser, S. F. Suchy et al., Alpha synuclein is present in Lewy bodies in sporadic Parkinson's disease, Mol. Psychiatry, vol.3, pp.493-499, 1998.

M. Mogi, M. Harada, H. Narabayashi, H. Inagaki, M. Minami et al., Interleukin (IL)-1?, IL-2, IL-4, IL-6 and transforming growth factor-? levels are elevated in ventricular cerebrospinal fluid in juvenile parkinsonism and Parkinson's disease, Neurosci. Lett, vol.211, pp.13-16, 1996.

A. Molfino, A. Formiconi, F. R. Fanelli, and M. Muscaritoli, Ghrelin: from discovery to cancer cachexia therapy, Curr. Opin. Clin. Nutr. Metab. Care, vol.17, pp.471-476, 2014.

C. Moolenbeek and E. J. Ruitenberg, The "Swiss roll": a simple technique for histological studies of the rodent intestine, Lab. Anim, vol.15, pp.57-59, 1981.

M. Moon, H. G. Kim, L. Hwang, J. Seo, S. Kim et al., Neuroprotective Effect of Ghrelin in the 1-Methyl-4-Phenyl-1,2,3,6-Tetrahydropyridine Mouse Model of Parkinson's Disease by Blocking Microglial Activation, Neurotox. Res, vol.15, pp.332-347, 2009.

J. F. Morley and J. E. Duda, Olfaction as a biomarker in Parkinson's disease, Biomark. Med, vol.4, pp.661-670, 2010.

P. Morrish, The meaning of negative DAT SPECT and F-Dopa PET scans in patients with clinical Parkinson's disease, Mov. Disord. Off. J. Mov. Disord. Soc, vol.20, 0117.

A. Mougenot, S. Nicot, A. Bencsik, E. Morignat, J. Verchère et al., Prion-like acceleration of a synucleinopathy in a transgenic mouse model, Neurobiol. Aging, vol.33, pp.2225-2228, 2012.

A. J. Mougenot, A. Bencsik, S. Nicot, J. Vulin, E. Morignat et al., Transmission of prion strains in a transgenic mouse model overexpressing human A53T mutated ?-synuclein, J. Neuropathol. Exp. Neurol, vol.70, pp.377-385, 2011.

, The Unified Parkinson's Disease Rating Scale (UPDRS): status and recommendations, Movement Disorder Society Task Force on Rating Scales for Parkinson's Disease, vol.18, pp.738-750, 2003.

T. D. Müller, R. Nogueiras, M. L. Andermann, Z. B. Andrews, S. D. Anker et al., Ghrelin. Mol. Metab, vol.4, pp.437-460, 2015.

E. Muñoz, R. Oliva, V. Obach, M. J. Martí, P. Pastor et al., Identification of Spanish familial Parkinson's disease and screening for the Ala53Thr mutation of the alphasynuclein gene in early onset patients, Neurosci. Lett, vol.235, pp.57-60, 1997.

A. B. Muñoz-manchado, J. Villadiego, S. Romo-madero, N. Suárez-luna, A. Bermejo-navas et al., Chronic and progressive Parkinson's disease MPTP model in adult and aged mice, J. Neurochem, vol.136, pp.373-387, 2016.

J. Munro, K. Steeghs, V. Morrison, H. Ireland, and K. Parkinsn, Human fibroblast replicative senescence can occur in the absence of extensive cell division and short telomeres, Publ. Online, vol.14, 2001.

E. Mutez, L. Larvor, F. Leprêtre, V. Mouroux, D. Hamalek et al., Transcriptional profile of Parkinson blood mononuclear cells with LRRK2 mutation, Neurobiol. Aging, vol.32, pp.1839-1848, 2011.

E. Mutez, A. Nkiliza, K. Belarbi, A. De-broucker, C. Vanbesien-mailliot et al., Involvement of the immune system, endocytosis and EIF2 signaling in both genetically determined and sporadic forms of Parkinson's disease, Neurobiol. Dis, vol.63, pp.165-170, 2014.

T. Nagatsu, M. Mogi, H. Ichinose, and A. Togari, Cytokines in Parkinson's disease, J. Neural Transm, pp.143-151, 2000.

K. Nakamura, V. P. Bindokas, J. D. Marks, D. A. Wright, D. M. Frim et al., The selective toxicity of 1-methyl-4-phenylpyridinium to dopaminergic neurons: the role of mitochondrial complex I and reactive oxygen species revisited, Mol. Pharmacol, vol.58, pp.271-278, 2000.

M. A. Nalls, N. Pankratz, C. M. Lill, C. B. Do, D. G. Hernandez et al., International Parkinson's Disease Genomics Consortium (ipdgc), Parkinson's Study Group (PSG) Parkinson's Research: The Organized GENetics Initiative (progeni), 23andMe, GenePD, NeuroGenetics Research Consortium (ngrc), Hussman Institute of Human Genomics (hihg), vol.46, pp.989-993, 2014.

D. Narendra, A. Tanaka, D. Suen, and R. J. Youle, Parkin is recruited selectively to impaired mitochondria and promotes their autophagy, J. Cell Biol, vol.183, pp.795-803, 2008.

, What are the Different Types of Atypical Parkinsonism Syndromes? | National Parkinson Foundation

A. Navarro and A. Boveris, Brain mitochondrial dysfunction in aging, neurodegeneration, and Parkinson's disease. Front, Aging Neurosci, vol.2, 2010.

C. Ng, S. Z. Mok, C. Koh, X. Ouyang, M. L. Fivaz et al., Parkin protects against LRRK2 G2019S mutant-induced dopaminergic neurodegeneration in Drosophila, J. Neurosci. Off. J. Soc. Neurosci, vol.29, pp.11257-11262, 2009.

K. Y. Ng, T. N. Chase, R. W. Colburn, and I. J. Kopin, L-Dopa-induced release of cerebral monoamines, Science, vol.170, pp.76-77, 1970.

K. Y. Ng, R. W. Colburn, and I. J. Kopin, Effects of L-dopa on efflux of cerebral monoamines from synaptosomes, Nature, vol.230, pp.331-332, 1971.

Y. Nishi, H. Mifune, A. Yabuki, Y. Tajiri, R. Hirata et al., Changes in Subcellular Distribution of n-Octanoyl or n-Decanoyl Ghrelin in Ghrelin-Producing Cells, 2013.

A. J. Noyce, A. J. Lees, and A. Schrag, The prediagnostic phase of Parkinson's disease, J. Neurol. Neurosurg. Psychiatry, 2016.

S. Nozaki, T. Saito, T. Matsumura, I. Miyai, and J. Kang, , 1999.

, Rinsho Shinkeigaku, vol.39, pp.1010-1014

R. L. Nussbaum and M. H. Polymeropoulos, Genetics of Parkinson's disease, Hum. Mol. Genet, vol.6, pp.1687-1691, 1997.

S. Nuvoli, A. Spanu, and G. Madeddu, Brain SPECT with perfusion radiopharmaceuticals and dopaminergic system radiocompounds in dementia disorders, Curr. Alzheimer Res, 2016.

J. A. Obeso, M. C. Rodríguez, A. Gorospe, J. Guridi, L. Alvarez et al., Surgical treatment of Parkinson's disease, Baillières Clin. Neurol, vol.6, pp.125-145, 1997.

W. Oertel and J. B. Schulz, Current and experimental treatments of Parkinson disease: A guide for neuroscientists, J. Neurochem, 2016.

N. U. Okubadejo, J. H. Bower, W. A. Rocca, and D. M. Maraganore, Parkinson's disease in Africa: A systematic review of epidemiologic and genetic studies, Mov. Disord. Off. J. Mov. Disord. Soc, vol.21, pp.2150-2156, 2006.

C. W. Olanow and K. S. Mcnaught, Ubiquitin-proteasome system and Parkinson's disease, Mov. Disord. Off. J. Mov. Disord. Soc, vol.21, pp.1806-1823, 2006.

C. W. Olanow and S. B. Prusiner, Is Parkinson's disease a prion disorder?, Proc. Natl. Acad. Sci. U. S. A, vol.106, pp.12571-12572, 2009.

J. Olesen and M. Leonardi, The burden of brain diseases in Europe, Eur. J. Neurol, vol.10, pp.471-477, 2003.

S. Olgiati, A. De-rosa, M. Quadri, C. Criscuolo, G. J. Breedveld et al., PARK20 caused by SYNJ1 homozygous Arg258Gln mutation in a new Italian family, Neurogenetics, vol.15, pp.183-188, 2014.

P. K. Olszewski, H. B. Schiöth, and A. S. Levine, Ghrelin in the CNS: from hunger to a rewarding and memorable meal?, Brain Res. Rev, vol.58, pp.160-170, 2008.

L. Oreland, Monoamine oxidase, dopamine and Parkinson's disease, Acta Neurol. Scand. Suppl, vol.136, pp.60-65, 1991.

C. F. Orr, D. B. Rowe, and G. M. Halliday, An inflammatory review of Parkinson's disease, Prog. Neurobiol, vol.68, pp.325-340, 2002.

A. Oueslati, K. E. Paleologou, B. L. Schneider, P. Aebischer, and H. A. Lashuel, Mimicking phosphorylation at serine 87 inhibits the aggregation of human ?-synuclein and protects against its toxicity in a rat model of Parkinson's disease, J. Neurosci. Off. J. Soc. Neurosci, vol.32, pp.1536-1544, 2012.

A. Özge, R. Bugdayci, E. Togrol, and M. Saracoglu, The relation of gastrointestinal symptoms to duration of loveo-dopa treatment and severity of Parkinson's disease, J. Appl. Res, vol.3, pp.349-355, 2003.

E. Özta? and T. Topal, A Cell Protective Mechanism in a Murine Model of Parkinson's Disease. Turk, J. Med. Sci, vol.33, pp.295-299, 2003.

F. Pagan, M. Hebron, E. H. Valadez, Y. Torres-yaghi, X. Huang et al., Nilotinib Effects in Parkinson's disease and Dementia with Lewy bodies, J. Park. Dis, vol.6, pp.503-517, 2016.

T. Pan, S. Kondo, W. Le, and J. Jankovic, The role of autophagy-lysosome pathway in neurodegeneration associated with Parkinson's disease, Brain J. Neurol, vol.131, 1969.

C. Pañeda, A. I. Arroba, L. M. Frago, A. M. Holm, J. Rømer et al., Growth hormone-releasing peptide-6 inhibits cerebellar cell death in aged rats, Neuroreport, vol.14, pp.1633-1635, 2003.

N. Pankratz, W. C. Nichols, S. K. Uniacke, C. Halter, J. Murrell et al., Genome-wide linkage analysis and evidence of gene-by-gene interactions in a sample of 362 multiplex Parkinson disease families, Hum. Mol. Genet, vol.12, pp.2599-2608, 2003.

N. Pankratz, J. B. Wilk, J. C. Latourelle, A. L. Destefano, C. Halter et al., Genomewide association study for susceptibility genes contributing to familial Parkinson disease, Hum. Genet, vol.124, pp.593-605, 2009.

F. Pan-montojo, O. Anichtchik, Y. Dening, L. Knels, S. Pursche et al., Progression of Parkinson's Disease Pathology Is Reproduced by Intragastric Administration of Rotenone in Mice, PLoS ONE, vol.5, 2010.

F. Pan-montojo, M. Schwarz, C. Winkler, M. Arnhold, G. A. O'sullivan et al., Environmental toxins trigger PD-like progression via increased alpha-synuclein release from enteric neurons in mice, Sci. Rep, vol.2, 2012.

J. Pantel, M. Legendre, S. Cabrol, L. Hilal, Y. Hajaji et al., Loss of constitutive activity of the growth hormone secretagogue receptor in familial short stature, J. Clin. Invest, vol.116, pp.760-768, 2006.
URL : https://hal.archives-ouvertes.fr/hal-02082146

S. Papapetropoulos, C. Paschalis, A. Athanassiadou, A. Papadimitriou, J. Ellul et al., Clinical phenotype in patients with ?-synuclein Parkinson's disease living in Greece in comparison with patients with sporadic Parkinson's disease, J. Neurol. Neurosurg. Psychiatry, vol.70, pp.662-665, 2001.

T. D. Papkovskaia, K. Chau, F. Inesta-vaquera, D. B. Papkovsky, D. G. Healy et al., G2019S leucine-rich repeat kinase 2 causes uncoupling protein-mediated mitochondrial depolarization, Hum. Mol. Genet, vol.21, pp.4201-4213, 2012.

J. Park, S. Y. Kim, G. Cha, S. B. Lee, S. Kim et al., Drosophila DJ-1 mutants show oxidative stress-sensitive locomotive dysfunction, Gene, vol.361, pp.133-139, 2005.

J. Park, S. B. Lee, S. Lee, Y. Kim, S. Song et al., Mitochondrial dysfunction in Drosophila PINK1 mutants is complemented by parkin, Nature, vol.441, pp.1157-1161, 2006.

J. Parkinson, An essay on the shaking palsy, J. Neuropsychiatry Clin. Neurosci, vol.14, pp.223-236, 2002.

, Pramipexole vs levodopa as initial treatment for Parkinson disease: A randomized controlled trial. Parkinson Study Group, JAMA, vol.284, pp.1931-1938, 2000.

L. Pasquali, S. Ruggieri, L. Murri, A. Paparelli, and F. Fornai, Does autophagy worsen or improve the survival of dopaminergic neurons?, Parkinsonism Relat. Disord, vol.15, pp.24-27, 2009.

W. Peelaerts, L. Bousset, A. Van-der-perren, A. Moskalyuk, R. Pulizzi et al., ?-Synuclein strains cause distinct synucleinopathies after local and systemic administration, Nature, vol.522, pp.340-344, 2015.

F. A. Perez and R. D. Palmiter, Parkin-deficient mice are not a robust model of parkinsonism, Proc. Natl. Acad. Sci. U. S. A, vol.102, pp.2174-2179, 2005.

C. Perier and M. Vila, Mitochondrial biology and Parkinson's disease. Cold Spring Harb, Perspect. Med, vol.2, p.9332, 2012.

M. Periquet, T. Fulga, L. Myllykangas, M. G. Schlossmacher, and M. B. Feany, Aggregated alphasynuclein mediates dopaminergic neurotoxicity in vivo, J. Neurosci. Off. J. Soc. Neurosci, vol.27, pp.3338-3346, 2007.

C. Perlemoine, F. Macia, F. Tison, I. Coman, D. Guehl et al., Effects of subthalamic nucleus deep brain stimulation and levodopa on energy production rate and substrate oxidation in Parkinson's disease, Br. J. Nutr, vol.93, pp.191-198, 2005.

S. Perrin, Preclinical research: Make mouse studies work, Nature, vol.507, pp.423-425, 2014.

Y. Pesah, T. Pham, H. Burgess, B. Middlebrooks, P. Verstreken et al., Drosophila parkin mutants have decreased mass and cell size and increased sensitivity to oxygen radical stress, Dev. Camb. Engl, vol.131, pp.2183-2194, 2004.

G. M. Petzinger and M. W. Jakowec, Animal models of basal ganglia injury and degeneration and their application to Parkinson's disease research, 2005.

R. Pfeiffer, . Zbigniew-wszolek, and M. Ebadi, Parkinson's Disease, Second Edition, second, 2012.

R. F. Pfeiffer, Gastrointestinal dysfunction in Parkinson's disease, Parkinsonism Relat. Disord, vol.17, pp.10-15, 2011.

P. Piccini, D. J. Burn, R. Ceravolo, D. Maraganore, and D. J. Brooks, The role of inheritance in sporadic Parkinson's disease: Evidence from a longitudinal study of dopaminergic function in twins, Ann. Neurol, vol.45, pp.577-582, 1999.

A. R. Pikstra, A. Van-der-hoorn, K. L. Leenders, and B. M. De-jong, Relation of 18-F-Dopa PET with hypokinesia-rigidity, tremor and freezing in Parkinson's disease, NeuroImage Clin, vol.11, pp.68-72, 2016.

A. Poehler, W. Xiang, P. Spitzer, V. E. May, H. Meixner et al., Autophagy modulates SNCA/?-synuclein release, thereby generating a hostile microenvironment, Autophagy, vol.10, pp.2171-2192, 2014.

W. Poewe, Non-motor symptoms in Parkinson's disease, Eur. J. Neurol, vol.15, pp.14-20, 2008.

W. Poewe, F. Gerstenbrand, G. Ransmayr, and S. Plörer, Premorbid personality of Parkinson patients, J. Neural Transm. Suppl, vol.19, pp.215-224, 1983.

M. Poletti, G. Perugi, C. Logi, A. Romano, P. Del-dotto et al., Dopamine agonists and delusional jealousy in Parkinson's disease: A cross-sectional prevalence study, Mov. Disord, vol.27, pp.1679-1682, 2012.

P. Pollak, A. L. Benabid, C. Gross, D. M. Gao, A. Laurent et al., , 1993.

, Rev. Neurol, vol.149, pp.175-176

M. H. Polymeropoulos, Autosomal dominant Parkinson's disease and alpha-synuclein, Ann. Neurol, vol.44, pp.63-64, 1998.

M. H. Polymeropoulos, J. J. Higgins, L. I. Golbe, W. G. Johnson, S. E. Ide et al., Mapping of a gene for Parkinson's disease to chromosome 4q21-q23, Science, vol.274, pp.1197-1199, 1996.

M. M. Ponsen, D. Stoffers, J. Booij, B. L. Van-eck-smit, E. C. Wolters et al., Idiopathic hyposmia as a preclinical sign of Parkinson's disease, Ann. Neurol, vol.56, pp.173-181, 2004.

R. Porcari, C. Proukakis, C. A. Waudby, B. Bolognesi, P. P. Mangione et al., The H50Q mutation induces a 10-fold decrease in the solubility of ?-synuclein, J. Biol. Chem, vol.290, pp.2395-2404, 2015.

P. E. Porporato, N. Filigheddu, S. Reano, M. Ferrara, E. Angelino et al., Acylated and unacylated ghrelin impair skeletal muscle atrophy in mice, J. Clin. Invest, vol.123, pp.611-622, 2013.

R. B. Postuma, D. Berg, M. Stern, W. Poewe, C. W. Olanow et al., MDS clinical diagnostic criteria for Parkinson's disease, Mov. Disord, vol.30, pp.1591-1601, 2015.

K. Prasad, E. Tarasewicz, J. Mathew, P. A. Strickland, B. Buckley et al., Toxicokinetics and toxicodynamics of paraquat accumulation in mouse brain, Exp. Neurol, vol.215, pp.358-367, 2009.

R. D. Prediger, L. C. Batista, R. Medeiros, P. Pandolfo, J. C. Florio et al., The risk is in the air: Intranasal administration of MPTP to rats reproducing clinical features of Parkinson's disease, Exp. Neurol, vol.202, pp.391-403, 2006.

R. D. Prediger, D. Rial, R. Medeiros, C. P. Figueiredo, R. L. Doty et al., Risk is in the air: an intranasal MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) rat model of Parkinson's disease, Ann. N. Y. Acad. Sci, vol.1170, pp.629-636, 2009.

T. Pringsheim, N. Jette, A. Frolkis, and T. D. Steeves, The prevalence of Parkinson's disease: a systematic review and meta-analysis, Mov. Disord. Off. J. Mov. Disord. Soc, vol.29, pp.1583-1590, 2014.

C. C. Probst and T. Van-eimeren, The Functional Anatomy of Impulse Control Disorders, Curr. Neurol. Neurosci. Rep, vol.13, 2013.

