W. Poewe, K. Seppi, and C. M. Tanner, Nat Rev DisPrimers, vol.23, p.17013, 2017.

M. Kestenbaum and R. N. Alcalay, Clinical features of LRRK2 carriers with Parkinson's disease, Advances in Neurobiology, vol.14, pp.31-48

H. T. Benamer and R. De-silva, LRRK2 G2019S in the North African popu-lation: a review, Eur Neurol, vol.63, issue.6, pp.321-325, 2010.

R. Inzelberg, S. Hassin-baer, and J. Jankovic, Genetic movement disorders in patients of Jewish ancestry, JAMA Neurol, vol.71, issue.12, pp.1567-1572, 2014.

R. B. Postuma, D. Berg, and M. Stern, MDS clinical diagnostic criteria for Parkinson's disease, Mov Disord, vol.30, issue.12, pp.1591-1601, 2015.

C. L. Tomlinson, R. Stowe, and S. Patel, Systematic review of levodopa dose equivalency reporting in Parkinson's disease, Mov Disord, vol.25, issue.15, pp.2649-2653, 2010.

S. Fahn and R. L. Elton, The unified Parkinson's disease rating scale, RecentDevelopments in Parkinson's Disease, vol.1987, pp.153-163

M. M. Hoehn and M. D. Yahr, Parkinsonism: onset progression and mortality, Neurology, vol.17, issue.5, pp.427-442, 1967.

M. Folstein, S. Folstein, and P. Mchugh, Mini--Mental State" A practical method for grading the cognitive state of patients for the clinician, J Psychiatr Res, vol.12, issue.3, pp.189-198, 1975.

R. F. Uhlmann and E. B. Larson, Effect of education on the mini--mental state examination as a screening test for dementia, J Am Geriatr Soc, vol.39, issue.9, pp.876-880, 1991.

A. T. Beck, R. A. Steer, and R. Ball, Comparison of Beck depression inventories--IA and --II in psychiatric outpatients, J Pers Assess, vol.67, issue.3, pp.588-597, 1996.

Y. P. Wang and C. Gorenstein, Assessment of depression in medical patients: a systematic review of the utility of the Beck depression inventory--II, Clinics (Sao Paulo), vol.68, issue.9, pp.1274-1287, 2013.

J. Jankovic, M. Mcdermott, and J. Carter, Variable expression of Parkinson's disease: a base--line analysis of the DATATOP cohort. The Parkinson study group, Neurology, vol.40, issue.10, pp.1529-1534, 1990.

S. Luciano, M. Wang, C. Ortega, and R. A. , Sex differences in LRRK2 G2019S and idiopathic Parkinson's disease, Ann Clin Transl Neurol, vol.4, issue.11, pp.801-810, 2017.

K. Marder, Y. Wang, and R. N. Alcalay, Age--specific penetrance of LRRK2 G2019S in the Michael J. Fox Ashkenazi Jewish LRRK2 con-sortium, Neurology, vol.85, issue.1, pp.89-95, 2015.

G. Yahalom, Y. Orlev, and O. S. Cohen, Motor progression of Parkinson's disease with the leucine--rich repeat kinase 2 G2019S mutation, Mov Disord, vol.29, issue.8, pp.1057-1060, 2014.

D. G. Healy, M. Falchi, and S. S. O'sullivan, Phenotype, genotype, and worldwide genetic penetrance of LRRK2--associated Parkinson's disease: a case--control study, Lancet Neurol, vol.7, issue.7, pp.583-590, 2008.

K. Nishioka, M. Kefi, and B. Jasinska-myga, A comparative study of LRRK2, PINK1 and genetically undefined familial Parkinson's dis-ease, J Neurol Neurosurg Psychiatry, vol.81, issue.4, pp.391-395, 2010.

C. Marras, B. Schule, and R. P. Munhoz, Phenotype in parkinsonian and nonparkinsonian LRRK2 G2019S mutation carriers, Neurology, vol.77, issue.4, pp.325-333, 2011.

R. N. Alcalay, A. Mirelman, and R. Saunders-pullman, Parkinson disease phenotype in Ashkenazi Jews with and without LRRK2 G2019S mutations, Mov Disord, vol.28, issue.14, pp.1966-1971, 2013.

J. Trinh, R. Amouri, and J. E. Dudac, A comparative study of Parkinson's disease and leucine--rich repeat kinase 2 p. G2019S par-kinsonism, Neurobiol Aging, vol.35, issue.5, pp.1125-1131, 2014.

A. Bouhouche, H. Tibar, B. E. Haj, and R. , LRRK2 G2019S mutation: prevalence and clinical features in moroccans with Parkinson's dis-ease, Parkinsons Dis, p.2412486, 2017.

L. Ishihara, R. A. Gibson, and L. Warren, Screening for Lrrk2 G2019S and clinical comparison of Tunisian and North American Caucasian Parkinson's disease families, Mov Disord, vol.22, issue.1, pp.55-61, 2007.

C. Marras, R. N. Alcalay, and C. Caspell-garcia, Motor and non--motor heterogeneity of LRRK2--related and idiopathic Parkinson's disease, Mov Disord, vol.31, issue.8, pp.1192-1202, 2016.

A. Mirelman, T. Heman, and K. Yasinovsky, Fall risk and gait in Parkinson's disease: the role of the LRRK2 G2019S mutation, MovDisord, vol.28, issue.12, pp.1683-1690, 2013.

R. N. Alcalay, M. Santana, H. Mirelman, and A. , Neuropsychological performance in LRRK2 G2019S carriers with Parkinson's disease, Parkinsonism Relat Disord, vol.21, pp.106-110, 2015.

R. Saunders-pullman, A. Mirelman, and R. N. Alcalay, Progression in the LRRK2--associated Parkinson disease population, JAMA Neurol, vol.75, issue.3, pp.312-319, 2018.