J. Pruszak, L. Just, O. Isacson, and G. Nikkhah, Isolation and culture of ventral mesencephalic precursor cells and dopaminergic neurons from rodent brains, Curr. Protoc. Stem Cell Biol. Chapter, 2009.

S. Przedborski, M. Levivier, H. Jiang, M. Ferreira, V. Jackson-lewis et al., Dose-dependent lesions of the dopaminergic nigrostriatal pathway induced by intrastriatal injection of 6-hydroxydopamine, Neuroscience, vol.67, pp.631-647, 1995.

M. Quaak, C. P. Van-schayck, A. M. Knaapen, and F. J. Van-schooten, Genetic variation as a predictor of smoking cessation success. A promising preventive and intervention tool for chronic respiratory diseases?, Eur. Respir. J, vol.33, pp.468-480, 2009.

K. Radad, G. Gille, and W. Rausch, Dopaminergic neurons are preferentially sensitive to longterm rotenone toxicity in primary cell culture, Toxicol. In Vitro, vol.22, pp.68-74, 2008.

K. Radad, R. Moldzio, M. Taha, and W. Rausch, Thymoquinone protects dopaminergic neurons against MPP + and rotenone, Phytother. Res, vol.23, pp.696-700, 2009.

D. Rail, C. Scholtz, and M. Swash, Post-encephalitic Parkinsonism: current experience, J. Neurol. Neurosurg. Psychiatry, vol.44, pp.670-676, 1981.

P. R. Rajendran, R. E. Thompson, and S. G. Reich, The use of alternative therapies by patients with Parkinson's disease, Neurology, vol.57, pp.790-794, 2001.

C. Ramaker, J. Marinus, A. M. Stiggelbout, and B. J. Van-hilten, Systematic evaluation of rating scales for impairment and disability in Parkinson's disease, Mov. Disord. Off. J. Mov. Disord. Soc, vol.17, pp.867-876, 2002.

D. Ramonet, J. P. Daher, B. M. Lin, K. Stafa, J. Kim et al., Dopaminergic neuronal loss, reduced neurite complexity and autophagic abnormalities in transgenic mice expressing G2019S mutant LRRK2, PloS One, vol.6, p.18568, 2011.

S. Reano, A. Graziani, and N. Filigheddu, Acylated and unacylated ghrelin administration to blunt muscle wasting, Curr. Opin. Clin. Nutr. Metab. Care, vol.17, pp.236-240, 2014.

A. Rediger, C. L. Piechowski, K. Habegger, A. Grüters, H. Krude et al., MC4R Dimerization in the Paraventricular Nucleus and GHSR/MC3R Heterodimerization in the Arcuate Nucleus: Is There Relevance for Body Weight Regulation, Neuroendocrinology, vol.95, pp.277-288, 2012.

N. L. Rey, S. George, and P. Brundin, Review: Spreading the word: precise animal models and validated methods are vital when evaluating prion-like behaviour of alpha-synuclein, 2016.

, Neuropathol. Appl. Neurobiol, vol.42, pp.51-76

J. R. Richardson, Y. Quan, T. B. Sherer, J. T. Greenamyre, and G. W. Miller, Paraquat neurotoxicity is distinct from that of MPTP and rotenone, Toxicol. Sci. Off. J. Soc. Toxicol, vol.88, pp.193-201, 2005.

P. Riederer and S. Wuketich, Time course of nigrostriatal degeneration in parkinson's disease. A detailed study of influential factors in human brain amine analysis, J. Neural Transm, vol.38, pp.277-301, 1976.

A. E. Rigamonti, A. I. Pincelli, B. Corrà, R. Viarengo, S. M. Bonomo et al., Plasma ghrelin concentrations in elderly subjects: comparison with anorexic and obese patients, J. Endocrinol, vol.175, pp.1-5, 2002.

A. Rodríguez, J. Gómez-ambrosi, V. Catalán, M. J. Gil, S. Becerril et al., Acylated and desacyl ghrelin stimulate lipid accumulation in human visceral adipocytes, Int. J. Obes, vol.33, pp.541-552, 2005.

G. Rosati, E. Granieri, L. Pinna, I. Aiello, R. Tola et al., The risk of Parkinson disease in Mediterranean people, Neurology, vol.30, pp.250-255, 1980.

C. A. Ross and M. A. Poirier, Opinion: What is the role of protein aggregation in neurodegeneration?, Nat. Rev. Mol. Cell Biol, vol.6, pp.891-898, 2005.

G. W. Ross and H. Petrovitch, Current evidence for neuroprotective effects of nicotine and caffeine against Parkinson's disease, Drugs Aging, vol.18, pp.797-806, 2001.

G. W. Ross, H. Petrovitch, R. D. Abbott, C. M. Tanner, J. Popper et al., Association of olfactory dysfunction with risk for future Parkinson's disease, Ann. Neurol, vol.63, pp.167-173, 2008.

Z. L. Rossetti, A. Sotgiu, D. E. Sharp, M. Hadjiconstantinou, and N. H. Neff, 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and free radicals in vitro, Biochem. Pharmacol, vol.37, pp.4573-4574, 1988.

C. Rossi, D. Frosini, D. Volterrani, P. De-feo, E. Unti et al., Differences in nigro-striatal impairment in clinical variants of early Parkinson's disease: evidence from a FP-CIT SPECT study, Eur. J. Neurol, vol.17, pp.626-630, 2010.

M. W. Rousseaux, P. C. Marcogliese, D. Qu, S. J. Hewitt, S. Seang et al., Progressive dopaminergic cell loss with unilateral-to-bilateral progression in a genetic model of Parkinson disease, Proc. Natl. Acad. Sci. U. S. A, vol.109, pp.15918-15923, 2012.

J. M. Ruijter, C. Ramakers, W. M. Hoogaars, Y. Karlen, O. Bakker et al., Amplification efficiency: linking baseline and bias in the analysis of quantitative PCR data, Nucleic Acids Res, vol.37, 2009.

B. J. Ryan, L. L. Lourenço-venda, M. J. Crabtree, A. B. Hale, K. M. Channon et al., ?-Synuclein and mitochondrial bioenergetics regulate tetrahydrobiopterin levels in a human dopaminergic model of Parkinson disease. Free Radic, Biol. Med, vol.67, pp.58-68, 2014.

K. Sääksjärvi, P. Knekt, H. Rissanen, M. A. Laaksonen, A. Reunanen et al., Prospective study of coffee consumption and risk of Parkinson's disease, Eur. J. Clin. Nutr, vol.62, pp.908-915, 2008.

K. Saigoh, Y. L. Wang, J. G. Suh, T. Yamanishi, Y. Sakai et al., Intragenic deletion in the gene encoding ubiquitin carboxy-terminal hydrolase in gad mice, Nat. Genet, vol.23, pp.47-51, 1999.

A. Sakaguchi-nakashima, J. Y. Meir, Y. Jin, K. Matsumoto, and N. Hisamoto, LRK-1, a C. elegans PARK8-related kinase, regulates axonal-dendritic polarity of SV proteins, Curr. Biol. CB, vol.17, pp.592-598, 2007.

J. Sämann, J. Hegermann, E. Von-gromoff, S. Eimer, R. Baumeister et al., Caenorhabditits elegans LRK-1 and PINK-1 act antagonistically in stress response and neurite outgrowth, J. Biol. Chem, vol.284, pp.16482-16491, 2009.

J. R. Santos, J. A. Cunha, A. L. Dierschnabel, C. L. Campêlo, A. H. Leão et al., Cognitive, motor and tyrosine hydroxylase temporal impairment in a model of parkinsonism induced by reserpine, Behav. Brain Res, vol.253, pp.68-77, 2013.

W. Satake, Y. Nakabayashi, I. Mizuta, Y. Hirota, C. Ito et al., Genome-wide association study identifies common variants at four loci as genetic risk factors for Parkinson's disease, Nat. Genet, vol.41, pp.1303-1307, 2009.

M. Satou, Y. Nishi, J. Yoh, Y. Hattori, and H. Sugimoto, Identification and characterization of acylprotein thioesterase 1/lysophospholipase I as a ghrelin deacylation/lysophospholipid hydrolyzing enzyme in fetal bovine serum and conditioned medium, Endocrinology, vol.151, pp.4765-4775, 2010.

M. Sawada, K. Imamura, and T. Nagatsu, Role of cytokines in inflammatory process in Parkinson's disease, J. Neural Transm, pp.373-381, 2006.

G. V. Sawle, S. J. Wroe, A. J. Lees, D. J. Brooks, and R. S. Frackowiak, The identification of presymptomatic parkinsonism: clinical and [18F]dopa positron emission tomography studies in an Irish kindred, Ann. Neurol, vol.32, pp.609-617, 1992.

M. Schaeffer, F. Langlet, C. Lafont, F. Molino, D. J. Hodson et al., Rapid sensing of circulating ghrelin by hypothalamic appetite-modifying neurons, Proc. Natl. Acad. Sci. U. S. A, vol.110, pp.1512-1517, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00796900

T. Schallert, S. M. Fleming, J. L. Leasure, J. L. Tillerson, and S. T. Bland, CNS plasticity and assessment of forelimb sensorimotor outcome in unilateral rat models of stroke, cortical ablation, parkinsonism and spinal cord injury, neuropharmacology, vol.39, pp.777-787, 2000.

A. Schanze, U. Reulbach, M. Scheuchenzuber, M. Groschl, J. Kornhuber et al., Ghrelin and eating disturbances in psychiatric disorders, Neuropsychobiology, vol.57, pp.126-130, 2008.

A. H. Schapira, Recent developments in biomarkers in Parkinson disease, Curr. Opin. Neurol, vol.26, pp.395-400, 2013.

A. H. Schapira, Neurobiology and treatment of Parkinson's disease, Trends Pharmacol. Sci, vol.30, pp.41-47, 2009.

A. H. Schapira, M. Emre, P. Jenner, and W. Poewe, Levodopa in the treatment of Parkinson's disease, Eur. J. Neurol, vol.16, pp.982-989, 2009.

H. Schellekens, T. G. Dinan, and J. F. Cryan, Taking two to tango: a role for ghrelin receptor heterodimerization in stress and reward, Front. Neurosci, vol.7, 2013.

C. H. Schenck, S. R. Bundlie, and M. W. Mahowald, Delayed emergence of a parkinsonian disorder in 38% of 29 older men initially diagnosed with idiopathic rapid eye movement sleep behavior disorder, Neurology, vol.46, pp.388-393, 1996.

M. G. Schlossmacher, M. P. Frosch, W. P. Gai, M. Medina, N. Sharma et al., Parkin localizes to the Lewy bodies of Parkinson disease and dementia with Lewy bodies, Am. J. Pathol, vol.160, issue.10, pp.61113-61116, 2002.

B. S. Schoenberg, B. O. Osuntokun, A. O. Adeuja, O. Bademosi, V. Nottidge et al., Comparison of the prevalence of Parkinson's disease in black populations in the rural United States and in rural Nigeria: door-to-door community studies, Neurology, vol.38, pp.645-646, 1988.

B. Schüle, R. A. Pera, and J. W. Langston, Can cellular models revolutionize drug discovery in Parkinson's disease?, Biochim. Biophys. Acta BBA -Mol. Basis Dis, vol.1792, pp.1043-1051, 2009.

F. Schuler and J. E. Casida, The insecticide target in the PSST subunit of complex I, Pest Manag. Sci, vol.57, pp.932-940, 2001.

M. Sedelis, K. Hofele, G. W. Auburger, S. Morgan, J. P. Huston et al., MPTP susceptibility in the mouse: behavioral, neurochemical, and histological analysis of gender and strain differences, Behav. Genet, vol.30, pp.171-182, 2000.

P. Seibler, J. Graziotto, H. Jeong, F. Simunovic, C. Klein et al., Mitochondrial Parkin recruitment is impaired in neurons derived from mutant PINK1 iPS cells, J. Neurosci. Off. J. Soc. Neurosci, vol.31, pp.5970-5976, 2011.

I. Seim, S. L. Carter, A. C. Herington, and L. K. Chopin, Complex organisation and structure of the ghrelin antisense strand gene GHRLOS, a candidate non-coding RNA gene, BMC Mol. Biol, vol.9, p.95, 2008.

I. Seim, S. L. Carter, A. C. Herington, and L. K. Chopin, Complex organisation and structure of the ghrelin antisense strand gene GHRLOS, a candidate non-coding RNA gene, BMC Mol. Biol, vol.9, p.95, 2008.

I. Seim, C. Collet, A. C. Herington, and L. K. Chopin, Revised genomic structure of the human ghrelin gene and identification of novel exons, alternative splice variants and natural antisense transcripts, BMC Genomics, vol.8, p.298, 2007.

M. Selikhova, D. R. Williams, P. A. Kempster, J. L. Holton, T. Revesz et al., A clinicopathological study of subtypes in Parkinson's disease, Brain J. Neurol, vol.132, pp.2947-2957, 2009.

T. J. Shafer and W. D. Atchison, Transmitter, ion channel and receptor properties of pheochromocytoma (PC12) cells: a model for neurotoxicological studies, Neurotoxicology, vol.12, pp.473-492, 1991.

P. Shahim, K. Blennow, P. Johansson, J. Svensson, S. Lista et al., Cerebrospinal Fluid Stanniocalcin-1 as a Biomarker for Alzheimer's Disease and Other Neurodegenerative Disorders, Neuromolecular Med, 2016.

S. Sharma, S. Singh, V. Sharma, V. P. Singh, and R. Deshmukh, Neurobiology of l-DOPA induced dyskinesia and the novel therapeutic strategies, Biomed. Pharmacother. Bioméd. Pharmacothérapie, vol.70, pp.283-293, 2015.

T. B. Sherer, R. Betarbet, C. M. Testa, B. B. Seo, J. R. Richardson et al., Mechanism of toxicity in rotenone models of Parkinson's disease, J. Neurosci. Off. J. Soc. Neurosci, vol.23, pp.10756-10764, 2003.

T. Shimada, H. Furuta, A. Doi, H. Ariyasu, H. Kawashima et al., Des-acyl ghrelin protects microvascular endothelial cells from oxidative stress-induced apoptosis through sirtuin 1 signaling pathway, Metabolism, vol.63, pp.469-474, 2014.

K. Shimizu, K. Ohtaki, K. Matsubara, K. Aoyama, T. Uezono et al., Carrier-mediated processes in blood--brain barrier penetration and neural uptake of paraquat, Brain Res, vol.906, pp.135-142, 2001.

A. Shin, M. Camilleri, I. Busciglio, D. Burton, S. A. Smith et al., The ghrelin agonist RM-131 accelerates gastric emptying of solids and reduces symptoms in patients with type 1 diabetes mellitus, Clin. Gastroenterol. Hepatol. Off. Clin. Pract. J. Am. Gastroenterol. Assoc, vol.11, pp.1453-1459, 2013.

S. Shojaee, F. Sina, S. S. Banihosseini, M. H. Kazemi, R. Kalhor et al., Genome-wide linkage analysis of a Parkinsonian-pyramidal syndrome pedigree by 500 K SNP arrays, Am. J. Hum. Genet, vol.82, pp.1375-1384, 2008.

J. Simón-sánchez, C. Schulte, J. M. Bras, M. Sharma, J. R. Gibbs et al., Genome-wide association study reveals genetic risk underlying Parkinson's disease, Nat. Genet, vol.41, pp.1308-1312, 2009.

A. B. Singleton, M. Farrer, J. Johnson, A. Singleton, S. Hague et al., Synuclein locus triplication causes Parkinson's disease, vol.302, p.841, 2003.

F. Sixel-döring, J. Zimmermann, A. Wegener, B. Mollenhauer, and C. Trenkwalder, The Evolution of REM Sleep Behavior Disorder in Early Parkinson Disease, 2016.

W. W. Smith, R. L. Margolis, X. Li, J. C. Troncoso, M. K. Lee et al., Alpha-synuclein phosphorylation enhances eosinophilic cytoplasmic inclusion formation in SH-SY5Y cells, J. Neurosci. Off. J. Soc. Neurosci, vol.25, pp.5544-5552, 2005.

S. S. Sommer and W. A. Rocca, Prion analogues and twin studies in Parkinson's disease, Neurology, vol.46, pp.273-275, 1996.

R. Soto-otero, E. Méndez-alvarez, A. Hermida-ameijeiras, A. M. Muñoz-patiño, and J. L. Labandeira-garcia, Autoxidation and neurotoxicity of 6-hydroxydopamine in the presence of some antioxidants: potential implication in relation to the pathogenesis of Parkinson's disease, J. Neurochem, vol.74, pp.1605-1612, 2000.

B. Spencer, R. Potkar, M. Trejo, E. Rockenstein, C. Patrick et al., Beclin 1 gene transfer activates autophagy and ameliorates the neurodegenerative pathology in alpha-synuclein models of Parkinson's and Lewy body diseases, J. Neurosci. Off. J. Soc. Neurosci, vol.29, pp.13578-13588, 2009.

B. A. Spengler, J. L. Biedler, and R. A. Ross, A corrected karyotype for the SH-SY5Y human neuroblastoma cell line, Cancer Genet. Cytogenet, vol.138, pp.177-178, 2002.

M. G. Spillantini, R. A. Crowther, R. Jakes, N. J. Cairns, P. L. Lantos et al., Filamentous alpha-synuclein inclusions link multiple system atrophy with Parkinson's disease and dementia with Lewy bodies, Neurosci. Lett, vol.251, pp.205-208, 1998.

M. G. Spillantini, R. A. Crowther, R. Jakes, M. Hasegawa, and M. Goedert, ?-Synuclein in filamentous inclusions of Lewy bodies from Parkinson's disease and dementia with Lewy bodies, Proc. Natl. Acad. Sci, vol.95, pp.6469-6473, 1998.

M. G. Spillantini and M. Goedert, The alpha-synucleinopathies: Parkinson's disease, dementia with Lewy bodies, and multiple system atrophy, Ann. N. Y. Acad. Sci, vol.920, pp.16-27, 2000.

W. Springer, T. Hoppe, E. Schmidt, and R. Baumeister, A Caenorhabditis elegans Parkin mutant with altered solubility couples alpha-synuclein aggregation to proteotoxic stress, Hum. Mol. Genet, vol.14, pp.3407-3423, 2005.

H. Stachowiak, O. Wien, M. Stamelou, and K. P. Bhatia, Atypical parkinsonism: diagnosis and treatment, Neurol. Clin, vol.33, pp.39-56, 1973.

S. E. Starkstein, R. G. Robinson, R. Leiguarda, and T. J. Preziosi, Anxiety and depression in Parkinson's disease, Behav. Neurol, vol.6, pp.151-154, 1993.

K. C. Stein and H. L. True, Prion strains and amyloid polymorphism influence phenotypic variation, PLoS Pathog, vol.10, p.1004328, 2014.

M. B. Stern and A. Siderowf, Parkinson's at risk syndrome: Can Parkinson's disease be predicted?, Mov. Disord, vol.25, pp.89-93, 2010.

F. J. Steyn, V. Tolle, C. Chen, and J. Epelbaum, Neuroendocrine Regulation of Growth Hormone Secretion, Compr. Physiol, vol.6, pp.687-735, 2016.

M. G. Stokholm, E. H. Danielsen, S. J. Hamilton-dutoit, and P. Borghammer, Pathological ?synuclein in gastrointestinal tissues from prodromal Parkinson disease patients, Ann. Neurol, vol.79, pp.940-949, 2016.

G. Stores, Clinical diagnosis and misdiagnosis of sleep disorders, J. Neurol. Neurosurg. Psychiatry, vol.78, pp.1293-1297, 2007.

K. M. Strauss, L. M. Martins, H. Plun-favreau, F. P. Marx, S. Kautzmann et al., Loss of function mutations in the gene encoding Omi/HtrA2 in Parkinson's disease, Hum. Mol. Genet, vol.14, pp.2099-2111, 2005.

L. Studer, V. Tabar, and R. D. Mckay, Transplantation of expanded mesencephalic precursors leads to recovery in parkinsonian rats, Nat. Neurosci, vol.1, pp.290-295, 1998.

S. R. Subramaniam, L. Vergnes, N. R. Franich, K. Reue, and M. Chesselet, Region specific mitochondrial impairment in mice with widespread overexpression of alpha-synuclein, 2014.

, Neurobiol. Dis, vol.70, pp.204-213

Y. Sui, K. M. Bullock, M. A. Erickson, J. Zhang, and W. A. Banks, Alpha synuclein is transported into and out of the brain by the blood-brain barrier, Peptides, vol.62, pp.197-202, 2014.

P. Svenningsson, C. Rosenblad, K. Af-edholm-arvidsson, K. Wictorin, C. Keywood et al., Eltoprazine counteracts l-DOPA-induced dyskinesias in Parkinson's disease: a dose-finding study, Brain J. Neurol, vol.138, pp.963-973, 2015.

R. Taipa, C. Pereira, I. Reis, I. Alonso, A. Bastos-lima et al., DJ-1 linked parkinsonism (PARK7) is associated with Lewy body pathology, Brain J. Neurol, vol.139, pp.1680-1687, 2016.

K. Takagi, R. Legrand, A. Asakawa, H. Amitani, M. François et al., Anti-ghrelin immunoglobulins modulate ghrelin stability and its orexigenic effect in obese mice and humans, Nat. Commun, vol.4, p.2685, 2013.

H. Takeda, S. Muto, K. Nakagawa, S. Ohnishi, and M. Asaka, Rikkunshito and ghrelin secretion, Curr. Pharm. Des, vol.18, pp.4827-4838, 2012.

L. Tönges, T. Frank, L. Tatenhorst, K. A. Saal, J. C. Koch et al., Inhibition of rho kinase enhances survival of dopaminergic neurons and attenuates axonal loss in a mouse model of Parkinson's disease, Brain J. Neurol, vol.135, pp.3355-3370, 2012.

H. T. Tran, C. H. Chung, .. Iba, M. Zhang, B. Trojanowski et al., ?synuclein immunotherapy blocks uptake and templated propagation of misfolded ?synuclein and neurodegeneration, Cell Rep, vol.7, pp.2054-2065, 2014.

K. N. Tretiakoff, Contribution à l'étude de l'anatomie pathologique du locus niger de Soemmering avec quelques deductions à la pathologie des troubles musculaires et de la maladie de Parkinson, 1919.

P. A. Trimmer, M. K. Borland, P. M. Keeney, J. P. Bennett, and W. D. Parker, Parkinson's disease transgenic mitochondrial cybrids generate Lewy inclusion bodies, J. Neurochem, vol.88, pp.800-812, 2004.

J. Q. Trojanowski and V. M. Lee, Fatal attractions" of proteins. A comprehensive hypothetical mechanism underlying Alzheimer's disease and other neurodegenerative disorders, Ann. N. Y. Acad. Sci, vol.924, pp.62-67, 2000.

E. Tsika, M. Kannan, C. S. Foo, .. Dikeman, D. Glauser et al., Conditional expression of Parkinson's disease-related R1441C LRRK2 in midbrain dopaminergic neurons of mice causes nuclear abnormalities without neurodegeneration, Neurobiol. Dis, vol.71, pp.345-358, 2014.

M. D. Tuttle, G. Comellas, A. J. Nieuwkoop, D. J. Covell, D. A. Berthold et al., Solid-state NMR structure of a pathogenic fibril of fulllength human ?-synuclein, Nat. Struct. Mol. Biol, vol.23, pp.409-415, 2016.

T. Tyson, J. A. Steiner, and P. Brundin, Sorting out release, uptake and processing of alphasynuclein during prion-like spread of pathology, J. Neurochem, 2015.

C. Tzoulis, G. T. Tran, T. Schwarzlmüller, K. Specht, K. Haugarvoll et al., Severe nigrostriatal degeneration without clinical parkinsonism in patients with polymerase gamma mutations, Brain J. Neurol, vol.136, pp.2393-2404, 2013.

M. M. Unger, J. C. Möller, K. Mankel, K. M. Eggert, K. Bohne et al., Postprandial ghrelin response is reduced in patients with Parkinson's disease and idiopathic REM sleep behaviour disorder: a peripheral biomarker for early Parkinson's disease?, J. Neurol, vol.258, pp.982-990, 2011.

U. Ungerstedt, 6-Hydroxy-dopamine induced degeneration of central monoamine neurons, Eur. J. Pharmacol, vol.5, pp.107-110, 1968.

E. M. Valente, P. M. Abou-sleiman, V. Caputo, M. M. Muqit, K. Harvey et al., Hereditary early-onset Parkinson's disease caused by mutations in PINK1, Science, vol.304, pp.1158-1160, 2004.

E. Valera and E. Masliah, Immunotherapy for neurodegenerative diseases: focus on ?synucleinopathies, Pharmacol. Ther, vol.138, pp.311-322, 2013.

C. M. Van-duijn, M. C. Dekker, V. Bonifati, R. J. Galjaard, J. J. Houwing-duistermaat et al., PARK7, a Novel Locus for Autosomal Recessive Early-Onset Parkinsonism, on Chromosome 1p36, vol.69, pp.629-634, 2001.

S. Venneti, J. L. Robinson, S. Roy, M. T. White, J. Baccon et al., Simulated brain biopsy for diagnosing neurodegeneration using autopsy-confirmed cases, Acta Neuropathol. (Berl.), vol.122, pp.737-745, 2011.

A. Verstraeten, J. Theuns, and C. Van-broeckhoven, Progress in unraveling the genetic etiology of Parkinson disease in a genomic era, Trends Genet. TIG, vol.31, pp.140-149, 2015.

C. Vilariño-güell, C. Wider, O. A. Ross, J. C. Dachsel, J. M. Kachergus et al., VPS35 mutations in Parkinson disease, Am. J. Hum. Genet, vol.89, pp.162-167, 2011.

L. A. Volpicelli-daley, K. L. Gamble, C. E. Schultheiss, D. M. Riddle, A. B. West et al., Formation of ?-synuclein Lewy neurite-like aggregates in axons impedes the transport of distinct endosomes, Mol. Biol. Cell, vol.25, pp.4010-4023, 2014.

L. A. Volpicelli-daley, K. C. Luk, T. P. Patel, S. A. Tanik, D. M. Riddle et al., Exogenous ?-synuclein fibrils induce Lewy body pathology leading to synaptic dysfunction and neuron death, Neuron, vol.72, pp.57-71, 2011.

S. Von-campenhausen, B. Bornschein, R. Wick, K. Bötzel, C. Sampaio et al., Prevalence and incidence of Parkinson's disease in Europe, Eur. Neuropsychopharmacol. J. Eur. Coll. Neuropsychopharmacol, vol.15, pp.473-490, 2005.

G. Walkinshaw and C. M. Waters, Neurotoxin-induced cell death in neuronal PC12 cells is mediated by induction of apoptosis, Neuroscience, vol.63, pp.975-987, 1994.

K. Walsh and G. Bennett, Parkinson's disease and anxiety, Postgrad. Med. J, vol.77, pp.89-93, 2001.

L. Wang, S. M. Fleming, M. Chesselet, and Y. Taché, Abnormal colonic motility in mice overexpressing human wild-type ?-synuclein, NeuroReport, vol.19, pp.873-876, 2008.

L. Wang, S. Mogami, H. Karasawa, C. Yamada, S. Yakabi et al., Preventive effect of rikkunshito on gastric motor function inhibited by l-dopa in rats, Peptides, vol.55, pp.136-144, 2014.

L. Wang, N. P. Murphy, A. Stengel, M. Goebel-stengel, D. H. Pierre et al., Ghrelin prevents levodopa-induced inhibition of gastric emptying and increases circulating levodopa in fasted rats: Ghrelin prevents l-dopa-delayed gastric emptying, Neurogastroenterol. Motil, vol.24, pp.235-245, 2012.

X. Wang and E. K. Michaelis, Selective Neuronal Vulnerability to Oxidative Stress in the Brain, Front. Aging Neurosci, vol.2, 2010.

C. D. Ward, R. C. Duvoisin, S. E. Ince, J. D. Nutt, R. Eldridge et al., Parkinson's disease in 65 pairs of twins and in a set of quadruplets, Neurology, vol.33, pp.815-815, 1983.

M. Watabe-uchida, L. Zhu, S. K. Ogawa, A. Vamanrao, and N. Uchida, Whole-brain mapping of direct inputs to midbrain dopamine neurons, Neuron, vol.74, pp.858-873, 2012.

J. C. Watts and S. B. Prusiner, Mouse models for studying the formation and propagation of prions, J. Biol. Chem, vol.289, 2014.

J. L. Webb, B. Ravikumar, J. Atkins, J. N. Skepper, and D. C. Rubinsztein, Alpha-Synuclein is degraded by both autophagy and the proteasome, J. Biol. Chem, vol.278, pp.25009-25013, 2003.

M. Wellman and A. Abizaid, Growth Hormone Secretagogue Receptor Dimers: A New Pharmacological Target. eNeuro 2, 2015.

C. H. Williams-gray, T. Foltynie, C. E. Brayne, T. W. Robbins, and R. A. Barker, Evolution of cognitive dysfunction in an incident Parkinson's disease cohort, Brain J. Neurol, vol.130, pp.1787-1798, 2007.

P. Willner, The validity of animal models of depression, Psychopharmacology (Berl.), vol.83, pp.1-16, 1984.

J. M. Wo, N. Ejskjaer, P. M. Hellström, R. A. Malik, J. C. Pezzullo et al., Randomised clinical trial: ghrelin agonist TZP-101 relieves gastroparesis associated with severe nausea and vomiting--randomised clinical study subset data, Aliment. Pharmacol. Ther, vol.33, pp.679-688, 2011.

D. Woolley, D. Gietzen, S. Gee, J. Magdalou, and B. Hammock, Does paraquat (PD) mimic MPP+ toxicity?, Proc. West. Pharmacol. Soc, pp.191-193, 1989.

A. M. Wren, L. J. Seal, M. A. Cohen, A. E. Brynes, G. S. Frost et al., Ghrelin enhances appetite and increases food intake in humans, J. Clin. Endocrinol. Metab, vol.86, p.5992, 2001.

X. Wu, M. L. Block, W. Zhang, L. Qin, B. Wilson et al., The role of microglia in paraquat-induced dopaminergic neurotoxicity, Antioxid. Redox Signal, vol.7, pp.654-661, 2005.

N. Xiong, J. Xiong, M. Jia, L. Liu, X. Zhang et al., The role of autophagy in Parkinson's disease: rotenone-based modeling, Behav. Brain Funct. BBF, vol.9, p.13, 2013.

L. Xu and J. Pu, Alpha-Synuclein in Parkinson's Disease: From Pathogenetic Dysfunction to Potential Clinical Application. Park. Dis, p.1720621, 2016.

D. Yang, Z. Liu, H. Zhang, and Q. Luo, Ghrelin protects human pulmonary artery endothelial cells against hypoxia-induced injury via PI3-kinase/Akt, Peptides, vol.42, pp.112-117, 2013.

Q. Yang, H. She, M. Gearing, E. Colla, M. Lee et al., Regulation of neuronal survival factor MEF2D by chaperone-mediated autophagy, Science, vol.323, pp.124-127, 2009.

W. L. Yang and A. Y. Sun, Paraquat-induced cell death in PC12 cells, Neurochem. Res, vol.23, pp.1387-1394, 1998.

J. Yu, H. Xu, X. Shen, and H. Jiang, Ghrelin protects MES23.5 cells against rotenone via inhibiting mitochondrial dysfunction and apoptosis, Neuropeptides, vol.56, pp.69-74, 2016.

J. Yu, H. Xu, X. Shen, and H. Jiang, Ghrelin protects MES23.5 cells against rotenone via inhibiting mitochondrial dysfunction and apoptosis, Neuropeptides, 2015.

J. J. Zarranz, J. Alegre, J. C. Gómez-esteban, E. Lezcano, R. Ros et al., The new mutation, E46K, of alpha-synuclein causes Parkinson and Lewy body dementia, Ann. Neurol, vol.55, pp.164-173, 2004.

W. J. Zetusky, J. Jankovic, and F. J. Pirozzolo, The heterogeneity of Parkinson's disease: clinical and prognostic implications, Neurology, vol.35, pp.522-526, 1985.

L. Zhang, Y. Dong, X. Xu, and Z. Xu, The role of autophagy in Parkinson's disease, Neural Regen. Res, vol.7, pp.141-145, 2012.

W. Zhang, B. Chai, J. Li, H. Wang, and M. W. Mulholland, Effect of Des-acyl Ghrelin on Adiposity and Glucose Metabolism, Endocrinology, vol.149, pp.4710-4716, 2008.

X. Zhang, M. Yin, and M. Zhang, Cell-based assays for Parkinson's disease using differentiated human LUHMES cells, Acta Pharmacol. Sin, vol.35, pp.945-956, 2014.

X. Zhu, Y. Cao, K. Voodg, and D. F. Steiner, On the Processing of Proghrelin to Ghrelin, J. Biol. Chem, vol.281, pp.38867-38870, 2006.

J. M. Zigman, S. G. Bouret, and Z. B. Andrews, Obesity Impairs the Action of the Neuroendocrine Ghrelin System, Trends Endocrinol. Metab. TEM, vol.27, pp.54-63, 2016.

J. M. Zigman, J. E. Jones, C. E. Lee, C. B. Saper, and J. K. Elmquist, Expression of ghrelin receptor mRNA in the rat and the mouse brain, J. Comp. Neurol, vol.494, pp.528-548, 2006.

A. Zimprich, S. Biskup, P. Leitner, P. Lichtner, M. Farrer et al., Mutations in LRRK2 cause autosomal-dominant parkinsonism with pleomorphic pathology, Neuron, vol.44, pp.601-607, 2004.

P. Zizzari, R. Hassouna, D. Grouselle, J. Epelbaum, and V. Tolle, Physiological roles of preproghrelin-derived peptides in GH secretion and feeding, Peptides, vol.32, pp.2274-2282, 2011.

, Dribbling of saliva during the daytime

. .. , Loss or change in your ability to taste or smell

, Difficulty swallowing food or drink or problems with choking

. .. , Vomiting or feelings of sickness (nausea)

. .. , Constipation (less than 3 bowel movements a week) or having to strain to pass a stool (faeces)

, Bowel (fecal) incontinence

, Feeling that your bowel emptying is incomplete after having been to the toilet

, A sense of urgency to pass urine makes you rush to the toilet

. .. , Getting up regularly at night to pass urine

, Unexplained pains (not due to known conditions such as arthritis)

, Problems remembering things that have happened recently or forgetting to do things

, Loss of interest in what is happening around you or doing things

, Seeing or hearing things that you know or are told are not there

. .. , Difficulty concentrating or staying focussed

, Feeling anxious, frightened or panicky

, Feeling less interested in sex or more interested in sex

, Feeling light headed, dizzy or weak standing from sitting or lying, p.20

, Finding it difficult to stay awake during activities such as working, driving or eating, p.22

. .. , Intense, vivid dreams or frightening dreams

, Unpleasant sensations in your legs at night or while resting, and a feeling that you need to move, Talking or moving about in your sleep as if you are 'acting' out a

, Believing things are happening to you that other people say are not true, p.30

R. Abbott, R. A. Cox, M. Markus, H. Tomkins, and A. , Diet, body size and micronutrient status in Parkinson's disease, Eur. J. Clin. Nutr, vol.46, pp.879-884, 1992.

A. Abizaid, Z. Liu, Z. B. Andrews, M. Shanabrough, E. Borok et al., Ghrelin modulates the activity and synaptic input organization of midbrain dopamine neurons while promoting appetite, J. Clin. Invest, vol.116, pp.3229-3239, 2006.

R. A. Adan, J. J. Hillebrand, U. N. Danner, S. Cardona-cano, M. J. Kas et al., Neurobiology driving hyperactivity in activity-based anorexia, Curr. Top. Behav. Neurosci, vol.6, pp.229-250, 2011.

T. Akamizu and K. Kangawa, Therapeutic applications of ghrelin to cachexia utilizing its appetite-stimulating effect, Peptides, vol.32, pp.2295-2300, 2011.

R. L. Albin, A. B. Young, and J. B. Penney, The functional anatomy of basal ganglia disorders, Trends Neurosci, vol.12, pp.366-375, 1989.

H. Alzahrani and A. Venneri, Cognitive and neuroanatomical correlates of neuropsychiatric symptoms in Parkinson's disease: A systematic review, J. Neurol. Sci, vol.356, pp.32-44, 2015.

, Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition, American Psychiatric Association, 2013.

Z. B. Andrews, D. Erion, R. Beiler, Z. Liu, A. Abizaid et al., Ghrelin Promotes and Protects Nigrostriatal Dopamine Function via a UCP2-Dependent Mitochondrial Mechanism, J. Neurosci, vol.29, pp.14057-14065, 2009.

Z. B. Andrews, Z. Liu, N. Walllingford, D. M. Erion, E. Borok et al., UCP2 mediates ghrelin's action on NPY/AgRP neurons by lowering free radicals, Nature, vol.454, pp.846-851, 2008.

S. V. Angeloni, N. Glynn, G. Ambrosini, M. J. Garant, J. D. Higley et al., Characterization of the rhesus monkey ghrelin gene and factors influencing ghrelin gene expression and fasting plasma levels, Endocrinology, vol.145, pp.2197-2205, 2004.

G. Arabia, B. R. Grossardt, R. C. Colligan, J. H. Bower, D. M. Maraganore et al., Novelty seeking and introversion do not predict the longterm risk of Parkinson disease, Neurology, vol.75, pp.349-357, 2010.

E. Arai, M. Arai, T. Uchiyama, Y. Higuchi, K. Aoyagi et al., Subthalamic deep brain stimulation can improve gastric emptying in Parkinson's disease, Brain, vol.135, pp.1478-1485, 2012.

F. Assogna, L. Cravello, M. D. Orfei, N. Cellupica, C. Caltagirone et al., Alexithymia in Parkinson's disease: A systematic review of the literature, Parkinsonism Relat. Disord, vol.28, pp.1-11, 2016.

B. B. Averbeck, S. S. O'sullivan, and A. Djamshidian, Impulsive and compulsive behaviors in Parkinson's disease, Annu. Rev. Clin. Psychol, vol.10, pp.553-580, 2014.

U. F. Bailer, G. K. Frank, J. C. Price, C. C. Meltzer, C. Becker et al., Interaction between serotonin transporter and dopamine D2/D3 receptor radioligand measures is associated with harm avoidant symptoms in anorexia and bulimia nervosa, Psychiatry Res, vol.211, pp.160-168, 2013.

G. Baldanzi, Ghrelin and des-acyl ghrelin inhibit cell death in cardiomyocytes and endothelial cells through ERK1/2 and PI 3-kinase/AKT, J. Cell Biol, vol.159, pp.1029-1037, 2002.

A. Bali and A. S. Jaggi, An Integrative Review on Role and Mechanisms of Ghrelin in Stress, Anxiety and Depression, Curr. Drug Targets, vol.17, pp.495-507, 2016.