F. Nabli, B. Sassi, S. Amouri, and R. , Motor phenotype of LRRK2--associated Parkinson's disease: a Tunisian longitudinal study, MovDisord, vol.30, issue.2, pp.253-258, 2015.

J. H. Somme, M. Salazar, A. Gonzalez, and A. , Cognitive and be-havioral symptoms in Parkinson's disease patients with the G2019S and R1441G mutations of the LRRK2 gene, Parkinsonism RelatDisord, vol.21, issue.5, pp.494-499, 2015.

S. Srivatsal, B. Cholerton, and J. B. Leverenz, Cognitive profile of LRRK2--related Parkinson's disease, Mov Disord, vol.30, issue.5, pp.728-733, 2015.

B. Sassi, S. Nabli, F. Hentati, and E. , Cognitive dysfunction in Tunisian LRRK2 associated Parkinson's disease, Parkinsonism RelatDisord, vol.18, issue.3, pp.243-246, 2012.

Z. Landoulsi, S. Ben-romdhan, B. Djebara, and M. , Using KASP technique to screen LRRK2 G2019S mutation in a large Tunisian cohort, BMC Med Genet, vol.18, issue.1, p.70, 2017.
URL : https://hal.archives-ouvertes.fr/pasteur-01639043

A. Orr-urtreger, C. Shifrin, and U. Rozovski, The LRRK2 G2019S mu-tation in Ashkenazi Jews with Parkinson disease: is there a gender effect?, Neurology, vol.69, issue.16, pp.1595-1602, 2007.

R. Cilia, C. Siri, and D. Rusconi, LRRK2 mutations in Parkinson's disease: confirmation of a gender effect in the Italian population, Parkinsonism Relat Disord, vol.20, issue.8, pp.911-914, 2014.

Z. Gan-or, C. S. Leblond, and V. Mallett, LRRK2 mutations in Parkinson disease; a sex effect or lack thereof? A meta--analysisParkinsonism Relat Disord, vol.21, pp.778-782, 2015.

D. Georgiev, K. Hamberg, and M. Hariz, Gender differences in Parkinson's disease: a clinical perspective, Acta Neurol Scand, vol.136, issue.6, pp.570-584, 2017.

L. Ishihara, L. Warren, and R. Gibson, Clinical features of Parkinson disease patients with homozygous leucine--rich repeat kinase 2 G2019S mutations, Arch Neurol, vol.63, issue.9, pp.1250-1254, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00221455

S. Ben-romdhan, N. Farhat, and A. Nasri, How to cite this article

, LRRK2 G2019S Parkinson's disease with more benign phenotype than idiopathic

, Acta Neurol Scand, vol.00, pp.1-7, 2018.

,

R. Di-paolo, G. , D. Camilli, and P. , Phosphoinositides in cell regulation and membrane dynamics, Nature, vol.443, pp.651-657, 2006.

,

D. A. Dyment, A. C. Smith, P. Humphreys, J. Schwartzentruber, C. L. Beaulieu et al., Homozygous nonsense mutation in SYNJ1 associated with intractable epilepsy and tau pathology, Neurobiol Aging, vol.36, pp.1222-1223, 2015.

C. Haffner, K. Takei, H. Chen, N. Ringstad, A. Hudson et al., Synaptojanin 1: localization on coated endocytic intermediates in nerve terminals and interaction of its 170 kDa isoform with Eps15, FEBS Lett, vol.419, pp.1451-1459, 1997.

K. Hardies, Y. Cai, C. Jardel, A. C. Jansen, M. Cao et al., Loss of SYNJ1 dual phosphatase activity leads to early onset refractory seizures and progressive neurological decline, EuroEPINOMICS RES Consortium, vol.139, pp.2420-2430, 2016.

H. Houlden and A. B. Singleton, The genetics and neuropathology of Parkinson's disease, Acta Neuropathol (Berl), vol.124, pp.325-338, 2012.

L. Kirola, M. Behari, C. Shishir, and B. K. Thelma, Identification of a novel homozygous mutation Arg459Pro in SYNJ1 gene of an Indian family with autosomal recessive juvenile Parkinsonism, Parkinsonism Relat Disord, vol.31, pp.124-128, 2016.

,

M. Krauß and V. Haucke, Phosphoinositide-metabolizing enzymes at the interface between membrane traffic and cell signalling, EMBO Rep, vol.8, pp.241-246, 2007.

C. E. Krebs, S. Karkheiran, J. C. Powell, M. Cao, V. Makarov et al., The Sac1 domain of SYNJ1 identified mutated in a family with early-onset progressive Parkinsonism with generalized seizures, Hum Mutat, vol.34, pp.1200-1207, 2013.

M. Mani, S. Y. Lee, L. Lucast, O. Cremona, G. Di-paolo et al., The dual phosphatase activity of synaptojanin1 is required for both efficient synaptic vesicle endocytosis and reavailability at nerve terminals, Neuron, vol.56, pp.1004-1018, 2007.

J. Massano and K. P. Bhatia, Clinical approach to Parkinson's disease: features, diagnosis, and principles of management. Cold Spring Harb Perspect Med 2:a008870, 2012.

P. S. Mcpherson, E. P. Garcia, and V. I. Slepnev, A presynaptic inositol-5-phosphatase, Nature, vol.379, pp.353-357, 1996.

M. Mettlen, M. Stoeber, D. Loerke, C. N. Antonescu, G. Danuser et al., Endocytic accessory proteins are functionally distinguished by their differential effects on the maturation of clathrin-coated pits, Mol Biol Cell, vol.20, pp.3251-3260, 2009.

S. Olgiati, D. Rosa, A. Quadri, and M. , PARK20 caused by SYNJ1 homozygous Arg258Gln mutation in a new Italian family, Neurogenetics, vol.15, pp.183-188, 2014.