B. W. Balleine, Sensation, Incentive Learning, and the Motivational Control of Goal-Directed Action, 2011.

W. A. Banks, Extent and Direction of Ghrelin Transport Across the Blood-Brain Barrier Is Determined by Its Unique Primary Structure, J. Pharmacol. Exp. Ther, vol.302, pp.822-827, 2002.

A. L. Bartels, A. T. Willemsen, R. Kortekaas, B. M. De-jong, R. De-vries et al., Decreased blood-brain barrier Pglycoprotein function in the progression of Parkinson's disease, PSP and MSA, J. Neural Transm, vol.115, pp.1001-1009, 1996.

J. A. Bayliss and Z. B. Andrews, Ghrelin is neuroprotective in Parkinson's disease: molecular mechanisms of metabolic neuroprotection, Ther. Adv. Endocrinol. Metab, vol.4, pp.25-36, 2013.

J. A. Bayliss, M. Lemus, V. V. Santos, M. Deo, J. Elsworth et al., Acylated but not des-acyl ghrelin is neuroprotective in an MPTP mouse model of Parkinson's Disease, J. Neurochem, 2016.

J. A. Bayliss, M. B. Lemus, R. Stark, V. V. Santos, A. Thompson et al., Ghrelin-AMPK Signaling Mediates the Neuroprotective Effects of Calorie Restriction in Parkinson's Disease, J. Neurosci. Off. J. Soc. Neurosci, vol.36, pp.3049-3063, 2016.

K. T. Beier, E. E. Steinberg, K. E. Deloach, S. Xie, K. Miyamichi et al., Circuit Architecture of VTA Dopamine Neurons Revealed by Systematic Input-Output Mapping, Cell, vol.162, pp.622-634, 2015.
URL : https://hal.archives-ouvertes.fr/hal-02187382

A. W. Bergen, M. Yeager, R. A. Welch, K. Haque, J. K. Ganjei et al., Association of multiple DRD2 polymorphisms with anorexia nervosa, Neuropsychopharmacol. Off. Publ. Am. Coll. Neuropsychopharmacol, vol.30, pp.1703-1710, 2005.

H. Bernheimer, W. Birkmayer, O. Hornykiewicz, K. Jellinger, and F. Seitelberger, Brain dopamine and the syndromes of Parkinson and Huntington. Clinical, morphological and neurochemical correlations, J. Neurol. Sci, vol.20, pp.415-455, 1973.

K. C. Berridge, C. Ho, J. M. Richard, and A. G. Difeliceantonio, The tempted brain eats: pleasure and desire circuits in obesity and eating disorders, Brain Res, vol.1350, pp.43-64, 2010.

K. C. Berridge and M. L. Kringelbach, Pleasure systems in the brain, Neuron, vol.86, pp.646-664, 2015.

K. C. Berridge and T. E. Robinson, What is the role of dopamine in reward: hedonic impact, reward learning, or incentive salience? Brain Res, Brain Res. Rev, vol.28, pp.309-369, 1998.

K. C. Berridge, T. E. Robinson, and J. W. Aldridge, Dissecting components of reward: "liking", "wanting", and learning, Curr. Opin. Pharmacol, vol.9, pp.65-73, 2009.

K. C. Berridge, I. L. Venier, and T. E. Robinson, Taste reactivity analysis of 6-hydroxydopamine-induced aphagia: implications for arousal and anhedonia hypotheses of dopamine function, Behav. Neurosci, vol.103, pp.36-45, 1989.

A. Björklund and S. B. Dunnett, Dopamine neuron systems in the brain: an update, Trends Neurosci, vol.30, pp.194-202, 2007.

J. W. B?aszczyk, Motor deficiency in Parkinson's disease, Acta Neurobiol. Exp. (Warsz.), vol.58, pp.79-93, 1998.

I. D. Blum, Z. Patterson, R. Khazall, E. W. Lamont, M. W. Sleeman et al., Reduced anticipatory locomotor responses to scheduled meals in ghrelin receptor deficient mice, Neuroscience, vol.164, pp.351-359, 2009.

L. Booij, K. F. Casey, J. M. Antunes, M. Szyf, R. Joober et al., DNA methylation in individuals with anorexia nervosa and in matched normal-eater controls: A genome-wide study, Int. J. Eat. Disord, vol.48, pp.874-882, 2015.

V. Boraska, C. S. Franklin, J. A. Floyd, L. M. Thornton, L. M. Huckins et al.,

J. I. Hudson, T. Reichborn-kjennerud, G. P. Knudsen, P. Monteleone, A. S. Kaplan et al.,

, Mol. Psychiatry, vol.19, pp.1085-1094

R. Bourdy, M. Sánchez-catalán, J. Kaufling, J. J. Balcita-pedicino, M. Freund-mercier et al., Control of the Nigrostriatal Dopamine Neuron Activity and Motor Function by the Tail of the Ventral Tegmental Area, Neuropsychopharmacology, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01160036

J. P. Britt, F. Benaliouad, R. A. Mcdevitt, G. D. Stuber, R. A. Wise et al., Synaptic and behavioral profile of multiple glutamatergic inputs to the nucleus accumbens, Neuron, vol.76, pp.790-803, 2012.

J. P. Britt and A. Bonci, Optogenetic interrogations of the neural circuits underlying addiction, Curr. Opin. Neurobiol, vol.23, pp.539-545, 2013.

V. Brochard, B. Combadière, A. Prigent, Y. Laouar, A. Perrin et al., Infiltration of CD4+ lymphocytes into the brain contributes to neurodegeneration in a mouse model of Parkinson disease, J. Clin. Invest, vol.119, pp.182-192, 2009.
URL : https://hal.archives-ouvertes.fr/inserm-00393947

F. Broglio, L. Gianotti, S. Destefanis, S. Fassino, G. Abbate-daga et al., The endocrine response to acute ghrelin administration is blunted in patients with anorexia nervosa, a ghrelin hypersecretory state, Clin. Endocrinol. (Oxf.), vol.60, pp.592-599, 2004.

P. Calabresi, B. Picconi, A. Tozzi, V. Ghiglieri, and M. Di-filippo, Direct and indirect pathways of basal ganglia: a critical reappraisal, Nat. Neurosci, vol.17, 2014.

B. Callaghan and J. B. Furness, Novel and Conventional Receptors for Ghrelin, Desacyl-Ghrelin, and Pharmacologically Related Compounds, Pharmacol. Rev, vol.66, pp.984-1001, 2014.

M. B. Callesen, J. Scheel-krüger, M. L. Kringelbach, and A. Møller, A systematic review of impulse control disorders in Parkinson's disease, J. Park. Dis, vol.3, pp.105-138, 2013.

J. P. Camiña, M. Lodeiro, O. Ischenko, A. C. Martini, and F. F. Casanueva, Stimulation by ghrelin of p42/p44 mitogen-activated protein kinase through the GHS-R1a receptor: role of G-proteins and beta-arrestins, J. Cell. Physiol, vol.213, pp.187-200, 2007.

S. Cardona-cano, M. Merkestein, K. P. Skibicka, S. L. Dickson, and R. A. Adan, Role of ghrelin in the pathophysiology of eating disorders: implications for pharmacotherapy, CNS Drugs, vol.26, pp.281-296, 2012.

M. Carta, T. Carlsson, A. Muñoz, D. Kirik, and A. Björklund, Serotonin-dopamine interaction in the induction and maintenance of L-DOPA-induced dyskinesias, Prog. Brain Res, vol.172, pp.465-478, 2008.

M. Cazorla, F. D. De-carvalho, M. O. Chohan, M. Shegda, N. Chuhma et al., Dopamine D2 receptors regulate the anatomical and functional balance of basal ganglia circuitry, Neuron, vol.81, pp.153-164, 2014.
URL : https://hal.archives-ouvertes.fr/inserm-02438087

M. Chartier-harlin, J. Kachergus, C. Roumier, V. Mouroux, X. Douay et al., ?-synuclein locus duplication as a cause of familial Parkinson's disease, The Lancet, vol.364, pp.1167-1169, 2004.

K. B. Chow, J. Sun, K. Chu, W. Tai-cheung, C. H. Cheng et al., The truncated ghrelin receptor polypeptide (GHS-R1b) is localized in the endoplasmic reticulum where it forms heterodimers with ghrelin receptors (GHS-R1a) to attenuate their cell surface expression, Mol. Cell. Endocrinol, vol.348, pp.247-254, 2012.

J. Clarke, N. Ramoz, A. Fladung, and P. Gorwood, Higher reward value of starvation imagery in anorexia nervosa and association with the Val66Met BDNF polymorphism, Transl. Psychiatry, vol.6, p.829, 2016.

J. Y. Cohen, S. Haesler, L. Vong, B. B. Lowell, and N. Uchida, Neuron-type-specific signals for reward and punishment in the ventral tegmental area, Nature, vol.482, pp.85-88, 2012.

G. Collden, E. Balland, J. Parkash, E. Caron, F. Langlet et al., Neonatal overnutrition causes early alterations in the central response to peripheral ghrelin, 2015.

. Mol and . Metab, , vol.4, pp.15-24

J. J. Cone, J. E. Mccutcheon, and M. F. Roitman, Ghrelin acts as an interface between physiological state and phasic dopamine signaling, J. Neurosci. Off. J. Soc. Neurosci, vol.34, pp.4905-4913, 2014.

B. Connolly and S. H. Fox, Treatment of cognitive, psychiatric, and affective disorders associated with Parkinson's disease, Neurother. J. Am. Soc. Exp. Neurother, vol.11, pp.78-91, 2014.

J. Corcuff, E. Krim, F. Tison, A. Foubert-sanier, D. Guehl et al., Subthalamic nucleus stimulation in patients with Parkinson's disease does not increase serum ghrelin levels, Br. J. Nutr, vol.95, pp.1028-1029, 2006.

J. L. Costa, D. Naot, J. Lin, M. Watson, K. E. Callon et al., Ghrelin is an Osteoblast Mitogen and Increases Osteoclastic Bone Resorption In Vitro, Int. J. Pept, p.605193, 2011.

M. A. Cowley, R. G. Smith, S. Diano, M. Tschöp, N. Pronchuk et al., The distribution and mechanism of action of ghrelin in the CNS demonstrates a novel hypothalamic circuit regulating energy homeostasis, Neuron, vol.37, pp.649-661, 2003.

D. E. Cummings, J. Q. Purnell, R. S. Frayo, K. Schmidova, B. E. Wisse et al., A preprandial rise in plasma ghrelin levels suggests a role in meal initiation in humans, Diabetes, vol.50, pp.1714-1719, 2001.

C. Da-cunha, S. L. Boschen, A. Gómez-a, E. K. Ross, W. S. Gibson et al., Toward sophisticated basal ganglia neuromodulation: Review on basal ganglia deep brain stimulation, Neurosci. Biobehav. Rev, vol.58, pp.186-210, 2015.

A. Dahlström and K. Fuxe, Localization of monoamines in the lower brain stem, Cell. Mol. Life Sci, vol.20, pp.398-399, 1964.

Y. Date, M. Nakazato, N. Murakami, M. Kojima, K. Kangawa et al., Ghrelin Acts in the Central Nervous System to Stimulate Gastric Acid Secretion, Biochem. Biophys. Res. Commun, vol.280, pp.904-907, 2001.

C. Davis, Eating disorders and hyperactivity: a psychobiological perspective, Can. J. Psychiatry Rev. Can. Psychiatr, vol.42, pp.168-175, 1997.

C. Davis and S. Kaptein, Anorexia nervosa with excessive exercise: a phenotype with close links to obsessive-compulsive disorder, Psychiatry Res, vol.142, pp.209-217, 2006.

C. Davis and D. B. Woodside, Sensitivity to the rewarding effects of food and exercise in the eating disorders, Compr. Psychiatry, vol.43, pp.189-194, 2002.

J. J. Day, J. L. Jones, R. M. Wightman, and R. M. Carelli, Phasic nucleus accumbens dopamine release encodes effort-and delay-related costs, Biol. Psychiatry, vol.68, pp.306-309, 2010.

P. J. Delhanty, Ghrelin and unacylated ghrelin stimulate human osteoblast growth via mitogen-activated protein kinase (MAPK)/phosphoinositide 3-kinase (PI3K) pathways in the absence of GHS-R1a, J. Endocrinol, vol.188, pp.37-47, 2006.

P. J. Delhanty, S. J. Neggers, and A. J. Van-der-lely, Should we consider des-acyl ghrelin as a separate hormone and if so, what does it do?, Front. Horm. Res, vol.42, pp.163-174, 2014.

M. R. Delong, Primate models of movement disorders of basal ganglia origin, Trends Neurosci, vol.13, pp.281-285, 1990.

C. Delporte, Structure and Physiological Actions of Ghrelin. Scientifica, vol.2013, pp.1-25, 2013.

G. Di-chiara, Nucleus accumbens shell and core dopamine: differential role in behavior and addiction, Behav. Brain Res, vol.137, pp.75-114, 2002.

S. Diano, S. A. Farr, S. C. Benoit, E. C. Mcnay, I. Da-silva et al., Ghrelin controls hippocampal spine synapse density and memory performance, Nat. Neurosci, vol.9, pp.381-388, 2006.

S. L. Dickson, E. Egecioglu, S. Landgren, K. P. Skibicka, J. A. Engel et al., The role of the central ghrelin system in reward from food and chemical drugs, Mol. Cell. Endocrinol, vol.340, pp.80-87, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00706318

J. Dong, N. Song, J. Xie, and H. Jiang, Ghrelin Antagonized 1-Methyl-4-Phenylpyridinium (MPP+)-Induced Apoptosis in MES23.5 Cells, J. Mol. Neurosci, vol.37, pp.182-189, 2009.

D. Santos, V. V. Rodrigues, A. L. De-lima, T. C. De-barioglio, S. R. Raisman-vozari et al., Ghrelin as a neuroprotective and palliative agent in Alzheimer's and Parkinson's disease, Curr. Pharm. Des, vol.19, pp.6773-6790, 2013.

D. L. Drazen, T. P. Vahl, D. A. D'alessio, R. J. Seeley, and S. C. Woods, Effects of a fixed meal pattern on ghrelin secretion: evidence for a learned response independent of nutrient status, Endocrinology, vol.147, pp.23-30, 2006.

E. Egecioglu, E. Jerlhag, N. Salomé, K. P. Skibicka, D. Haage et al., Ghrelin increases intake of rewarding food in rodents, Addict. Biol, vol.15, pp.304-311, 2010.

N. Ejskjaer, G. Dimcevski, J. Wo, P. M. Hellström, L. C. Gormsen et al., Safety and efficacy of ghrelin agonist TZP-101 in relieving symptoms in patients with diabetic gastroparesis: a randomized, placebo-controlled study, Neurogastroenterol. Motil. Off. J. Eur. Gastrointest. Motil. Soc, vol.22, pp.1069-281, 2010.

N. Ejskjaer, J. M. Wo, T. Esfandyari, M. Jamal, G. Dimcevski et al., A phase 2a, randomized, double-blind 28-day study of TZP-102 a ghrelin receptor agonist for diabetic gastroparesis, Neurogastroenterol. Motil. Off. J. Eur. Gastrointest. Motil. Soc, vol.25, pp.140-150, 2013.

L. Erdur, B. Kallenbach-dermutz, V. Lehmann, F. Zimmermann-viehoff, W. Köpp et al., Somatic comorbidity in anorexia nervosa: First results of a 21-year follow-up study on female inpatients, Biopsychosoc. Med, vol.6, p.4, 2012.

A. H. Evans, A. D. Lawrence, J. Potts, L. Macgregor, R. Katzenschlager et al., Relationship between impulsive sensation seeking traits, smoking, alcohol and caffeine intake, and Parkinson's disease, J. Neurol. Neurosurg. Psychiatry, vol.77, pp.317-321, 2006.

J. G. Ferreira, F. Del-fava, R. H. Hasue, and S. J. Shammah-lagnado, Organization of ventral tegmental area projections to the ventral tegmental area-nigral complex in the rat, Neuroscience, vol.153, pp.196-213, 2008.

D. F. Fiorino and A. G. Phillips, Facilitation of sexual behavior and enhanced dopamine efflux in the nucleus accumbens of male rats after D-amphetamine-induced behavioral sensitization, J. Neurosci. Off. J. Soc. Neurosci, vol.19, pp.456-463, 1999.

D. F. Fiorino and A. G. Phillips, Facilitation of sexual behavior in male rats following damphetamine-induced behavioral sensitization, Psychopharmacology (Berl.), vol.142, pp.200-208, 1999.

U. Fiszer, M. Micha?owska, B. Baranowska, E. Woli?ska-witort, W. Jeske et al., Leptin and ghrelin concentrations and weight loss in Parkinson's disease, Acta Neurol. Scand, vol.121, pp.230-236, 2010.

A. Fladung, U. M. Schulze, F. Schöll, K. Bauer, and G. Grön, Role of the ventral striatum in developing anorexia nervosa, Transl. Psychiatry, vol.3, p.315, 2013.

L. Frago, Neuroprotective actions of ghrelin and growth hormone secretagogues, Front. Mol. Neurosci, vol.4, 2011.

M. François, S. Barde, N. Achamrah, J. Breton, J. Do-rego et al., The number of preproghrelin mRNA expressing cells is increased in mice with activity-based anorexia, Neuropeptides, vol.51, pp.17-23, 2015.

G. K. Frank, U. F. Bailer, S. E. Henry, W. Drevets, C. C. Meltzer et al., Increased dopamine D2/D3 receptor binding after recovery from anorexia nervosa measured by positron emission tomography and [11c]raclopride, Biol. Psychiatry, vol.58, pp.908-912, 2005.

G. K. Frank, J. R. Reynolds, M. E. Shott, L. Jappe, T. T. Yang et al., Anorexia nervosa and obesity are associated with opposite brain reward response, Neuropsychopharmacol. Off. Publ. Am. Coll. Neuropsychopharmacol, vol.37, pp.2031-2046, 2012.

J. B. Furness, B. Hunne, N. Matsuda, L. Yin, D. Russo et al., Investigation of the presence of ghrelin in the central nervous system of the rat and mouse, Neuroscience, vol.193, pp.1-9, 2011.

M. A. Gates, E. M. Torres, A. White, R. A. Fricker-gates, and S. B. Dunnett, Re-examining the ontogeny of substantia nigra dopamine neurons, Eur. J. Neurosci, vol.23, pp.1384-1390, 2006.

C. Gauna, P. J. Delhanty, L. J. Hofland, J. A. Janssen, F. Broglio et al., Ghrelin stimulates, whereas des-octanoyl ghrelin inhibits, glucose output by primary hepatocytes, J. Clin. Endocrinol. Metab, vol.90, pp.1055-1060, 2005.

N. Germain, B. Galusca, D. Grouselle, D. Frere, S. Billard et al., Ghrelin and obestatin circadian levels differentiate bingeing-purging from restrictive anorexia nervosa, J. Clin. Endocrinol. Metab, vol.95, pp.3057-3062, 2010.

N. Germain, B. Galusca, D. Grouselle, D. Frere, V. Tolle et al., Ghrelin/obestatin ratio in two populations with low bodyweight: constitutional thinness and anorexia nervosa, Psychoneuroendocrinology, vol.34, pp.413-419, 2009.

E. Gershon and W. W. Vale, CRF type 2 receptors mediate the metabolic effects of ghrelin in C2C12 cells, Obes. Silver Spring Md, vol.22, pp.380-389, 2014.

G. Gervasini, I. Gordillo, A. García-herráiz, I. Flores, M. Jiménez et al., Influence of dopamine polymorphisms on the risk for anorexia nervosa and associated psychopathological features, J. Clin. Psychopharmacol, vol.33, pp.551-555, 2013.