R. M. Perera, R. Zoncu, L. Lucast, P. De-camilli, and D. Toomre, Two synaptojanin 1 isoforms are recruited to clathrin-coated pits at different stages, Proc Natl Acad Sci U S A, vol.103, pp.19332-19337, 2006.

M. Picillo, A. Ranieri, G. Orefice, V. Bonifati, and P. Barone, Clinical progression of SYNJ1-related early onset atypical parkinsonism: 3-year follow up of the original Italian family, J Neurol, vol.261, pp.823-824, 2014.

G. J. Praefcke, M. G. Ford, and E. M. Schmid, Evolving nature of the AP2 ?-appendage hub during clathrin, 2004.

, Monogenic Parkinson's disease and parkinsonism: clinical phenotypes and frequencies of known mutations, Parkinsonism Relat Disord, vol.23, pp.407-415, 2013.

A. Puschmann, New genes causing hereditary Parkinson's disease or parkinsonism, Curr Neurol Neurosci Rep, vol.17, p.66, 2017.

M. Quadri, M. Fang, M. Picillo, S. Olgiati, G. J. Breedveld et al., Mutation in the SYNJ1 gene associated with autosomal recessive, early-onset Parkinsonism, Hum Mutat, vol.34, pp.1208-1215, 2013.

C. A. Ross and W. W. Smith, Gene-environment interactions in Parkinson's disease, Parkinsonism Relat Disord, vol.13, issue.3, pp.309-315, 2007.

E. M. Schmid, M. Ford, A. Burtey, G. Praefcke, S. Y. Peak-chew et al., Role of the AP2 ?appendage hub in recruiting partners for clathrin, 2006.

, PLoS Biol, vol.4, p.262

K. Soda, D. M. Balkin, S. M. Ferguson, S. Paradise, I. Milosevic et al., Role of dynamin, synaptojanin, and endophilin in podocyte foot processes, J Clin Invest, vol.122, pp.4401-4411, 2012.

S. Taghavi, R. Chaouni, A. Tafakhori, L. J. Azcona, S. G. Firouzabadi et al., A clinical and molecular genetic study of 50 families with autosomal recessive parkinsonism revealed known and novel gene mutations, Mol Neurobiol, vol.55, pp.3477-3489, 2018.

Y. Tsujishita, S. Guo, L. E. Stolz, J. D. York, and J. H. Hurley, Specificity determinants in phosphoinositide dephosphorylation: crystal struc-ture of an archetypal inositol polyphosphate 5-phosphatase, Cell, vol.105, pp.379-389, 2001.

J. Parkinson, An essay on the shaking palsy. 1817, J Neuropsychiatry Clin Neurosci, vol.14, pp.223-236, 2002.

J. Charcot and M. Jean, Bourneville b 1840 (1884) Leçons sur les maladies du système nerveux faites à la salpêtrière, A. Delahaye 3. Charcot J-M (1987) Charcot, the Clinician: The Tuesday Lessons: Excerpts from Nine Case Presentations on General Neurology Delivered at the Salpetriere Hospital in 1887-88

D. Hirtz, D. J. Thurman, and K. Gwinn-hardy, How common are the "common" neurologic disorders, Neurology, vol.68, pp.326-337, 2007.

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

L. Hirsch, N. Jette, and A. Frolkis, The Incidence of Parkinson's Disease: A Systematic Review and Meta-Analysis, NED, vol.46, pp.292-300, 2016.

R. Cacabelos, Parkinson's Disease: From Pathogenesis to Pharmacogenomics, Int J Mol Sci, vol.18, 2017.

K. Taylor, J. A. Cook, and C. E. Counsell, Heterogeneity in male to female risk for Parkinson's disease, J Neurol Neurosurg Psychiatry, vol.78, pp.905-906, 2007.

G. Wooten, L. Currie, and V. Bovbjerg, Are men at greater risk for Parkinson's disease than women?, J Neurol Neurosurg Psychiatry, vol.75, pp.637-639, 2004.

F. Moisan, S. Kab, and F. Mohamed, Parkinson disease male-to-female ratios increase with age: French nationwide study and meta-analysis, J Neurol Neurosurg Psychiatry, vol.87, pp.952-957, 2016.

M. D. Benedetti, D. M. Maraganore, and J. H. Bower, Hysterectomy, menopause, and estrogen use preceding Parkinson's disease: an exploratory case-control study, Mov Disord, vol.16, pp.830-837, 2001.

W. A. Rocca, J. H. Bower, and D. M. Maraganore, Increased risk of parkinsonism in women who underwent oophorectomy before menopause, Neuro, vol.70, pp.200-209, 2008.

Y. Alamri, M. Macaskill, T. Anderson, and H. Benamer, Parkinson's Disease in the Gulf Countries: An Updated Review, ENE, vol.74, pp.222-225, 2015.

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, Movement Disorders, vol.21, pp.2150-2156, 2006.

J. Blanckenberg, S. Bardien, and B. Glanzmann, The prevalence and genetics of Parkinson's disease in sub-Saharan Africans, Journal of the Neurological Sciences, vol.335, pp.22-25, 2013.

N. Attia-romdhane, B. Hamida, M. Mrabet, and A. , Prevalence study of neurologic disorders in Kelibia (Tunisia), Neuroepidemiology, vol.12, pp.285-299, 1993.

R. Mehanna, S. Moore, and J. G. Hou, Comparing clinical features of young onset, middle onset and late onset Parkinson's disease, Parkinsonism & Related Disorders, vol.20, pp.530-534, 2014.

L. Defebvre and M. Vérin, La maladie de Parkinson, 2e édition, 2011.

J. Jankovic, Parkinson's disease: clinical features and diagnosis, J Neurol Neurosurg Psychiatry, vol.79, pp.368-376, 2008.