E. Ghigo, F. Broglio, E. Arvat, M. Maccario, M. Papotti et al., Ghrelin: more than a natural GH secretagogue and/or an orexigenic factor, Clin. Endocrinol. (Oxf.), vol.62, pp.1-17, 2005.

S. Ghods-sharifi and S. B. Floresco, Differential effects on effort discounting induced by inactivations of the nucleus accumbens core or shell, Behav. Neurosci, vol.124, pp.179-191, 2010.

S. Gnanapavan, B. Kola, S. A. Bustin, D. G. Morris, P. Mcgee et al., The tissue distribution of the mRNA of the ghrelin and subtypes of its receptor, GHS-R, in humans, J. Clin. Endocrinol. Metab, vol.87, pp.2988-2991, 2002.

J. L. Goldstein, T. -. Zhao, R. L. Li, D. P. Sherbet, G. Liang et al., Surviving starvation: essential role of the ghrelin-growth hormone axis, Cold Spring Harb. Symp. Quant. Biol, vol.76, pp.121-127, 2011.

P. Gorwood, S. Richard-devantoy, O. Sentissi, Y. Le-strat, and J. P. Olié, The number of past manic episodes is the best predictor of antidepressant-emergent manic switch in a cohort of bipolar depressed patients, Psychiatry Res, vol.240, pp.288-294, 2016.

R. Granata, F. Settanni, L. Biancone, L. Trovato, R. Nano et al., Acylated and unacylated ghrelin promote proliferation and inhibit apoptosis of pancreatic beta-cells and human islets: involvement of 3',5'-cyclic adenosine monophosphate/protein kinase A, extracellular signal-regulated kinase 1/2, and phosphatidyl inositol 3-Kinase/Akt signaling, Endocrinology, vol.148, pp.512-529, 2007.

M. T. Gray and J. M. Woulfe, Striatal blood-brain barrier permeability in Parkinson's disease, J. Cereb. Blood Flow Metab. Off. J. Int. Soc. Cereb. Blood Flow Metab, vol.35, pp.747-750, 2015.

X. Guan, H. Yu, O. C. Palyha, K. K. Mckee, S. D. Feighner et al., Distribution of mRNA encoding the growth hormone secretagogue receptor in brain and peripheral tissues, Mol. Brain Res, vol.48, pp.23-29, 1997.

E. Guatteo, M. L. Cucchiaroni, and N. B. Mercuri, Substantia nigra control of basal ganglia nuclei, J. Neural Transm. Suppl, pp.91-101, 2009.

J. A. Gutierrez, P. J. Solenberg, D. R. Perkins, J. A. Willency, M. D. Knierman et al., Ghrelin octanoylation mediated by an orphan lipid transferase, Proc. Natl. Acad. Sci, vol.105, pp.6320-6325, 2008.

S. N. Haber, The place of dopamine in the cortico-basal ganglia circuit, Neuroscience, vol.282, pp.248-257, 2014.

S. N. Haber and J. L. Fudge, The primate substantia nigra and VTA: integrative circuitry and function, Crit. Rev. Neurobiol, vol.11, pp.323-342, 1997.

S. N. Haber and B. Knutson, The reward circuit: linking primate anatomy and human imaging, Neuropsychopharmacol. Off. Publ. Am. Coll. Neuropsychopharmacol, vol.35, pp.4-26, 2010.

L. Heimer, D. S. Zahm, L. Churchill, P. W. Kalivas, and C. Wohltmann, Specificity in the projection patterns of accumbal core and shell in the rat, Neuroscience, vol.41, pp.89-125, 1991.

C. L. Heusner, T. S. Hnasko, M. S. Szczypka, Y. Liu, M. J. During et al., Viral restoration of dopamine to the nucleus accumbens is sufficient to induce a locomotor response to amphetamine, Brain Res, vol.980, pp.266-274, 2003.

B. Holst, A. Cygankiewicz, T. H. Jensen, M. Ankersen, and T. W. Schwartz, High Constitutive Signaling of the Ghrelin Receptor-Identification of a Potent Inverse Agonist, 2003.

, Mol. Endocrinol, vol.17, pp.2201-2210

M. Hotta, R. Ohwada, T. Akamizu, T. Shibasaki, K. Takano et al., Ghrelin increases hunger and food intake in patients with restricting-type anorexia nervosa: a pilot study, Endocr. J, vol.56, pp.1119-1128, 2009.

A. Howard, S. Feighner, D. Cully, J. Arena, P. Liberator et al., A receptor in pituitary and hypothalamus that functions in growth hormone release, vol.16, pp.974-977, 1996.

Z. Hu, M. Cooper, D. P. Crockett, and R. Zhou, Differentiation of the midbrain dopaminergic pathways during mouse development, J. Comp. Neurol, vol.476, pp.301-311, 2004.

S. Hwang, M. Moon, S. Kim, L. Hwang, K. J. Ahn et al., Neuroprotective effect of ghrelin is associated with decreased expression of prostate apoptosis response-4, Endocr. J, vol.56, pp.609-617, 2009.

S. Hyman, D. Chisholm, R. Kessler, V. Patel, H. Whiteford et al., Mental Disorders, Disease Control Priorities in Developing Countries, 2006.

S. Ikemoto, Dopamine reward circuitry: two projection systems from the ventral midbrain to the nucleus accumbens-olfactory tubercle complex, Brain Res. Rev, vol.56, pp.27-78, 2007.

S. Ikemoto, C. Yang, and A. Tan, Basal ganglia circuit loops, dopamine and motivation: A review and enquiry, Behav. Brain Res, vol.290, pp.17-31, 2015.

A. Ilango, J. Shumake, W. Wetzel, and F. W. Ohl, Contribution of emotional and motivational neurocircuitry to cue-signaled active avoidance learning, Front. Behav. Neurosci, vol.8, p.372, 2014.

E. Jerlhag, E. Egecioglu, S. L. Dickson, M. Andersson, L. Svensson et al., Ghrelin stimulates locomotor activity and accumbal dopamine-overflow via central cholinergic systems in mice: implications for its involvement in brain reward, Addict. Biol, vol.11, pp.45-54, 2006.

E. Jerlhag, E. Egecioglu, S. L. Dickson, A. Douhan, L. Svensson et al., Ghrelin administration into tegmental areas stimulates locomotor activity and increases extracellular concentration of dopamine in the nucleus accumbens, Addict. Biol, vol.12, pp.6-16, 2007.

E. Jerlhag, E. Egecioglu, S. L. Dickson, and J. A. Engel, Glutamatergic regulation of ghrelin-induced activation of the mesolimbic dopamine system, Addict. Biol, vol.16, pp.82-91, 2011.

H. Jiang, L. Betancourt, and R. G. Smith, Ghrelin Amplifies Dopamine Signaling by Cross Talk Involving Formation of Growth Hormone Secretagogue Receptor/Dopamine Receptor Subtype 1 Heterodimers, Mol. Endocrinol, vol.20, pp.1772-1785, 2006.

P. M. Johnson and P. J. Kenny, Dopamine D2 receptors in addiction-like reward dysfunction and compulsive eating in obese rats, Nat. Neurosci, vol.13, pp.635-641, 2010.

J. Kamegai, I. Wakabayashi, R. D. Kineman, and L. A. Frohman, Growth hormonereleasing hormone receptor (GHRH-R) and growth hormone secretagogue receptor (GHS-R) mRNA levels during postnatal development in male and female rats, J. Neuroendocrinol, vol.11, pp.299-306, 1999.

H. Karasawa, C. Pietra, C. Giuliano, S. Garcia-rubio, X. Xu et al., New ghrelin agonist, HM01 alleviates constipation and L-dopa-delayed gastric emptying in 6-hydroxydopamine rat model of Parkinson's disease, Neurogastroenterol. Motil, 2014.

J. Kask, L. Ekselius, L. Brandt, N. Kollia, A. Ekbom et al., Mortality in Women With Anorexia Nervosa: The Role of Comorbid Psychiatric Disorders, Psychosom. Med, 2016.

Y. Kawahara, F. Kaneko, M. Yamada, Y. Kishikawa, H. Kawahara et al., Food reward-sensitive interaction of ghrelin and opioid receptor pathways in mesolimbic dopamine system, Neuropharmacology, vol.67, pp.395-402, 2013.

Y. Kawahara, H. Kawahara, F. Kaneko, M. Yamada, Y. Nishi et al., Peripherally administered ghrelin induces bimodal effects on the mesolimbic dopamine system depending on food-consumptive states, Neuroscience, vol.161, pp.855-864, 2009.

W. H. Kaye, G. K. Frank, and C. Mcconaha, Altered dopamine activity after recovery from restricting-type anorexia nervosa, Neuropsychopharmacol. Off. Publ. Am. Coll. Neuropsychopharmacol, vol.21, pp.503-506, 1999.

W. H. Kaye, J. L. Fudge, and M. Paulus, New insights into symptoms and neurocircuit function of anorexia nervosa, Nat. Rev. Neurosci, vol.10, pp.573-584, 2009.

W. H. Kaye, C. E. Wierenga, U. F. Bailer, A. N. Simmons, and A. Bischoff-grethe, Nothing tastes as good as skinny feels: the neurobiology of anorexia nervosa, Trends Neurosci, vol.36, pp.110-120, 2013.

E. Keen-rhinehart and T. J. Bartness, Peripheral ghrelin injections stimulate food intake, foraging, and food hoarding in Siberian hamsters, Am. J. Physiol. Regul. Integr. Comp. Physiol, vol.288, pp.716-722, 2005.

A. Kern, R. Albarran-zeckler, H. E. Walsh, and R. G. Smith, Apo-Ghrelin Receptor Forms Heteromers with DRD2 in Hypothalamic Neurons and Is Essential for Anorexigenic Effects of DRD2 Agonism, Neuron, vol.73, pp.317-332, 2012.

J. H. Kim and P. Vezina, Blockade of glutamate reuptake in the rat nucleus accumbens increases locomotor activity, Brain Res, vol.819, pp.165-169, 1999.

M. S. Kim, C. Y. Yoon, P. G. Jang, Y. J. Park, C. S. Shin et al., The mitogenic and antiapoptotic actions of ghrelin in 3T3-L1 adipocytes, Mol. Endocrinol. Baltim. Md, vol.18, pp.2291-2301, 2004.

M. Kohl, C. Foulon, and J. Guelfi, Hyperactivity and anorexia nervosa: behavioural and biological perspective, 2004.

M. Kojima, H. Hosoda, Y. Date, M. Nakazato, and H. Matsuo, Ghelin is a gowthhormone-releasing acylated peptide from stomach, Nature, vol.402, pp.656-660, 1999.

M. Kojima and K. Kangawa, Ghrelin: structure and function, Physiol. Rev, vol.85, pp.495-522, 2005.

D. Kontis and E. Theochari, Dopamine in anorexia nervosa: a systematic review, Behav. Pharmacol, vol.23, pp.496-515, 2012.

R. Kortekaas, K. L. Leenders, J. C. Van-oostrom, W. Vaalburg, J. Bart et al., Blood-brain barrier dysfunction in parkinsonian midbrain in vivo, Ann. Neurol, vol.57, pp.176-179, 2005.

A. Labarthe, O. Fiquet, R. Hassouna, P. Zizzari, L. Lanfumey et al., Ghrelin-Derived Peptides: A Link between Appetite/Reward, GH Axis, and Psychiatric Disorders?, Front. Endocrinol, vol.5, 2014.

R. Lage, M. J. Vázquez, L. Varela, A. K. Saha, A. Vidal-puig et al., Ghrelin effects on neuropeptides in the rat hypothalamus depend on fatty acid metabolism actions on BSX but not on gender, FASEB J, vol.24, pp.2670-2679, 2010.

S. Lammel, A. Hetzel, O. Häckel, I. Jones, B. Liss et al., Unique properties of mesoprefrontal neurons within a dual mesocorticolimbic dopamine system, Neuron, vol.57, pp.760-773, 2008.

S. Lammel, E. E. Steinberg, C. Földy, N. R. Wall, K. Beier et al., Diversity of transgenic mouse models for selective targeting of midbrain dopamine neurons, Neuron, vol.85, pp.429-438, 2015.

S. Lammel, K. M. Tye, and M. R. Warden, Progress in understanding mood disorders: optogenetic dissection of neural circuits, Genes Brain Behav, vol.13, pp.38-51, 2014.

J. L. Lanciego, N. Luquin, and J. A. Obeso, Functional neuroanatomy of the basal ganglia. Cold Spring Harb, Perspect. Med, vol.2, p.9621, 2012.

A. D. Lawrence, A. H. Evans, and A. J. Lees, Compulsive use of dopamine replacement therapy in Parkinson's disease: reward systems gone awry?, Lancet Neurol, vol.2, pp.595-604, 2003.

A. J. Lees, M. Selikhova, L. A. Andrade, and C. Duyckaerts, The black stuff and Konstantin Nikolaevich Tretiakoff, Mov. Disord. Off. J. Mov. Disord. Soc, vol.23, pp.777-783, 2008.

R. Legrand, N. Lucas, J. Breton, S. Azhar, J. Do-rego et al., Ghrelin treatment prevents development of activity based anorexia in mice, Eur. Neuropsychopharmacol. J. Eur. Coll. Neuropsychopharmacol, vol.26, pp.948-958, 2016.
URL : https://hal.archives-ouvertes.fr/hal-02295806

F. Lemarié, E. Beauchamp, P. Legrand, and V. Rioux, Revisiting the metabolism and physiological functions of caprylic acid (C8:0) with special focus on ghrelin octanoylation, Biochimie, vol.120, pp.40-48, 2016.

F. Levin, T. Edholm, P. T. Schmidt, P. Grybäck, H. Jacobsson et al., Ghrelin Stimulates Gastric Emptying and Hunger in Normal-Weight Humans, J. Clin. Endocrinol. Metab, vol.91, pp.3296-3302, 2006.

F. Lewy, Paralysis agitans. I. Pathologische Anatomie, 1912.

M. Lewandowski and . Berlin, , pp.920-933

M. Leyton, I. Boileau, C. Benkelfat, M. Diksic, G. Baker et al., Amphetamineinduced increases in extracellular dopamine, drug wanting, and novelty seeking: a PET/[11C]raclopride study in healthy men, Neuropsychopharmacol. Off. Publ. Am. Coll. Neuropsychopharmacol, vol.27, pp.1027-1035, 2002.

X. Li, J. Qi, T. Yamaguchi, H. Wang, and M. Morales, Heterogeneous composition of dopamine neurons of the rat A10 region: molecular evidence for diverse signaling properties, Brain Struct. Funct, vol.218, pp.1159-1176, 2013.

K. J. Livak and T. D. Schmittgen, Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method, Methods San Diego Calif, vol.25, pp.402-408, 2001.

S. H. Lockie and Z. B. Andrews, The hormonal signature of energy deficit: Increasing the value of food reward, Mol. Metab, vol.2, pp.329-336, 2013.

K. Majchrzak, K. M. Paw?owski, E. J. Orzechowska, I. Dolka, J. Mucha et al., A role of ghrelin in canine mammary carcinoma cells proliferation, apoptosis and migration, BMC Vet. Res, vol.8, p.170, 2012.

C. S. Maldonado-irizarry and A. E. Kelley, Excitotoxic lesions of the core and shell subregions of the nucleus accumbens differentially disrupt body weight regulation and motor activity in rat, Brain Res. Bull, vol.38, pp.551-559, 1995.

B. K. Mani, A. K. Walker, E. J. Lopez-soto, J. Raingo, C. E. Lee et al., Neuroanatomical characterization of a growth hormone secretagogue receptor-green fluorescent protein reporter mouse: Characterization of Ghsr-egfp reporter mouse, J. Comp. Neurol, vol.522, pp.3644-3666, 2014.

E. Markaki, J. Ellul, Z. Kefalopoulou, E. Trachani, A. Theodoropoulou et al., The Role of Ghrelin, Neuropeptide Y and Leptin Peptides in Weight Gain after Deep Brain Stimulation for Parkinson's Disease, Stereotact. Funct. Neurosurg, vol.90, pp.104-112, 2012.

S. Marrinan, A. V. Emmanuel, and D. J. Burn, Delayed gastric emptying in Parkinson's disease: Delayed Gastric Emptying in PD, Mov. Disord, vol.29, pp.23-32, 2014.

L. Martins, D. Fernández-mallo, M. G. Novelle, M. J. Vázquez, M. Tena-sempere et al., Hypothalamic mTOR Signaling Mediates the Orexigenic Action of Ghrelin, PLoS ONE, vol.7, 2012.

S. Mary, J. Fehrentz, M. Damian, G. Gaibelet, H. Orcel et al., Heterodimerization with Its Splice Variant Blocks the Ghrelin Receptor 1a in a Non-signaling Conformation, J. Biol. Chem, vol.288, pp.24656-24665, 2013.

N. Maswood, J. Young, E. Tilmont, Z. Zhang, D. M. Gash et al., Caloric restriction increases neurotrophic factor levels and attenuates neurochemical and behavioral deficits in a primate model of Parkinson's disease, Proc. Natl. Acad. Sci. U. S. A, vol.101, pp.18171-18176, 2004.

R. W. Mccallum, A. Lembo, T. Esfandyari, B. R. Bhandari, N. Ejskjaer et al., Phase 2b, randomized, double-blind 12-week studies of TZP-102, a ghrelin receptor agonist for diabetic gastroparesis, Neurogastroenterol. Motil, vol.25, pp.705-717, 2013.

M. R. Mcfarlane, M. S. Brown, J. L. Goldstein, and T. Zhao, Induced ablation of ghrelin cells in adult mice does not decrease food intake, body weight, or response to high-fat diet, Cell Metab, vol.20, pp.54-60, 2014.

Y. Mear, A. Enjalbert, and S. Thirion, GHS-R1a constitutive activity and its physiological relevance, Front. Neurosci, vol.7, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00966209

M. Méquinion, E. Caron, S. Zgheib, A. Stievenard, P. Zizzari et al., Physical activity: benefit or weakness in metabolic adaptations in a mouse model of chronic food restriction?, Am. J. Physiol. -Endocrinol. Metab, vol.308, pp.241-255, 2015.

M. Méquinion, F. Langlet, S. Zgheib, S. Dickson, B. Dehouck et al., Ghrelin: Central and Peripheral Implications in Anorexia Nervosa, 2013.

M. Merkestein, M. A. Brans, M. C. Luijendijk, J. W. De-jong, E. Egecioglu et al., Ghrelin Mediates Anticipation to a Palatable Meal in Rats, Obesity, vol.20, pp.963-971, 2012.

D. Miljic, S. Pekic, M. Djurovic, M. Doknic, N. Milic et al., Ghrelin has partial or no effect on appetite, growth hormone, prolactin, and cortisol release in patients with anorexia nervosa, J. Clin. Endocrinol. Metab, vol.91, pp.1491-1495, 2006.

C. Missale, S. R. Nash, S. W. Robinson, M. Jaber, and M. G. Caron, Dopamine receptors: from structure to function, Physiol. Rev, vol.78, pp.189-225, 1998.

R. E. Mistlberger, Circadian food-anticipatory activity: formal models and physiological mechanisms, Neurosci. Biobehav. Rev, vol.18, pp.171-195, 1994.

M. Moon, H. G. Kim, L. Hwang, J. Seo, S. Kim et al., Neuroprotective Effect of Ghrelin in the 1-Methyl-4-Phenyl-1,2,3,6-Tetrahydropyridine Mouse Model of Parkinson's Disease by Blocking Microglial Activation, Neurotox. Res, vol.15, pp.332-347, 2009.