, Gowers W (1893) A manual of diseases of the nervous system

H. Ling, L. A. Massey, and A. J. Lees, Hypokinesia without decrement distinguishes progressive supranuclear palsy from Parkinson's disease, Brain, vol.135, pp.1141-1153, 2012.

D. R. Williams, H. C. Watt, and A. J. Lees, Predictors of falls and fractures in bradykinetic rigid syndromes: a retrospective study, J Neurol Neurosurg Psychiatry, vol.77, pp.468-473, 2006.

M. Macht, Y. Kaussner, and J. C. Möller, Predictors of freezing in Parkinson's disease: a survey of 6,620 patients, Mov Disord, vol.22, pp.953-956, 2007.

R. F. Pfeiffer, Non-motor symptoms in Parkinson's disease, Parkinsonism Relat Disord, vol.22, pp.119-122, 2016.

S. Sveinbjornsdottir, The clinical symptoms of Parkinson's disease, J Neurochem, vol.139, pp.318-324, 2016.

A. Schapira, K. R. Chaudhuri, and P. Jenner, Non-motor features of Parkinson disease, Nature Reviews Neuroscience, vol.18, p.435, 2017.

A. Schrag, L. Horsfall, and K. Walters, Prediagnostic presentations of Parkinson's disease in primary care: a case-control study, Lancet Neurol, vol.14, pp.57-64, 2015.

H. Chen, E. A. Burton, and G. W. Ross, Research on the Premotor Symptoms of Parkinson's Disease: Clinical and Etiological Implications, Environmental Health Perspectives, vol.121, p.1245, 2013.

W. H. Jost, Autonomic dysfunctions in idiopathic Parkinson's disease, J Neurol, vol.250, pp.28-30, 2003.

H. Lahrmann, P. Cortelli, and M. Hilz, EFNS guidelines on the diagnosis and management of orthostatic hypotension, Eur J Neurol, vol.13, pp.930-936, 2006.

S. E. Mathers, P. A. Kempster, and P. J. Law, Anal sphincter dysfunction in Parkinson's disease, Arch Neurol, vol.46, pp.1061-1064, 1989.

P. Derkinderen, T. Rouaud, and T. Lebouvier, Parkinson disease: the enteric nervous system spills its guts, Neurology, vol.77, pp.1761-1767, 2011.

W. H. Jost, Gastrointestinal dysfunction in Parkinson's Disease, J Neurol Sci, vol.289, pp.69-73, 2010.

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

K. Winge, Lower urinary tract dysfunction in patients with parkinsonism and other neurodegenerative disorders, Handb Clin Neurol, vol.130, pp.335-356, 2015.

I. Araki, M. Kitahara, T. Oida, and S. Kuno, Voiding dysfunction and Parkinson's disease: urodynamic abnormalities and urinary symptoms, J Urol, vol.164, pp.1640-1643, 2000.

M. Hirayama, Sweating dysfunctions in Parkinson's disease, J Neurol, vol.253, pp.42-47, 2006.

M. Fischer, I. Gemende, W. C. Marsch, and P. A. Fischer, Skin function and skin disorders in Parkinson's disease, J Neural Transm, vol.108, pp.205-213, 2001.

M. G. Cersósimo, O. R. Tumilasci, and G. B. Raina, Hyposialorrhea as an early manifestation of Parkinson disease, Auton Neurosci, vol.150, pp.150-151, 2009.

J. Palma and H. Kaufmann, Autonomic disorders predicting Parkinson disease, Parkinsonism Relat Disord, vol.20, pp.94-98, 2014.

S. H. Mehta, J. C. Morgan, and K. D. Sethi, Sleep disorders associated with Parkinson's disease: role of dopamine, epidemiology, and clinical scales of assessment, CNS Spectr, vol.13, pp.6-11, 2008.

C. W. Olanow, A. H. Schapira, and T. Roth, Waking up to sleep episodes in Parkinson's disease, Mov Disord, vol.15, pp.212-215, 2000.

J. P. Larsen and E. Tandberg, Sleep disorders in patients with Parkinson's disease: epidemiology and management, CNS Drugs, vol.15, pp.267-275, 2001.

M. Louter, M. A. Van-der-marck, D. Pevernagie, and .. A. , Sleep matters in Parkinson's disease: use of a priority list to assess the presence of sleep disturbances, Eur J Neurol, vol.20, pp.259-265, 2013.

R. Monderer and M. Thorpy, Sleep disorders and daytime sleepiness in Parkinson's disease, Curr Neurol Neurosci Rep, vol.9, pp.173-180, 2009.

B. Knie, M. T. Mitra, K. Logishetty, and K. R. Chaudhuri, Excessive daytime sleepiness in patients with Parkinson's disease, CNS Drugs, vol.25, pp.203-212, 2011.

H. Pfeiffer, A. Løkkegaard, and M. Zoetmulder, Cognitive impairment in early-stage non-demented Parkinson's disease patients, Acta Neurologica Scandinavica, vol.129, pp.307-318, 2014.

A. A. Kehagia, R. A. Barker, and T. W. Robbins, Cognitive Impairment in Parkinson's Disease: The Dual Syndrome Hypothesis, Neurodegener Dis, vol.11, pp.79-92, 2012.

V. S. Mattay, A. Tessitore, and J. H. Callicott, Dopaminergic modulation of cortical function in patients with Parkinson's disease, Ann Neurol, vol.51, pp.156-164, 2002.

D. Aarsland, K. Andersen, and J. P. Larsen, Prevalence and characteristics of dementia in Parkinson disease: an 8-year prospective study, Arch Neurol, vol.60, pp.387-392, 2003.

D. J. Irwin, M. T. White, and J. B. Toledo, Neuropathologic substrates of Parkinson's disease dementia, Ann Neurol, vol.72, pp.587-598, 2012.

G. Alves, J. P. Larsen, and M. Emre, Changes in motor subtype and risk for incident dementia in Parkinson's disease, Mov Disord, vol.21, pp.1123-1130, 2006.

J. Wang, H. You, and J. Liu, Association of Olfactory Bulb Volume and Olfactory Sulcus Depth with Olfactory Function in Patients with Parkinson Disease, American Journal of Neuroradiology, vol.32, pp.677-681, 2011.

N. K. Archibald, M. P. Clarke, U. P. Mosimann, and D. J. Burn, Visual symptoms in Parkinson's disease and Parkinson's disease dementia, Mov Disord, vol.26, pp.2387-2395, 2011.

W. L. Baker, D. Silver, and C. M. White, Dopamine agonists in the treatment of early Parkinson's disease: A meta-analysis, Parkinsonism & Related Disorders, vol.15, pp.287-294, 2009.

G. Fénelon, F. Mahieux, R. Huon, and M. Ziégler, Hallucinations in Parkinson's disease: prevalence, phenomenology and risk factors, Brain, vol.123, pp.733-745, 2000.

A. J. Muller, J. M. Shine, G. M. Halliday, and S. Lewis, Visual hallucinations in Parkinson's disease: theoretical models, Mov Disord, vol.29, pp.1591-1598, 2014.

N. K. Archibald, S. B. Hutton, and M. P. Clarke, Visual exploration in Parkinson's disease and Parkinson's disease dementia, Brain, vol.136, pp.739-750, 2013.

A. G. Beiske, J. H. Loge, A. Rønningen, and E. Svensson, Pain in Parkinson's disease: Prevalence and characteristics, PAIN®, vol.141, pp.173-177, 2009.

Z. Rihmer, X. Gonda, and P. Döme, Depression in Parkinson's disease, Ideggyogy Sz, vol.67, pp.229-236, 2014.

L. Tan, Mood disorders in Parkinson's disease, Parkinsonism & Related Disorders, vol.18, issue.11, pp.70024-70028, 2012.

J. L. Fontoura, C. Baptista, and B. Pedroso-f-de, Depression in Parkinson's Disease: The Contribution from Animal Studies, Parkinsons Dis, 2017.

D. J. Burn, Beyond the iron mask: towards better recognition and treatment of depression associated with Parkinson's disease, Mov Disord, vol.17, pp.445-454, 2002.

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

C. G. Goetz, B. C. Tilley, and S. R. Shaftman, Movement Disorder Societysponsored revision of the Unified Parkinson's Disease Rating Scale (MDS-UPDRS): scale presentation and clinimetric testing results, Mov Disord, vol.23, pp.2129-2170, 2008.

R. B. Postuma, D. Berg, and M. Stern, MDS clinical diagnostic criteria for Parkinson's disease, Mov Disord, vol.30, pp.1591-1601, 2015.

S. Fahn and R. Elton, The Unified Parkinson's Disease Rating Scale, Recent Developments in Parkinson's Disease, pp.153-163, 1987.

C. G. Goetz, S. Fahn, and P. Martinez-martin, Movement Disorder Societysponsored revision of the Unified Parkinson's Disease Rating Scale (MDS-UPDRS): Process, format, and clinimetric testing plan, Mov Disord, vol.22, pp.41-47, 2007.

M. M. Hoehn and M. D. Yahr, Parkinsonism: onset, progression and mortality, Neurology, vol.17, pp.427-442, 1967.

A. C. England and R. S. Schwab, Postoperative medical evaluation of 26 selected patients with Parkinson's disease, J Am Geriatr Soc, vol.4, pp.1219-1232, 1956.

W. Guy, ECDEU assessment manual for psychopharmacology, Rev. 1976. U.S. Dept. of Health, Education, and Welfare, Public Health Service, Alcohol, Drug Abuse, and Mental Health Administration, 1976.

M. F. Folstein, S. E. Folstein, and P. R. Mchugh, A practical method for grading the cognitive state of patients for the clinician, J Psychiatr Res, vol.12, pp.189-198, 1975.

R. M. Crum, J. C. Anthony, S. S. Bassett, and M. F. Folstein, Population-based norms for the Mini-Mental State Examination by age and educational level, JAMA, vol.269, pp.2386-2391, 1993.

Z. S. Nasreddine, N. A. Phillips, and V. Bédirian, MoCA: a brief screening tool for mild cognitive impairment, The Montreal Cognitive Assessment, vol.53, pp.695-699, 2005.

B. Dubois, A. Slachevsky, I. Litvan, and B. Pillon, The FAB: a Frontal Assessment Battery at bedside, Neurology, vol.55, pp.1621-1626, 2000.

J. A. Yesavage, T. L. Brink, and T. L. Rose, Development and validation of a geriatric depression screening scale: a preliminary report, J Psychiatr Res, vol.17, pp.37-49, 1982.

A. T. Beck, R. A. Steer, R. Ball, and W. Ranieri, Comparison of Beck Depression Inventories -IA and -II in psychiatric outpatients, J Pers Assess, vol.67, pp.588-597, 1996.

J. E. Ware and C. D. Sherbourne, The MOS 36-item short-form health survey, 1992.

I. , Conceptual framework and item selection, Med Care, vol.30, pp.473-483

V. Peto, C. Jenkinson, R. Fitzpatrick, and R. Greenhall, The development and validation of a short measure of functioning and well being for individuals with Parkinson's disease, Qual Life Res, vol.4, pp.241-248, 1995.

C. Foix and J. Nicolesco, Anatomie cérébrale. Les noyaux gris centraux et la région mesencephalo-sous-optique ; suivi d'une appendice sur l'anatomie pathologique de la maladie de Parkinson, 1925.