T. D. Müller, R. Nogueiras, M. L. Andermann, Z. B. Andrews, S. D. Anker et al., Ghrelin. Mol. Metab, vol.4, pp.437-460, 2015.

E. Mutez, L. Larvor, F. Leprêtre, V. Mouroux, D. Hamalek et al., Transcriptional profile of Parkinson blood mononuclear cells with LRRK2 mutation, Neurobiol. Aging, vol.32, pp.1839-1848, 2011.

E. Mutez, A. Nkiliza, K. Belarbi, A. De-broucker, C. Vanbesien-mailliot et al., Involvement of the immune system, endocytosis and EIF2 signaling in both genetically determined and sporadic forms of Parkinson's disease, Neurobiol. Dis, vol.63, pp.165-170, 2014.

R. G. Nair-roberts, S. D. Chatelain-badie, E. Benson, H. White-cooper, J. P. Bolam et al., Stereological estimates of dopaminergic, GABAergic and glutamatergic neurons in the ventral tegmental area, substantia nigra and retrorubral field in the rat, Neuroscience, vol.152, pp.1024-1031, 2008.

A. M. Naleid, M. K. Grace, D. E. Cummings, and A. S. Levine, Ghrelin induces feeding in the mesolimbic reward pathway between the ventral tegmental area and the nucleus accumbens, Peptides, vol.26, pp.2274-2279, 2005.

K. Y. Ng, T. N. Chase, R. W. Colburn, and I. J. Kopin, L-Dopa-induced release of cerebral monoamines, Science, vol.170, pp.76-77, 1970.

K. Y. Ng, R. W. Colburn, and I. J. Kopin, Effects of L-dopa on efflux of cerebral monoamines from synaptosomes, Nature, vol.230, pp.331-332, 1971.

Y. Nishi, H. Mifune, A. Yabuki, Y. Tajiri, R. Hirata et al., Changes in Subcellular Distribution of n-Octanoyl or n-Decanoyl Ghrelin in Ghrelin-Producing Cells, 2013.

C. Nocjar and J. Panksepp, Chronic intermittent amphetamine pretreatment enhances future appetitive behavior for drug-and natural-reward: interaction with environmental variables, Behav. Brain Res, vol.128, pp.189-203, 2002.

L. Novakova, M. Haluzik, R. Jech, D. Urgosik, F. Ruzicka et al., Hormonal regulators of food intake and weight gain in Parkinson's disease after subthalamic nucleus stimulation, Neuro Endocrinol. Lett, vol.32, pp.437-441, 2011.

R. D. Oades and G. M. Halliday, Ventral tegmental (A10) system: neurobiology. 1. Anatomy and connectivity, Brain Res, vol.434, pp.117-165, 1987.

L. A. O'connell and H. A. Hofmann, Evolution of a Vertebrate Social Decision-Making Network, Science, vol.336, pp.1154-1157, 2012.

C. B. O'hara, I. C. Campbell, and U. Schmidt, A reward-centred model of anorexia nervosa: a focussed narrative review of the neurological and psychophysiological literature, Neurosci. Biobehav. Rev, vol.52, pp.131-152, 2015.

P. K. Olszewski, H. B. Schiöth, and A. S. Levine, Ghrelin in the CNS: from hunger to a rewarding and memorable meal?, Brain Res. Rev, vol.58, pp.160-170, 2008.

Y. Ono, T. Nakatani, Y. Sakamoto, E. Mizuhara, Y. Minaki et al., Differences in neurogenic potential in floor plate cells along an anteroposterior location: midbrain dopaminergic neurons originate from mesencephalic floor plate cells, Dev. Camb. Engl, vol.134, pp.3213-3225, 2007.

S. S. O'sullivan, K. Wu, M. Politis, A. D. Lawrence, A. H. Evans et al., Cue-induced striatal dopamine release in Parkinson's disease-associated impulsive-compulsive behaviours, Brain J. Neurol, vol.134, pp.969-978, 2011.

J. Overduin, D. P. Figlewicz, J. Bennett-jay, S. Kittleson, and D. E. Cummings, Ghrelin increases the motivation to eat, but does not alter food palatability, Am. J. Physiol. Regul. Integr. Comp. Physiol, vol.303, pp.259-269, 2012.

R. D. Palmiter, Dopamine signaling in the dorsal striatum is essential for motivated behaviors: lessons from dopamine-deficient mice, Ann. N. Y. Acad. Sci, vol.1129, pp.35-46, 2008.

C. Pañeda, A. I. Arroba, L. M. Frago, A. M. Holm, J. Rømer et al., Growth hormone-releasing peptide-6 inhibits cerebellar cell death in aged rats, Neuroreport, vol.14, pp.1633-1635, 2003.

J. Pantel, M. Legendre, S. Cabrol, L. Hilal, Y. Hajaji et al., Loss of constitutive activity of the growth hormone secretagogue receptor in familial short stature, J. Clin. Invest, vol.116, pp.760-768, 2006.
URL : https://hal.archives-ouvertes.fr/hal-02082146

G. Paxinos and K. B. Franklin, The mouse brain in stereotaxic coordinates, 2001.

S. Peng, S. Yu, Q. Wang, Q. Kang, Y. Zhang et al., Dopamine receptor D2 and catechol-Omethyltransferase gene polymorphisms associated with anorexia nervosa in Chinese Han population: DRD2 and COMT gene polymorphisms were associated with AN, Neurosci. Lett, vol.616, pp.147-151, 2016.

C. Perlemoine, F. Macia, F. Tison, I. Coman, D. Guehl et al., Effects of subthalamic nucleus deep brain stimulation and levodopa on energy production rate and substrate oxidation in Parkinson's disease, Br. J. Nutr, vol.93, pp.191-198, 2005.

A. L. Phu, Z. Xu, V. Brakoulias, N. Mahant, V. S. Fung et al., Effect of impulse control disorders on disability and quality of life in Parkinson's disease patients, J. Clin. Neurosci. Off. J. Neurosurg. Soc. Australas, vol.21, pp.63-66, 2014.

W. Poewe, Non-motor symptoms in Parkinson's disease, Eur. J. Neurol, vol.15, pp.14-20, 2008.

W. Poewe, F. Gerstenbrand, G. Ransmayr, and S. Plörer, Premorbid personality of Parkinson patients, J. Neural Transm. Suppl, vol.19, pp.215-224, 1983.

M. Poletti, G. Perugi, C. Logi, A. Romano, P. Del-dotto et al., Dopamine agonists and delusional jealousy in Parkinson's disease: A cross-sectional prevalence study, Mov. Disord, vol.27, pp.1679-1682, 2012.

P. E. Porporato, N. Filigheddu, S. Reano, M. Ferrara, E. Angelino et al., Acylated and unacylated ghrelin impair skeletal muscle atrophy in mice, J. Clin. Invest, vol.123, pp.611-622, 2013.

R. B. Postuma, D. Berg, M. Stern, W. Poewe, C. W. Olanow et al., MDS clinical diagnostic criteria for Parkinson's disease, Mov. Disord, vol.30, pp.1591-1601, 2015.

C. C. Probst and T. Van-eimeren, The Functional Anatomy of Impulse Control Disorders, Curr. Neurol. Neurosci. Rep, vol.13, 2013.

L. Pulvirenti, R. Berrier, M. Kreifeldt, and G. F. Koob, Modulation of locomotor activity by NMDA receptors in the nucleus accumbens core and shell regions of the rat, Brain Res, vol.664, pp.231-236, 1994.

D. Quarta, C. Di-francesco, S. Melotto, L. Mangiarini, C. Heidbreder et al., Systemic administration of ghrelin increases extracellular dopamine in the shell but not the core subdivision of the nucleus accumbens, Neurochem. Int, vol.54, pp.89-94, 2009.

M. Rask-andersen, P. K. Olszewski, A. S. Levine, and H. B. Schiöth, Molecular mechanisms underlying anorexia nervosa: focus on human gene association studies and systems controlling food intake, Brain Res. Rev, vol.62, pp.147-164, 2010.

S. Reano, A. Graziani, and N. Filigheddu, Acylated and unacylated ghrelin administration to blunt muscle wasting, Curr. Opin. Clin. Nutr. Metab. Care, vol.17, pp.236-240, 2014.

A. Rediger, C. L. Piechowski, K. Habegger, A. Grüters, H. Krude et al., MC4R Dimerization in the Paraventricular Nucleus and GHSR/MC3R Heterodimerization in the Arcuate Nucleus: Is There Relevance for Body Weight Regulation, Neuroendocrinology, vol.95, pp.277-288, 2012.

A. C. Ribeiro, G. Ceccarini, C. Dupré, J. M. Friedman, D. W. Pfaff et al., Contrasting effects of leptin on food anticipatory and total locomotor activity, PloS One, vol.6, 2011.

P. Riederer and S. Wuketich, Time course of nigrostriatal degeneration in parkinson's disease. A detailed study of influential factors in human brain amine analysis, J. Neural Transm, vol.38, pp.277-301, 1976.

T. E. Robinson and K. C. Berridge, Review. The incentive sensitization theory of addiction: some current issues, Philos. Trans. R. Soc. Lond. B. Biol. Sci, vol.363, pp.3137-3146, 2008.
URL : https://hal.archives-ouvertes.fr/insu-00357971

T. E. Robinson and K. C. Berridge, The neural basis of drug craving: an incentivesensitization theory of addiction, Brain Res. Brain Res. Rev, vol.18, pp.247-291, 1993.

M. A. Rossi, T. Sukharnikova, V. Y. Hayrapetyan, L. Yang, and H. H. Yin, Operant selfstimulation of dopamine neurons in the substantia nigra, PloS One, vol.8, p.65799, 2013.

H. Roux, C. Blanchet, C. Stheneur, E. Chapelon, and N. Godart, Somatic outcome among patients hospitalised for anorexia nervosa in adolescence: disorders reported and links with global outcome, Eat. Weight Disord. EWD, vol.18, pp.175-182, 2013.

Y. Saegusa, T. Hattori, M. Nahata, C. Yamada, and H. Takeda, A New Strategy Using Rikkunshito to Treat Anorexia and Gastrointestinal Dysfunction. Evid.-Based Complement, 2015.

, Altern. Med. ECAM, p.364260, 2015.

N. Sáez-francàs, G. Martí-andrés, N. Ramírez, O. De-fàbregues, J. Álvarez-sabín et al., Clinical and psychopathological factors associated with impulse control disorders in Parkinson's disease, Neurol. Barc. Spain, vol.31, pp.231-238, 2016.

J. D. Salamone and M. Correa, The mysterious motivational functions of mesolimbic dopamine, Neuron, vol.76, pp.470-485, 2012.

J. D. Salamone, K. Mahan, and S. Rogers, Ventrolateral striatal dopamine depletions impair feeding and food handling in rats, Pharmacol. Biochem. Behav, vol.44, pp.605-610, 1993.

M. Satou, Y. Nishi, J. Yoh, Y. Hattori, and H. Sugimoto, Identification and characterization of acyl-protein thioesterase 1/lysophospholipase I as a ghrelin deacylation/lysophospholipid hydrolyzing enzyme in fetal bovine serum and conditioned medium, Endocrinology, vol.151, pp.4765-4775, 2010.

G. Scanelli, M. Gualandi, M. Simoni, and E. Manzato, Somatic involvement assessed through a cumulative score of clinical severity in patients with eating disorders, Eat. Weight Disord. EWD, vol.19, pp.49-59, 2014.

M. Schaeffer, F. Langlet, C. Lafont, F. Molino, D. J. Hodson et al., Rapid sensing of circulating ghrelin by hypothalamic appetite-modifying neurons, Proc. Natl. Acad. Sci. U. S. A, vol.110, pp.1512-1517, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00796900

E. Schéle, T. Bake, C. Rabasa, and S. L. Dickson, Centrally Administered Ghrelin Acutely Influences Food Choice in Rodents, PloS One, vol.11, 2016.

H. Schellekens, T. G. Dinan, and J. F. Cryan, Taking two to tango: a role for ghrelin receptor heterodimerization in stress and reward, Front. Neurosci, vol.7, 2013.

A. J. Scheurink, G. J. Boersma, R. Nergårdh, and P. Södersten, Neurobiology of hyperactivity and reward: agreeable restlessness in anorexia nervosa, Physiol. Behav, vol.100, pp.490-495, 2010.

I. Seim, S. L. Carter, A. C. Herington, and L. K. Chopin, Complex organisation and structure of the ghrelin antisense strand gene GHRLOS, a candidate non-coding RNA gene, BMC Mol. Biol, vol.9, p.95, 2008.

S. Sharma, S. Singh, V. Sharma, V. P. Singh, and R. Deshmukh, Neurobiology of l-DOPA induced dyskinesia and the novel therapeutic strategies, Biomed. Pharmacother. Bioméd. Pharmacothérapie, vol.70, pp.283-293, 2015.

A. Shin, M. Camilleri, I. Busciglio, D. Burton, S. A. Smith et al., The ghrelin agonist RM-131 accelerates gastric emptying of solids and reduces symptoms in patients with type 1 diabetes mellitus, Clin. Gastroenterol. Hepatol. Off. Clin. Pract. J. Am. Gastroenterol. Assoc, vol.11, pp.1453-1459, 2013.

H. Sienkiewicz-jarosz, A. Scinska, L. Swiecicki, W. Lipczynska-lojkowska, W. Kuran et al., Sweet liking in patients with Parkinson's disease, J. Neurol. Sci, vol.329, pp.17-22, 2013.

E. H. Simpson, J. A. Waltz, C. Kellendonk, and P. D. Balsam, Schizophrenia in translation: dissecting motivation in schizophrenia and rodents, Schizophr. Bull, vol.38, pp.1111-1117, 2012.

K. P. Skibicka, C. Hansson, M. Alvarez-crespo, P. A. Friberg, and S. L. Dickson, Ghrelin directly targets the ventral tegmental area to increase food motivation, Neuroscience, vol.180, pp.129-137, 2011.

K. P. Skibicka, R. H. Shirazi, C. Rabasa-papio, M. Alvarez-crespo, C. Neuber et al., Divergent circuitry underlying food reward and intake effects of ghrelin: Dopaminergic VTA-accumbens projection mediates ghrelin's effect on food reward but not food intake, Neuropharmacology, vol.73, pp.274-283, 2013.

K. S. Smith, K. C. Berridge, and J. W. Aldridge, Disentangling pleasure from incentive salience and learning signals in brain reward circuitry, Proc. Natl. Acad. Sci. U. S. A, vol.108, 2011.

P. Södersten, C. Bergh, M. Leon, and M. Zandian, Dopamine and anorexia nervosa, Neurosci. Biobehav. Rev, vol.60, pp.26-30, 2016.

J. E. Steinglass, B. Figner, S. Berkowitz, H. B. Simpson, E. U. Weber et al., Increased capacity to delay reward in anorexia nervosa, J. Int. Neuropsychol. Soc. JINS, vol.18, pp.773-780, 2012.

F. J. Steyn, V. Tolle, C. Chen, and J. Epelbaum, Neuroendocrine Regulation of Growth Hormone Secretion, Compr. Physiol, vol.6, pp.687-735, 2016.

V. St-onge, A. Watts, and A. Abizaid, Ghrelin enhances cue-induced bar pressing for high fat food, Horm. Behav, vol.78, pp.141-149, 2016.

C. M. Stopper and S. B. Floresco, Contributions of the nucleus accumbens and its subregions to different aspects of risk-based decision making, Cogn. Affect. Behav. Neurosci, vol.11, pp.97-112, 2011.

P. Svenningsson, C. Rosenblad, K. Af-edholm-arvidsson, K. Wictorin, C. Keywood et al., Eltoprazine counteracts l-DOPAinduced dyskinesias in Parkinson's disease: a dose-finding study, Brain J. Neurol, vol.138, pp.963-973, 2015.

L. W. Swanson, The projections of the ventral tegmental area and adjacent regions: a combined fluorescent retrograde tracer and immunofluorescence study in the rat, Brain Res. Bull, vol.9, pp.321-353, 1982.

M. S. Szczypka, K. Kwok, M. D. Brot, B. T. Marck, A. M. Matsumoto et al., Dopamine production in the caudate putamen restores feeding in dopamine-deficient mice, Neuron, vol.30, pp.819-828, 2001.

K. Takagi, R. Legrand, A. Asakawa, H. Amitani, M. François et al., Anti-ghrelin immunoglobulins modulate ghrelin stability and its orexigenic effect in obese mice and humans, Nat. Commun, vol.4, p.2685, 2013.

H. Takeda, S. Muto, K. Nakagawa, S. Ohnishi, and M. Asaka, Rikkunshito and ghrelin secretion, Curr. Pharm. Des, vol.18, pp.4827-4838, 2012.

H. Takeda, S. Muto, K. Nakagawa, S. Ohnishi, C. Sadakane et al., Rikkunshito as a ghrelin enhancer, Methods Enzymol, vol.514, pp.333-351, 2012.

C. M. Tanner, F. Kamel, G. W. Ross, J. A. Hoppin, S. M. Goldman et al., Rotenone, Paraquat, and Parkinson's Disease, Environ. Health Perspect, vol.119, pp.866-872, 2011.

S. R. Taylor, S. Badurek, R. J. Dileone, R. Nashmi, L. Minichiello et al., GABAergic and glutamatergic efferents of the mouse ventral tegmental area, J. Comp. Neurol, vol.522, pp.3308-3334, 2014.

L. A. Tellez, W. Han, X. Zhang, T. L. Ferreira, I. O. Perez et al., Separate circuitries encode the hedonic and nutritional values of sugar, Nat. Neurosci, vol.19, pp.465-470, 2016.

C. Theander-carrillo, P. Wiedmer, P. Cettour-rose, R. Nogueiras, D. Perez-tilve et al., Ghrelin action in the brain controls adipocyte metabolism, J. Clin. Invest, vol.116, 1983.

N. M. Thompson, D. A. Gill, R. Davies, N. Loveridge, P. A. Houston et al., Ghrelin and des-octanoyl ghrelin promote adipogenesis directly in vivo by a mechanism independent of the type 1a growth hormone secretagogue receptor, Endocrinology, vol.145, pp.234-242, 2004.

C. J. Todes and A. J. Lees, The pre-morbid personality of patients with Parkinson's disease, J. Neurol. Neurosurg. Psychiatry, vol.48, pp.97-100, 1985.

C. Tomasetto, C. Wendling, M. C. Rio, and P. Poitras, Identification of cDNA encoding motilin related peptide/ghrelin precursor from dog fundus, Peptides, vol.22, pp.2055-2059, 2001.

K. N. Tretiakoff, Contribution à l'étude de l'anatomie pathologique du locus niger de Soemmering avec quelques deductions à la pathologie des troubles musculaires et de la maladie de Parkinson, 1919.

N. X. Tritsch, W. Oh, C. Gu, and B. L. Sabatini, Midbrain dopamine neurons sustain inhibitory transmission using plasma membrane uptake of GABA, not synthesis, vol.3, p.1936, 2014.

H. Ueno, T. Shiiya, and M. Nakazato, Translational research of ghrelin, Ann. N. Y. Acad. Sci, vol.1200, pp.120-127, 2010.

M. M. Unger, J. C. Möller, K. Mankel, K. M. Eggert, K. Bohne et al., Postprandial ghrelin response is reduced in patients with Parkinson's disease and idiopathic REM sleep behaviour disorder: a peripheral biomarker for early Parkinson's disease?, J. Neurol, vol.258, pp.982-990, 2011.

U. Ungerstedt, Stereotaxic mapping of the monoamine pathways in the rat brain, Acta Physiol. Scand. Suppl, vol.367, pp.1-48, 1971.