J. Yelnik, C. François, G. Percheron, and D. Tandé, Morphological taxonomy of the neurons of the primate striatum, J Comp Neurol, vol.313, pp.273-294, 1991.

J. B. Penney and A. B. Young, GABA as the pallidothalamic neurotransmitter: implications for basal ganglia function, Brain Res, vol.207, pp.195-199, 1981.

E. Rinvik and O. P. Ottersen, Terminals of subthalamonigral fibres are enriched with glutamate-like immunoreactivity: an electron microscopic, immunogold analysis in the cat, J Chem Neuroanat, vol.6, pp.19-30, 1993.

G. E. Alexander, M. R. Delong, and P. L. Strick, Parallel organization of functionally segregated circuits linking basal ganglia and cortex, Annu Rev Neurosci, vol.9, pp.357-381, 1986.

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.

G. E. Alexander, M. D. Crutcher, and M. R. Delong, Basal ganglia-thalamocortical circuits: parallel substrates for motor, oculomotor, "prefrontal" and "limbic" functions, Prog Brain Res, vol.85, pp.119-146, 1990.

J. Viallet, D. Gayraud, and E. Bonnefoi-kyriacou, Maladie de Parkinson idiopathique : aspects cliniques, diagnostiques et thérapeutiques, Encycl Med Chir Elsevier SAS, 2001.

R. Levy, L. Hazrati, and M. Herrero, Re-evaluation of the functional anatomy of the basal ganglia in normal and Parkinsonian states, Neuroscience, vol.76, pp.335-343, 1997.

D. W. Dickson, Neuropathology of Parkinson disease, Parkinsonism & Related Disorders, vol.46, pp.30-33, 2018.

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

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

O. Hornykiewicz, Biochemical aspects of Parkinson's disease, Neurology, vol.51, pp.2-9, 1998.

D. C. German, K. Manaye, and W. K. Smith, Midbrain dopaminergic cell loss in Parkinson's disease: computer visualization, Ann Neurol, vol.26, pp.507-514, 1989.

P. Damier, E. C. Hirsch, Y. Agid, and A. M. Graybiel, The substantia nigra of the human brain. II. Patterns of loss of dopamine-containing neurons in Parkinson's disease, Brain, vol.122, pp.1437-1448, 1999.

H. Cheng, C. M. Ulane, and R. E. Burke, Clinical progression in Parkinson disease and the neurobiology of axons, Ann Neurol, vol.67, pp.715-725, 2010.

C. Tretiakoff, Contributions a l'etude de l'anatomie pathologique du locus niger de soemmering avec quelques deductions relatives a la pathogenie des troubles de tonus musculaire et de la maladie de Parkinson, 1919.

C. Zarow, S. A. Lyness, J. A. Mortimer, and H. C. Chui, Neuronal loss is greater in the locus coeruleus than nucleus basalis and substantia nigra in Alzheimer and Parkinson diseases, Arch Neurol, vol.60, pp.337-341, 2003.

W. P. Gai, P. C. Blumbergs, L. B. Geffen, and W. W. Blessing, Age-related loss of dorsal vagal neurons in Parkinson's disease, Neurology, vol.42, pp.2106-2111, 1992.

E. C. Hirsch, A. M. Graybiel, C. Duyckaerts, and F. Javoy-agid, Neuronal loss in the pedunculopontine tegmental nucleus in Parkinson disease and in progressive supranuclear palsy, Proc Natl Acad Sci, vol.84, pp.5976-5980, 1987.

K. A. Jellinger, Pathology of Parkinson's disease. Changes other than the nigrostriatal pathway, Mol Chem Neuropathol, vol.14, pp.153-197, 1991.

M. G. Cersosimo and E. E. Benarroch, Autonomic involvement in Parkinson's disease: pathology, pathophysiology, clinical features and possible peripheral biomarkers, J Neurol Sci, vol.313, pp.57-63, 2012.

S. Orimo, T. Uchihara, and A. Nakamura, Axonal alpha-synuclein aggregates herald centripetal degeneration of cardiac sympathetic nerve in Parkinson's disease, Brain, vol.131, pp.642-650, 2008.

R. K. Pearce, C. H. Hawkes, and S. E. Daniel, The anterior olfactory nucleus in Parkinson's disease, Mov Disord, vol.10, pp.283-287, 1995.

S. Greffard, M. Verny, and A. Bonnet, 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.

K. Jellinger, Neuropathological substrates of Alzheimer's disease and Parkinson's disease, J Neural Transm Suppl, vol.24, pp.109-129, 1987.

, lewy FH (1912) Paralysis agitans. 1. Pathologische Anatomie, pp.920-933

M. G. Spillantini, M. L. Schmidt, and V. M. Lee, Alpha-synuclein in Lewy bodies, Nature, vol.388, pp.839-840, 1997.

H. Fujishiro, T. J. Ferman, and B. F. Boeve, Validation of the Neuropathologic Criteria of the Third Consortium for Dementia with Lewy Bodies for Prospectively Diagnosed Cases, J Neuropathol Exp Neurol, vol.67, pp.649-656, 2008.

G. E. Barreto, A. Iarkov, and V. E. Moran, Beneficial effects of nicotine, cotinine and its metabolites as potential agents for Parkinson's disease, Front Aging Neurosci, vol.6, 2015.

C. A. Ross and W. W. Smith, Gene-environment interactions in Parkinson's disease, Parkinsonism Relat Disord, vol.13, issue.08, pp.70022-70023, 2007.

A. Ascherio and M. A. Schwarzschild, The epidemiology of Parkinson's disease: risk factors and prevention, The Lancet Neurology, vol.15, pp.1257-1272, 2016.

K. Kieburtz and K. B. Wunderle, Parkinson's disease: Evidence for environmental risk factors, Mov Disord, vol.28, pp.8-13, 2013.