D. E. Vaillancourt, D. Schonfeld, Y. Kwak, N. I. Bohnen, and R. Seidler, Dopamine overdose hypothesis: Evidence and clinical implications: Dopamine Overdose Hypothesis, Mov. Disord, vol.28, pp.1920-1929, 2013.

L. A. Verhagen, E. Egecioglu, M. C. Luijendijk, J. J. Hillebrand, R. A. Adan et al., Acute and chronic suppression of the central ghrelin signaling system reveals a role in food anticipatory activity, Eur. Neuropsychopharmacol, vol.21, pp.384-392, 2011.

, Vicq d'Azyr, 1786. Traité d'anatomie et de physiologie, avec des planches coloriées. Représentant au naturel les divers organes de l'Homme et des Animaux. F.A. Didot

T. Vitalis, O. Cases, and J. G. Parnavelas, Development of the dopaminergic neurons in the rodent brainstem, Exp. Neurol, issue.1, pp.104-112, 0191.

N. D. Volkow, J. S. Fowler, and G. Wang, Role of dopamine in drug reinforcement and addiction in humans: results from imaging studies, Behav. Pharmacol, vol.13, pp.355-366, 2002.

N. D. Volkow and M. Morales, The Brain on Drugs: From Reward to Addiction, Cell, vol.162, pp.712-725, 2015.

A. Wagner, N. C. Barbarich-marsteller, G. K. Frank, U. F. Bailer, S. A. Wonderlich et al., Personality traits after recovery from eating disorders: do subtypes differ?, Int. J. Eat. Disord, vol.39, pp.276-284, 2006.

L. Wang, S. Mogami, H. Karasawa, C. Yamada, S. Yakabi et al., Preventive effect of rikkunshito on gastric motor function inhibited by l-dopa in rats, Peptides, vol.55, pp.136-144, 2014.

L. Wang, N. P. Murphy, A. Stengel, M. Goebel-stengel, D. H. Pierre et al., Ghrelin prevents levodopa-induced inhibition of gastric emptying and increases circulating levodopa in fasted rats: Ghrelin prevents l-dopa-delayed gastric emptying, Neurogastroenterol. Motil, vol.24, pp.235-245, 2012.

M. Watabe-uchida, L. Zhu, S. K. Ogawa, A. Vamanrao, and N. Uchida, Whole-brain mapping of direct inputs to midbrain dopamine neurons, Neuron, vol.74, pp.858-873, 2012.

D. Weintraub, J. Koester, M. N. Potenza, A. D. Siderowf, M. Stacy et al., Impulse control disorders in Parkinson disease: a cross-sectional study of 3090 patients, Arch. Neurol, vol.67, pp.589-595, 2010.

E. C. Weiselberg, M. Gonzalez, and M. Fisher, Eating disorders in the twenty-first century, Minerva Ginecol, vol.63, pp.531-545, 2011.

M. Wellman and A. Abizaid, Growth Hormone Secretagogue Receptor Dimers: A New Pharmacological Target. eNeuro 2, 2015.

T. Wichmann and M. R. Delong, Functional and pathophysiological models of the basal ganglia, Curr. Opin. Neurobiol, vol.6, pp.751-758, 1996.

R. A. Wise, Dopamine and reward: the anhedonia hypothesis 30 years on, Neurotox. Res, vol.14, pp.169-183, 2008.

J. M. Wo, N. Ejskjaer, P. M. Hellström, R. A. Malik, J. C. Pezzullo et al., Randomised clinical trial: ghrelin agonist TZP-101 relieves gastroparesis associated with severe nausea and vomiting--randomised clinical study subset data, Aliment. Pharmacol. Ther, vol.33, pp.679-688, 2011.

A. M. Wren, L. J. Seal, M. A. Cohen, A. E. Brynes, G. S. Frost et al., Ghrelin enhances appetite and increases food intake in humans, J. Clin. Endocrinol. Metab, vol.86, p.5992, 2001.

M. Wu, S. M. Brudzynski, and G. J. Mogenson, Functional interaction of dopamine and glutamate in the nucleus accumbens in the regulation of locomotion. Can, J. Physiol. Pharmacol, vol.71, pp.407-413, 1993.

C. L. Wyvell and K. C. Berridge, Incentive sensitization by previous amphetamine exposure: increased cue-triggered "wanting" for sucrose reward, J. Neurosci. Off. J. Soc. Neurosci, vol.21, pp.7831-7840, 2001.

T. Yamaguchi, J. Qi, H. Wang, S. Zhang, and M. Morales, Glutamatergic and dopaminergic neurons in the mouse ventral tegmental area, Eur. J. Neurosci, vol.41, pp.760-772, 2015.

T. Yamaguchi, H. Wang, X. Li, T. H. Ng, and M. Morales, Mesocorticolimbic glutamatergic pathway, J. Neurosci. Off. J. Soc. Neurosci, vol.31, pp.8476-8490, 2011.

K. Yamamoto and P. Vernier, The evolution of dopamine systems in chordates, Front. Neuroanat, vol.5, p.21, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00585570

D. Yang, Z. Liu, H. Zhang, and Q. Luo, Ghrelin protects human pulmonary artery endothelial cells against hypoxia-induced injury via PI3-kinase/Akt, Peptides, vol.42, pp.112-117, 2013.

J. Yu, H. Xu, X. Shen, and H. Jiang, Ghrelin protects MES23.5 cells against rotenone via inhibiting mitochondrial dysfunction and apoptosis, Neuropeptides, vol.56, pp.69-74, 2016.

A. Zastrow, S. Kaiser, C. Stippich, S. Walther, W. Herzog et al., Neural correlates of impaired cognitivebehavioral flexibility in anorexia nervosa, Am. J. Psychiatry, vol.166, pp.608-616, 2009.

Q. Y. Zhou and R. D. Palmiter, Dopamine-deficient mice are severely hypoactive, adipsic, and aphagic, Cell, vol.83, pp.1197-1209, 1995.

X. Zhu, Y. Cao, K. Voodg, and D. F. Steiner, On the Processing of Proghrelin to Ghrelin, 2006.

, J. Biol. Chem, vol.281, pp.38867-38870

J. M. Zigman, S. G. Bouret, and Z. B. Andrews, Obesity Impairs the Action of the Neuroendocrine Ghrelin System, Trends Endocrinol. Metab. TEM, vol.27, pp.54-63, 2016.

J. M. Zigman, J. E. Jones, C. E. Lee, C. B. Saper, and J. K. Elmquist, Expression of ghrelin receptor mRNA in the rat and the mouse brain, J. Comp. Neurol, vol.494, pp.528-548, 2006.

P. Zizzari, R. Hassouna, D. Grouselle, J. Epelbaum, and V. Tolle, Physiological roles of preproghrelin-derived peptides in GH secretion and feeding, Peptides, vol.32, pp.2274-2282, 2011.

, Dopaminergic neurons are mainly found in the parabrachial pigmented area (PBP, in green) largely spread out from bregma -3.08mm to bregma -3.68mm, and in the more posterior paranigral nucleus (PN, in green) located from bregma -3.40mm to bregma -3.68mm (Ikemoto, 2007), Schematic coronal representation of the antero-posterior organization of the ventral tegmental area in rodents, vol.8

;. Ikemoto and . Yamaguchi, Conversely, glutamatergic neurons are present at a lower density (~1 to 3%) in the PBP and the PN, 2007.

. Nair-roberts, They are also largely present in the rostromedial tegmental nucleus (RMTg, dark blue area) at bregma -4.04mm (Ikemoto, GABA-ergic neurons represent 30% of the PBP and the PN and are localized between bregma -3.40mm and bregma -3.68mm, 2007.

. Zigman, The different neuronal populations are shown in green for the dopamine, orange for the glutamate and blue for the GABA. (A8, retrorubral fields/A8 dopamine cells; IPN, Interpeduncular nucleus; ml, 2006.

, Diagnostic and statistical manual of mental disorders. Fourth Edition, 1994.

, American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition, DSM-5, 2013.

A. Asakawa, A. Inui, T. Kaga, H. Yuzuriha, T. Nagata et al., Ghrelin is an appetite-stimulatory signal from stomach with structural resemblance to motilin, Gastroenterology, vol.120, issue.2, pp.337-382, 2001.

S. N. Austad, Does caloric restriction in the laboratory simply prevent overfeeding and return house mice to their natural level of food intake?, Sci Aging Knowledge Environ, issue.6, p.3, 2001.

N. M. Avena and M. E. Bocarsly, Dysregulation of brain reward systems in eating disorders: neurochemical information from animal models of binge eating, bulimia nervosa, and anorexia nervosa, Neuropharmacology, vol.63, issue.1, pp.87-96, 2012.

N. Bellefontaine, K. Chachlaki, J. Parkash, C. Vanacker, W. Colledge et al., Leptin-dependent neuronal NO signaling in the preoptic hypothalamus facilitates reproduction, J Clin Invest, vol.2, issue.6, pp.2550-2559, 2014.

R. A. Boakes and I. Juraskova, The role of drinking in the suppression of food intake by recent activity, Behav Neurosci, vol.115, issue.3, pp.718-748, 2001.

R. A. Boakes, Self-starvation in the rat: running versus eating, Span J Psychol, vol.10, issue.2, pp.251-258, 2007.

D. W. Brann and V. B. Mahesh, Role of corticosteroids in female reproduction, FASEB J, vol.5, issue.12, pp.2691-2699, 1991.

K. M. Breen, V. G. Thackray, T. Hsu, R. A. Mak-mccully, D. Coss et al., Stress levels of glucocorticoids inhibit LH?-subunit gene expression in gonadotrope cells, Mol Endocrinol, vol.26, issue.10, pp.1716-1747, 2012.

M. D. Bruss, C. F. Khambatta, M. A. Ruby, I. Aggarwal, and M. K. Hellerstein, Calorie restriction increases fatty acid synthesis and whole body fat oxidation rates, Am J Physiol Endocrinol Metab, vol.298, issue.1, pp.108-124, 2010.

R. C. Casper, G. Pandey, J. B. Jaspan, and A. H. Rubenstein, Eating attitudes and glucose tolerance in anorexia nervosa patients at 8-year followup compared to control subjects, Psychiatry Res, vol.25, issue.3, pp.283-99, 1988.

R. C. Casper, Carbohydrate metabolism and its regulatory hormones in anorexia nervosa, Psychiatry Res, vol.16, issue.1, pp.85-96, 1996.

R. Dalle-grave, S. Calugi, and G. Marchesini, Compulsive exercise to control shape or weight in eating disorders: prevalence, associated features, and treatment outcome, Compr Psychiatry, vol.49, issue.4, pp.346-52, 2008.

C. Davis, Eating disorders and hyperactivity: a psychobiological perspective, Can J Psychiatry, vol.42, issue.2, pp.168-75, 1997.

L. P. De-moura, A. C. Sponton, M. B. De-araújo, R. A. Dalia, J. R. Pauli et al., Moderate physical activity from childhood contributes to metabolic health and reduces hepatic fat accumulation in adult rats, Lipids Health Dis, vol.6, p.29, 2013.

P. J. Delhanty, S. J. Neggers, and A. J. Van-der-lely, Should we cons, der des-acyl ghrelin as a separate hormone and if so, what does it do?, Front Horm Res, vol.42, pp.163-74, 2014.

C. De-luca, T. J. Kowalski, Y. Zhang, J. K. Elmquist, C. Lee et al., Complete rescue of obesity, diabetes, and infertility in db/db mice by neuron-specific LEPR-B transgenes, J Clin Invest, vol.115, issue.12, pp.3484-93, 2005.

M. J. Devlin, A. M. Cloutier, N. A. Thomas, D. A. Panus, S. Lotinun et al., Caloric restriction leads to high marrow adiposity and low bone mass in growing mice, J Bone Miner Res, vol.25, issue.9, pp.2078-88, 2010.

J. P. Difiori, H. J. Benjamin, J. S. Brenner, A. Gregory, N. Jayanthi et al., Overuse injuries and burnout in youth sports: a position statement from the American Medical Society for Sports Medicine, Br J Sports Med, vol.48, issue.4, pp.287-295, 2014.

D. Santos, Z. A. , D. Silva, R. J. Bacurau, R. F. Tirapegui et al., Effect of food restriction and intense physical training on estrous cyclicity and plasma leptin concentrations in rats, J Nutr Sci Vitaminol (Tokyo), vol.57, issue.1, pp.1-8, 2011.

M. Duclos, C. Gatti, B. Bessière, and P. Mormède, Tonic and phasic effects of corticosterone on food restriction-induced hyperactivity in rats, Psychoneuroendocrinology, vol.34, issue.3, pp.436-481, 2009.

M. Duclos, A. Ouerdani, P. Mormède, and J. P. Konsman, Food restriction-induced hyperactivity: addiction or adaptation to famine?, Psychoneuroendocrinology, vol.38, issue.6, pp.884-97, 2013.

B. Estour, N. Germain, E. Diconne, D. Frere, J. M. Cottet-emard et al., Hormonal profile heterogeneity and short-term physical risk in restrictive anorexia nervosa, J Clin Endocrinol Metab, vol.95, issue.5, pp.2203-2213, 2010.

C. Exner, J. Hebebrand, H. Remschmidt, C. Wewetzer, A. Ziegler et al., Leptin suppresses semistarvation induced hyperactivity in rats: implications for anorexia nervosa, Mol Psychiatry, vol.5, issue.5, pp.476-81, 2000.

R. Fernández-fernández, M. Tena-sempere, J. Roa, J. M. Castellano, V. M. Navarro et al., Direct stimulatory effect of ghrelin on pituitary release of LH through a nitric oxidedependent mechanism that is modulated by estrogen, vol.133, pp.1223-1255, 2007.

S. E. Gairdner and C. E. Amara, Serum leptin is not correlated with body fat in severe food restriction, Appl Physiol Nutr Metab, vol.37, issue.6, pp.1063-71, 2012.

C. Gelegen, D. A. Collier, I. C. Campbell, H. Oppelaar, J. Van-den-heuvel et al., Difference in susceptibility to activity-based anorexia in two inbred strains of mice, Eur Neuropsychopharmacol, vol.17, issue.3, pp.199-205, 2007.

N. Germain, B. Galusca, D. Grouselle, D. Frere, S. Billard et al., Ghrelin and obestatin circadian levels differentiate bingeing-purging from restrictive anorexia nervosa, J Clin Endocrinol Metab, vol.95, issue.6, pp.3057-62, 2010.

N. Germain, B. Galusca, L. Roux, C. W. Bossu, C. Ghatei et al., Constitutional thinness and lean anorexia nervosa display opposite concentrations of peptide YY, glucagon-like peptide 1, ghrelin, and leptin, Am J Clin Nutr, vol.85, issue.4, pp.967-71, 2007.

B. N. Greenwood, T. E. Foley, T. V. Le, P. V. Strong, A. B. Loughridge et al., Long-term voluntary wheel running is rewarding and produces plasticity in the mesolimbic reward pathway, Behav Brain Res, vol.1, issue.2, pp.354-62, 2011.

M. W. Hamrick, K. H. Ding, S. Ponnala, S. L. Ferrari, and C. M. Isales, Caloric restriction decreases cortical bone mass but spares trabecular bone in the mouse skeleton: implications for the regulation of bone mass by body weight, J Bone Miner Res, vol.23, issue.6, pp.870-878, 2008.

J. J. Hillebrand, M. P. Koeners, C. E. De-rijke, M. J. Kas, and R. A. Adan, Leptin treatment in activitybased anorexia, Biol Psychiatry, vol.15, issue.2, pp.165-71, 2005.

J. J. Hillebrand, A. A. Van-elburg, M. J. Kas, H. Van-engeland, and A. Ra, Olanzapine reduces physical activity in rats exposed to activity-based anorexia: possible implications for treatment of anorexia nervosa?, Biol Psychiatry, vol.15, issue.8, pp.651-658, 2005.

H. W. Hoek, Incidence, prevalence and mortality of anorexia nervosa and other eating disorders, Curr Opin Psychiatry, vol.19, issue.4, pp.389-94, 2006.

P. Jésus, W. Ouelaa, M. François, L. Riachy, C. Guérin et al., Alteration of intestinal barrier function during activity-based anorexia in mice, Clin Nutr, vol.15, issue.13, pp.310-315, 2013.

R. B. Kanarek, D. 'anci, K. E. Jurdak, N. Mathes, and W. F. , Running and addiction: precipitated withdrawal in a rat model of activity-based anorexia, Behav Neurosci, vol.123, issue.4, pp.905-917, 2009.

P. Kirialanis, P. Malliou, A. Beneka, and K. Giannakopoulos, Occurrence of acute lower limb injuries in artistic gymnasts in relation to event and exercise phase, Br J Sports Med, vol.37, issue.2, pp.137-146, 2003.

S. J. Klenotich and S. C. Dulawa, The activity-based anorexia mouse model, Methods Mol Biol, vol.829, pp.377-93, 2012.

I. Legroux-gérot, J. Vignau, E. Biver, P. Pigny, F. Collier et al., Anorexia nervosa, osteoporosis and circulating leptin: the missing link, Osteoporos Int, vol.21, issue.10, pp.1715-1737, 2010.

I. Legroux-gerot, J. Vignau, F. Collier, and B. Cortet, Bone loss associated with anorexia nervosa, Joint Bone Spine, vol.72, issue.6, pp.489-95, 2005.

I. Legroux-gerot, J. Vignau, F. Collier, and B. Cortet, Factors influencing changes in bone mineral density in patients with anorexia nervosa-related osteoporosis: the effect of hormone replacement therapy, Calcif Tissue Int, vol.83, issue.5, pp.315-338, 2008.

I. Legroux-gérot, J. Vignau, D. 'herbomez, M. Collier, F. Marchandise et al., Evaluation of bone loss and its mechanisms in anorexia nervosa, Calcif Tissue Int, vol.81, issue.3, pp.174-82, 2007.

A. R. Lucas, C. M. Beard, W. M. O'fallon, and L. T. Kurland, 50-year trends in the incidence of anorexia nervosa in Rochester, Minn.: a population-based study, Am J Psychiatry, vol.148, issue.7, pp.917-939, 1991.

G. Lusk, Animal calorimetry analysis of the oxidation of mixtures of carbohydrate and fat. A correction, J Biol Chem, vol.59, pp.41-42, 1924.

J. A. Mclean and G. Tobin, Animal and human calorimetry, 1987.

M. Méquinion, F. Langlet, S. Zgheib, S. Dickson, B. Dehouck et al., Ghrelin: central and peripheral implications in anorexia nervosa, Front Endocrinol, vol.4, p.15, 2013.

M. Merkestein, M. A. Brans, M. C. Luijendijk, J. W. De-jong, E. Egecioglu et al., Ghrelin mediates anticipation to a palatable meal in rats, Obesity, vol.20, issue.5, pp.963-71, 2012.

M. Merkestein, M. A. Van-gestel, E. M. Van-der-zwaal, M. A. Brans, M. C. Luijendijk et al., GHS-R1a signaling in the DMH and VMH contributes to food anticipatory activity, Int J Obes (Lond), vol.38, issue.4, pp.610-618, 2014.

M. Misra, K. K. Miller, C. Almazan, K. Ramaswamy, A. Aggarwal et al., Hormonal and body composition predictors of soluble leptin receptor, leptin, and free leptin index in adolescent girls with anorexia nervosa and controls and relation to insulin sensitivity, J Clin Endocrinol Metab, vol.89, issue.7, pp.3486-95, 2004.

G. J. Morton, K. J. Kaiyala, J. D. Fisher, K. Ogimoto, M. W. Schwartz et al., Identification of a physiological role for leptin in the regulation of ambulatory activity and wheel running in mice, Am J Physiol Endocrinol Metab, vol.300, issue.2, pp.392-401, 2011.