J. W. Langston, P. Ballard, J. W. Tetrud, and I. Irwin, Chronic Parkinsonism in humans due to a product of meperidine-analog synthesis, Science, vol.219, pp.979-980, 1983.

T. B. Sherer, J. H. Kim, R. Betarbet, and J. T. Greenamyre, Subcutaneous rotenone exposure causes highly selective dopaminergic degeneration and alpha-synuclein aggregation, Exp Neurol, vol.179, pp.9-16, 2003.

C. M. Tanner, G. W. Ross, and S. A. Jewell, Occupation and risk of parkinsonism: a multicenter case-control study, Arch Neurol, vol.66, pp.1106-1113, 2009.

,

C. M. Tanner, F. Kamel, and G. W. Ross, Rotenone, paraquat, and Parkinson's disease, Environ Health Perspect, vol.119, pp.866-872, 2011.

S. Costello, M. Cockburn, and J. Bronstein, Parkinson's Disease and Residential Exposure to Maneb and Paraquat From Agricultural Applications in the Central Valley of California, Am J Epidemiol, vol.169, pp.919-926, 2009.

K. Curtin, A. E. Fleckenstein, and R. J. Robison, , 2015.

, Methamphetamine/amphetamine abuse and risk of Parkinson's disease in Utah: A population-based assessment, Drug and Alcohol Dependence, vol.146, pp.30-38

T. R. Guilarte, M. K. Nihei, J. L. Mcglothan, and A. S. Howard, Methamphetamineinduced deficits of brain monoaminergic neuronal markers: distal axotomy or neuronal plasticity, Neuroscience, vol.122, pp.499-513, 2003.

S. M. Goldman, C. M. Tanner, and D. Oakes, Head injury and Parkinson's disease risk in twins, Ann Neurol, vol.60, pp.65-72, 2006.

C. Marras, C. A. Hincapié, and V. L. Kristman, Systematic Review of the Risk of Parkinson's Disease After Mild Traumatic Brain Injury: Results of the International Collaboration on Mild Traumatic Brain Injury Prognosis, Archives of Physical Medicine and Rehabilitation, vol.95, pp.238-244, 2014.

M. A. Hernán, B. Takkouche, F. Caamaño-isorna, and J. J. Gestal-otero, A metaanalysis of coffee drinking, cigarette smoking, and the risk of Parkinson's disease, 2002.

, Ann Neurol, vol.52, pp.276-284

A. Kachroo, M. C. Irizarry, and M. A. Schwarzschild, Caffeine protects against combined paraquat and maneb-induced dopaminergic neuron degeneration, Experimental Neurology, vol.223, pp.657-661, 2010.

K. Xu, Y. Xu, J. Chen, and M. A. Schwarzschild, Neuroprotection by caffeine: time course and role of its metabolites in the MPTP model of Parkinson's disease, Neuroscience, vol.167, pp.475-481, 2010.

M. Kitagawa, H. Houzen, and K. Tashiro, Caffeine in Parkinson's disease: Comment on its importance and the dose proposal, Mov Disord, vol.27, pp.808-808, 2012.

R. B. Postuma, A. E. Lang, and R. P. Munhoz, Caffeine for treatment of Parkinson disease, Neurology, vol.79, pp.651-658, 2012.

M. G. Weisskopf, E. O'reilly, and H. Chen, Plasma urate and risk of Parkinson's disease, Am J Epidemiol, vol.166, pp.561-567, 2007.

M. A. Schwarzschild, S. R. Schwid, and K. Marek, Serum urate as a predictor of clinical and radiographic progression in Parkinson disease, Arch Neurol, vol.65, pp.716-723, 2008.

S. M. Zhang, M. A. Hernán, and H. Chen, Intakes of vitamins E and C, carotenoids, vitamin supplements, and PD risk, Neurology, vol.59, pp.1161-1169, 2002.

C. D. Marsden, Parkinson's disease in twins, J Neurol Neurosurg Psychiatry, vol.50, pp.105-106, 1987.

R. J. Marttila, J. Kaprio, M. Koskenvuo, and U. K. Rinne, Parkinson's disease in a nationwide twin cohort, Neurology, vol.38, pp.1217-1219, 1988.

C. D. Ward, R. C. Duvoisin, and S. E. Ince, Parkinson's disease in 65 pairs of twins and in a set of quadruplets, Neurology, vol.33, pp.815-824, 1983.

W. G. Johnson, S. E. Hodge, and R. Duvoisin, Twin studies and the genetics of Parkinson's disease--a reappraisal, Mov Disord, vol.5, pp.187-194, 1990.

C. M. Tanner, R. Ottman, and S. M. Goldman, Parkinson disease in twins: an etiologic study, JAMA, vol.281, pp.341-346, 1999.

A. M. Lazzarini, R. H. Myers, and T. R. Zimmerman, A clinical genetic study of Parkinson's disease: evidence for dominant transmission, Neurology, vol.44, pp.499-506, 1994.

M. H. Polymeropoulos, J. J. Higgins, and L. I. Golbe, Mapping of a Gene for Parkinson's Disease to Chromosome 4q21-q23, Science, vol.274, pp.1197-1199, 1996.

M. H. Polymeropoulos, C. Lavedan, and E. Leroy, Mutation in the ?-Synuclein Gene Identified in Families with Parkinson's Disease, Science, vol.276, pp.2045-2047, 1997.

H. Houlden and A. B. Singleton, The genetics and neuropathology of Parkinson's disease, Acta Neuropathol, vol.124, pp.325-338, 2012.

A. Puschmann, Monogenic Parkinson's disease and parkinsonism: clinical phenotypes and frequencies of known mutations, Parkinsonism Relat Disord, vol.19, pp.407-415, 2013.

A. Puschmann, New Genes Causing Hereditary Parkinson's Disease or Parkinsonism, Curr Neurol Neurosci Rep, vol.17, p.66, 2017.