L. Nybo and N. H. Secher, Cerebral perturbations provoked by prolonged exercise, Prog Neurobiol, vol.72, issue.4, pp.223-61, 2004.

R. Ogata, T. Matsuzaki, T. Iwasa, M. Kiyokawa, N. Tanaka et al., Hypothalamic Ghrelin suppresses pulsatile secretion of luteinizing hormone via betaendorphin in ovariectomized rats, Neuroendocrinology, vol.90, issue.4, pp.364-70, 2009.

M. Padovani, J. A. Lavigne, G. V. Chandramouli, S. N. Perkins, J. C. Barrett et al., Distinct effects of calorie restriction and exercise on mammary gland gene expression in C57BL/6 mice, Cancer Prev Res (Phila), vol.2, issue.12, pp.1076-87, 2009.

K. M. Pirke, J. Pahl, U. Schweiger, and M. Warnhoff, Metabolic and endocrine indices of starvation in bulimia: a comparison with anorexia nervosa, Psychiatry Res, vol.15, issue.1, pp.33-42, 1985.

A. Routtenberg and A. W. Kuznesof, Self-starvation of rats living in activity wheels on a restricted feeding schedule, J Comp Physiol Psychol, vol.64, issue.3, pp.414-435, 1967.

J. Russell, L. A. Baur, P. J. Beumont, S. Byrnes, G. Gross et al., Altered energy metabolism in anorexia nervosa, Psychoneuroendocrinology, vol.26, issue.1, pp.51-63, 2001.

S. K. Sasse, B. N. Greenwood, C. V. Masini, T. J. Nyhuis, M. Fleshner et al., Chronic voluntary wheel running facilitates corticosterone response habituation to repeated audiogenic stress exposure in male rats, Stress, vol.11, issue.6, pp.425-462, 2008.

A. J. Scheurink, A. A. Ammar, B. Benthem, G. Van-dijk, and P. A. Södersten, Exercise and the regulation of energy intake, Int J Obes Relat Metab Disord, vol.3, pp.1-6, 1999.

A. J. Scheurink, G. J. Boersma, R. Nergårdh, and P. Södersten, Neurobiology of hyperactivity and reward: agreeable restlessness in anorexia nervosa, Physiol Behav, vol.14, issue.5, pp.490-495, 2010.

L. Smolak, S. K. Murnen, and A. E. Ruble, Female athletes and eating problems: a meta-analysis, Int J Eat Disord, vol.27, issue.4, pp.371-80, 2000.

J. R. Speakman and S. E. Mitchell, Caloric restriction, Mol Aspects Med, vol.32, issue.3, pp.159-221, 2011.

S. Tatsumi, M. Ito, Y. Asaba, K. Tsutsumi, and K. Ikeda, Life-long caloric restriction reveals biphasic and dimorphic effects on bone metabolism in rodents, Endocrinology, vol.149, issue.2, pp.634-675, 2008.

M. K. Torstveit, J. H. Rosenvinge, and J. Sundgot-borgen, Prevalence of eating disorders and the predictive power of risk models in female elite athletes: a controlled study, Scand J Med Sci Sports, vol.18, issue.1, pp.108-126, 2008.

M. K. Torstveit and J. Sundgot-borgen, Participation in leanness sports but not training volume is associated with menstrual dysfunction: a national survey of 1276 elite athletes and controls, Br J Sports Med, vol.39, issue.3, pp.141-148, 2005.

A. K. Uzum, B. Yucel, B. Omer, H. Issever, and N. C. Ozbey, Leptin concentration indexed to fat mass is increased in untreated anorexia nervosa (AN) patients, Clin Endocrinol (Oxf), vol.71, issue.1, pp.33-42, 2009.

L. A. Verhagen, E. Egecioglu, M. C. Luijendijk, J. J. Hillebrand, R. A. Adan et al., Acute and chronic suppression of the central ghrelin signaling system reveals a role in food anticipatory activity, Eur Neuropsychopharmacol, vol.21, issue.5, pp.384-92, 2011.

L. A. Verhagen, M. C. Luijendijk, J. J. Hillebrand, and R. A. Adan, Dopamine antagonism inhibits anorectic behavior in an animal model for anorexia nervosa, Eur Neuropsychopharmacol, vol.19, issue.3, pp.153-60, 2009.

F. Wasinski, R. F. Bacurau, M. R. Moraes, A. S. Haro, P. M. Moraes-vieira et al., Exercise and caloric restriction alter the immune system of mice submitted to a high-fat diet, Mediators Inflamm, p.395672, 2013.

K. Watanabe, C. Hara, and N. Ogawa, Feeding conditions and estrous cycle of female rats under the activity-stress procedure from aspects of anorexia nervosa, Physiol Behav, vol.51, issue.4, pp.827-859, 1992.

S. C. Woods, The eating paradox: how we tolerate food, Psychol Rev, vol.98, issue.4, pp.488-505, 1991.

A. ?ák, M. Vecka, E. Tvrzická, M. Hrubý, F. Novák et al., Composition of plasma fatty acids and non-cholesterol sterols in anorexia nervosa, Physiol Res, vol.54, issue.4, pp.443-51, 2005.

S. Zgheib, M. Méquinion, S. Lucas, D. Leterme, O. Ghali et al., Longterm physiological alterations and recovery in a mouse model of separation associated with time restricted feeding : a tool to study anorexia nervosa related consequences, 2014.

W. Zhang, A. Majumder, X. Wu, and M. W. Mulholland, The optimal EFS parameters were determined after using several values of voltage (5, 10 and 15V) and frequencies (10, 16 and 80 Hz) on test samples. We decided to stimulate the ENS from 2 months treated mice using three different pulse durations (200, 400 and 800 µs) and reduced the number to two different pulse durations (200 and 400 µs) for the 4 months treated mice. In all cases a train duration of 30 seconds with a voltage of 10V, a pulse delay of 0 seconds and a pulse rate of 16Hz were used. The reaction of the sample to EFS was determined after the last change of bath solution and after adding each of the following substances, atropine (ATR, 10 -5 M), guanethidine (GUA, 10 -5 M), serotonin (SHT, 10 -4 M) and tetrodotoxin, vol.297, pp.10-16, 2009.

, Additionally, to characterize the sudden increases and decreases in tension caused by the addition of the different substances resulting in an excitation or relaxation curve (see Figure?), an interface was created to manually determine the beginning of the curve, the maximum or minimum of the curve and the end of the curve. The program created with Matlab then automatically determined the slope, the amplitude of the increase/decrease, the e-Factor and the e-Exponent of the curve. The effect for each substance concentration on the different an ice-cold eppendorf holder using ice cold protein extraction buffer (60 mM TrisHCl (pH 6,8), 1mM Na3VO4, 1% SDS) (300µl) in the presence of protease and phosphatase inhibitors (phenylmethylsulphonyl fluoride (PMSF, Sigma Aldrich 78830-5g), leupeptin (Roche, 11017101001) and aprotinin (Roche, 10236624001), sodium fluoride, Contractility data for each time-point was translated into a curve by the data acquisition system. Parameters such as maximal force, minimal force, amplitude and frequency before and after each treatment were automatically extracted using a self-made program written with Matlab (version R2014b

, Samples were then diluted in each case to have the same end protein concentration of 30µg and 20 µl of protein extract were loaded together with 5 µl of loading buffer (Invitrogen, Germany, EU) into a 4-containing 5% methanol, at 10V during 3 hours. To verify the efficiency of the transfer, membranes were incubated in a Red Ponceau solution during 2 min and washed 3 times (3*5min) in deionised water. The aspecific signal was then blocked by incubation of the membranes in Tris-buffered saline + 0.1% Tween 20 (TBST 1X) and either 5% (w/v) non-fat dry milk for total protein study or 5% (w/v) bovine serum albumine for phosphorylated-antigens, SDS-PAGE and Western Blotting Protein concentration from intestine extracts was measured using the Nanodrop X100, pp.40101-150

, On the following day, blots were washed 3 x 5 min. with TBST 1X and incubated with horseradish peroxidase-conjugated anti-rabbit and anti-mouse (1:5000, provider) at room temperature for 1 in TBST 1X and developed with the Prime Western Blotting detection Reagent (GE Healthcare Amersham RPN2232) using the LAS3000 bioimager (Fujifilm

, dopamine and electric field stimulation before and after atropine, guanethidine and serotonin addition on the motility of duodenum, jejunum, ileum and colon from vehicle and rotenone treated mice. Our results show that rotenone induces changes in the dose-response curves to carbachol or dopamine in all tested regions. These alterations are region and treatment-time dependent. We did not observe difference in the basal frequency or the basal mean amplitude (max-min) (i.e. after stabilizing the samples and before adding any substances) between the samples coming from rotenone-treated or control mice (Supplementary Figure 1). contractility of the intestine. We compared the GC dose-response curves to carbachol in all different segments from 2 months and 4 months treated control and rotenone-treated mice

, After 4 months of treatment, differences are observed in the slope and in the amplitude of increase of the curve generated by carbachol in rotenone treated mice when compared to controls (slope: p< 0.01;amplitude of increase: p=0.0038, see Figure 2A and 2B). No differences between control and rotenone treated samples were, After two months of treatment, we observed an increased reaction to carbachol in the duodenum of rotenone treated samples when compared to control. Specifically the ?maximum (p<0.001)

, 001) in rotenone-treated samples when compared to control after 2 months (Figure 1D and 1E). Interestingly, the post-pre differences in the frequency (p<0.05), the slope (p<0.01) and the amplitude of increase (p<0.05) were higher in samples coming from control mice (Figure 1F-H), the jejunum, we observed an increase reaction to carbachol in the ?maximum (p<0.05) and the ?mean amplitude (p<0

, Whereas the mean amplitude was higher on rotenone treated samples (p<0.01), the slope (p<0.001), the amplitude of increase (p<0.001) and the e-factor (p<0.05) were higher on control mice (Figure 1I-L). Interestingly, after 4 months of rotenone treatment carbachol induced a higher reaction on samples coming from vehicle treated animals when compared to rotenone treated samples (Figure 2G)(p<0.01). No significant difference was observed in any of the other measured parameters, Interestingly the effect of rotenone on the ileum was more heterogeneous after 2 months of treatment

, After 4 months, rotenone treatment increased the mean amplitude (p<0.05) and decreased the frequency (p<0.05) in rotenone-treated mice when compared to control (Figure 2H-I)

, The differences observed on the effect of AT on the C-Off phase between rotenone treated and control samples was opposite to the one observed in the duodenum, the ileum and the colon. AT induced a significant decrease in the contractility of control mice, the jejunum, we observed no differences in the effect of AT on the R1 or C-Off phases of EFS 200 or EFS 400 after 2 months of treatment (Figure 6E-H), vol.87

, the R1 phase with higher relaxations during EFS 400 in rotenone treated mice (EFS 400: 136.076±36.44, p>0.05) when compared to control samples (EFS 400: 30.042±21.168, p>0.01) (Figure 6K)

, The effect of AT on the C-Off contractility was higher on treated samples, Further 2 months of treatment induced a non-significant higher R1-relaxation in the presence of AT on control mice

, After 4 months of treatment, AT had no effect on the C-Off contractility in control or treated mice (Figure 7N and P). Interestingly, the effect of AT on the R1 EFS-induced relaxation was opposite to all other intestinal regions and to the effect after 2 months of treatment on treated samples. AT diminished the relaxation induced by EFS in both control, p>0.05) and rotenone treated (EFS 200: 230.193%±98.75, p>0.05) samples in the R1 phase during the EFS 200 (Figure 6M), vol.87

, Altogether these results suggest that rotenone treatment affects cholinergic innervation in the intestine. The quality and degree of dysfunction depends on the region affected

H. Braak, M. Sastre, J. R. Bohl, and R. A. De-vos, Del Tredici K: Parkinson's disease: lesions in dorsal horn layer I, involvement of parasympathetic and sympathetic pre-and postganglionic neurons, Acta Neuropathol, vol.113, issue.4, pp.421-429, 2007.

K. Wakabayashi, H. Takahashi, E. Ohama, and F. Ikuta, Parkinson's disease: an immunohistochemical study of Lewy body-containing neurons in the enteric nervous system, Acta Neuropathol, vol.79, issue.6, pp.581-583, 1990.

K. Wakabayashi and H. Takahashi, Cellular pathology in multiple system atrophy, Neuropathology, vol.26, issue.4, pp.338-345, 2006.

H. Braak, R. A. De-vos, J. Bohl, D. Tredici, and K. , Gastric alpha-synuclein immunoreactive inclusions in Meissner's and Auerbach's plexuses in cases staged for Parkinson's disease-related brain pathology, Neurosci Lett, vol.396, issue.1, pp.67-72, 2006.

H. Braak, E. Ghebremedhin, U. Rub, and H. Bratzke, Del Tredici K: Stages in the development of Parkinson's disease-related pathology, Cell Tissue Res, vol.318, issue.1, pp.121-134, 2004.

E. Wolters and H. Braak, Parkinson's disease: premotor clinico-pathological correlations, J Neural Transm Suppl, issue.70, pp.309-319, 2006.

K. Krogh and P. Christensen, Neurogenic colorectal and pelvic floor dysfunction. Best practice & research Clinical gastroenterology, vol.23, pp.531-543, 2009.

S. Marrinan, A. V. Emmanuel, and D. J. Burn, Delayed gastric emptying in Parkinson's disease, Mov Disord, vol.29, issue.1, pp.23-32, 2014.

R. F. Pfeiffer, Gastrointestinal dysfunction in Parkinson's disease, Lancet Neurol, vol.2, issue.2, pp.107-116, 2003.

F. J. Pan-montojo and R. H. Funk, Oral administration of rotenone using a gavage and image analysis of alpha-synuclein inclusions in the enteric nervous system, Journal of visualized experiments, issue.44, 2010.

F. Pan-montojo, M. Schwarz, C. Winkler, M. Arnhold, G. A. O&apos;sullivan et al., Environmental toxins trigger PD-like progression via increased alpha-synuclein release from enteric neurons in mice, Scientific reports, vol.2012, p.898

J. I. Nagy, V. Urena-ramirez, and J. E. Ghia, Functional alterations in gut contractility after connexin36 ablation and evidence for gap junctions forming electrical synapses between nitrergic enteric neurons, FEBS Lett, vol.588, issue.8, pp.1480-1490, 2014.

S. Bertoni, G. Gabella, P. Ghizzardi, V. Ballabeni, M. Impicciatore et al., Motor responses of rat hypertrophic intestine following chronic obstruction. Neurogastroenterology and motility : the official journal of the, European Gastrointestinal Motility Society, vol.16, issue.3, pp.365-374, 2004.

A. Maass and H. Reichmann, Sleep and non-motor symptoms in Parkinson's disease, J Neural Transm, vol.120, issue.4, pp.565-569, 2013.

L. Klingelhoefer and H. Reichmann, Pathogenesis of Parkinson disease-the gutbrain axis and environmental factors, Nature reviews Neurology, vol.11, issue.11, pp.625-636, 2015.

D. Aarsland, S. Pahlhagen, C. G. Ballard, U. Ehrt, and P. Svenningsson, Depression in Parkinson disease--epidemiology, mechanisms and management, Nature reviews Neurology, vol.2012, issue.1, pp.35-47

W. Chen, Z. M. Xu, G. Wang, and S. D. Chen, Non-motor symptoms of Parkinson's disease in China: a review of the literature, Parkinsonism Relat Disord, vol.18, issue.5, pp.446-452, 2012.

R. F. Pfeiffer, Autonomic dysfunction in Parkinson's disease. Expert review of neurotherapeutics, vol.12, pp.697-706, 2012.

G. Defazio, A. Gigante, P. Mancino, and M. Tinazzi, The epidemiology of pain in Parkinson's disease, J Neural Transm, vol.120, issue.4, pp.583-586, 2013.

A. Todorova, P. Jenner, R. Chaudhuri, and K. , Non-motor Parkinson's: integral to motor Parkinson's, yet often neglected, Practical neurology, vol.14, issue.5, pp.310-322, 2014.

T. Amino, S. Orimo, Y. Itoh, A. Takahashi, T. Uchihara et al., Profound cardiac sympathetic denervation occurs in Parkinson disease, Brain Pathol, vol.15, issue.1, pp.29-34, 2005.

S. Orimo, T. Amino, Y. Itoh, A. Takahashi, T. Kojo et al., Cardiac sympathetic denervation precedes neuronal loss in the sympathetic ganglia in Lewy body disease, Acta Neuropathol, vol.109, issue.6, pp.583-588, 2005.

A. B. Manning-bog, A. L. Mccormack, J. Li, V. N. Uversky, A. L. Fink et al., The herbicide paraquat causes up-regulation and aggregation of alphasynuclein in mice: paraquat and alpha-synuclein, J Biol Chem, vol.277, issue.3, pp.1641-1644, 2002.

A. B. Manning-bog, A. L. Mccormack, M. G. Purisai, L. M. Bolin, D. Monte et al., Alphasynuclein overexpression protects against paraquat-induced neurodegeneration, The Journal of neuroscience : the official journal of the Society for Neuroscience, vol.23, issue.8, pp.3095-3099, 2003.

D. A. Di-monte, The environment and Parkinson's disease: is the nigrostriatal system preferentially targeted by neurotoxins?, Lancet Neurol, vol.2, issue.9, pp.531-538, 2003.

J. M. Gorell, C. C. Johnson, B. A. Rybicki, E. L. Peterson, and R. J. Richardson, The risk of Parkinson's disease with exposure to pesticides, farming, well water, and rural living, Neurology, vol.50, issue.5, pp.1346-1350, 1998.

C. M. Tanner, F. Kamel, G. W. Ross, J. A. Hoppin, S. M. Goldman et al., Rotenone, paraquat, and Parkinson's disease, vol.119, pp.866-872, 2011.

R. Betarbet, T. B. Sherer, G. Mackenzie, M. Garcia-osuna, A. V. Panov et al., Chronic systemic pesticide exposure reproduces features of Parkinson's disease, Nat Neurosci, vol.3, issue.12, pp.1301-1306, 2000.

T. B. Sherer, R. Betarbet, C. M. Testa, B. B. Seo, J. R. Richardson et al., Mechanism of toxicity in rotenone models of Parkinson's disease, J Neurosci, vol.23, issue.34, pp.10756-10764, 2003.

T. B. Sherer, R. Betarbet, and J. T. Greenamyre, Pesticides and Parkinson's disease, ScientificWorldJournal, vol.1, pp.207-208, 2001.

S. Holmqvist, O. Chutna, L. Bousset, P. Aldrin-kirk, W. Li et al., Direct evidence of Parkinson pathology spread from the gastrointestinal tract to the brain in rats, Acta Neuropathol, vol.128, issue.6, pp.805-820, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01155404

K. C. Luk, V. Kehm, J. Carroll, B. Zhang, P. O&apos;brien et al., Pathological alpha-synuclein transmission initiates Parkinson-like neurodegeneration in nontransgenic mice, Science, vol.2012, issue.6109, pp.949-953

P. Desplats, H. J. Lee, E. J. Bae, C. Patrick, E. Rockenstein et al., Inclusion formation and neuronal cell death through neuron-toneuron transmission of alpha-synuclein, Proc Natl Acad Sci, vol.106, issue.31, pp.13010-13015, 2009.

E. Svensson, E. Horvath-puho, R. W. Thomsen, J. C. Djurhuus, L. Pedersen et al., Vagotomy and subsequent risk of Parkinson's disease, Ann Neurol, vol.78, issue.4, pp.522-529, 2015.