H. Deng, P. Wang, and J. Jankovic, The genetics of Parkinson disease, Ageing Research Reviews, vol.42, pp.72-85, 2018.

R. Alcalay, E. Caccappolo, and H. Mejia-santana, Frequency of known mutations in early onset PD; implication for genetic counseling: the CORE-PD study, Arch Neurol, vol.67, pp.1116-1122, 2010.

L. L. Kilarski, J. P. Pearson, and V. Newsway, Systematic Review and UK-Based Study of PARK2 (parkin), PINK1, PARK7 (DJ-1) and LRRK2 in early-onset Parkinson's disease, Mov Disord, vol.27, pp.1522-1529, 2012.

S. Lesage and A. Brice, Parkinson's disease: from monogenic forms to genetic susceptibility factors, Hum Mol Genet, vol.18, pp.48-59, 2009.

D. Crosiers, J. Theuns, P. Cras, and C. Van-broeckhoven, Parkinson disease: insights in clinical, genetic and pathological features of monogenic disease subtypes, J Chem Neuroanat, vol.42, pp.131-141, 2011.

J. W. Touchman, A. Dehejia, and O. Chiba-falek, Human and Mouse ?-Synuclein Genes: Comparative Genomic Sequence Analysis and Identification of a Novel Gene Regulatory Element, Genome Res, vol.11, pp.78-86, 2001.

R. Jakes, M. G. Spillantini, and M. Goedert, Identification of two distinct synucleins from human brain, FEBS Lett, vol.345, pp.27-32, 1994.

S. Liu, I. Ninan, and I. Antonova, ) ?-Synuclein produces a long-lasting increase in neurotransmitter release, The EMBO Journal, vol.23, pp.4506-4516, 2004.

A. Abeliovich, Y. Schmitz, and I. Fariñas, Mice Lacking ?-Synuclein Display Functional Deficits in the Nigrostriatal Dopamine System, Neuron, vol.25, pp.239-252, 2000.

K. Nakamura, V. M. Nemani, and F. Azarbal, Direct Membrane Association Drives Mitochondrial Fission by the Parkinson Disease-associated Protein ?-Synuclein, J Biol Chem, vol.286, pp.20710-20726, 2011.

A. A. Cooper, A. D. Gitler, and A. Cashikar, ) ?-Synuclein Blocks ER-Golgi Traffic and Rab1 Rescues Neuron Loss in Parkinson's Models, Science, vol.313, pp.324-328, 2006.

L. Stefanis, K. E. Larsen, and H. J. Rideout, Expression of A53T Mutant But Not Wild-Type ?-Synuclein in PC12 Cells Induces Alterations of the Ubiquitin-Dependent Degradation System, Loss of Dopamine Release, and Autophagic Cell Death, J Neurosci, vol.21, pp.9549-9560, 2001.

R. Krüger, W. Kuhn, and T. Müller, AlaSOPro mutation in the gene encoding ?-synuclein in Parkinson's disease, Nature Genetics, vol.18, pp.106-108, 1998.

J. J. Zarranz, J. Alegre, and J. C. Gómez-esteban, The new mutation, E46K, of ?synuclein causes parkinson and Lewy body dementia, Ann Neurol, vol.55, pp.164-173, 2004.

P. Ibáñez, S. Lesage, and S. Janin, ) ?-Synuclein Gene Rearrangements in Dominantly Inherited Parkinsonism: Frequency, Phenotype, and Mechanisms, Arch Neurol, vol.66, pp.102-108, 2009.

A. Zimprich, S. Biskup, and P. Leitner, Mutations in LRRK2 Cause Autosomal-Dominant Parkinsonism with Pleomorphic Pathology, Neuron, vol.44, pp.601-607, 2004.

C. Paisán-ru??, S. Jain, and E. W. Evans, Cloning of the Gene Containing Mutations that Cause PARK8-Linked Parkinson's Disease, Neuron, vol.44, pp.595-600, 2004.

L. Bosgraaf and P. Van-haastert, Roc, a Ras/GTPase domain in complex proteins, Biochimica et Biophysica Acta (BBA) -Molecular Cell Research, vol.1643, pp.5-10, 2003.

D. Macleod, J. Dowman, and R. Hammond, The Familial Parkinsonism Gene LRRK2 Regulates Neurite Process Morphology, Neuron, vol.52, pp.587-593, 2006.

G. Piccoli, S. B. Condliffe, and M. Bauer, LRRK2 controls synaptic vesicle storage and mobilization within the recycling pool, J Neurosci, vol.31, pp.2225-2237, 2011.

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

N. Shin, H. Jeong, and J. Kwon, LRRK2 regulates synaptic vesicle endocytosis, Exp Cell Res, vol.314, pp.2055-2065, 2008.

W. W. Smith, Z. Pei, and H. Jiang, Leucine-rich repeat kinase 2 (LRRK2) interacts with parkin, and mutant LRRK2 induces neuronal degeneration, PNAS, vol.102, pp.18676-18681, 2005.

H. Qing, W. Wong, E. G. Mcgeer, and P. L. Mcgeer, Lrrk2 phosphorylates alpha synuclein at serine 129: Parkinson disease implications, Biochem Biophys Res Commun, vol.387, pp.149-152, 2009.

A. R. Esteves, R. H. Swerdlow, and S. M. Cardoso, LRRK2, a puzzling protein: insights into Parkinson's disease pathogenesis, Exp Neurol, vol.0, pp.206-216, 2014.

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

J. C. Dächsel, M. J. Farrer, and P. Disease, Arch Neurol, vol.67, pp.542-547, 2010.

I. F. Mata, C. M. Hutter, and M. C. González-fernández, Lrrk2 R1441G-related Parkinson's disease: evidence of a common founding event in the seventh century in Northern Spain, Neurogenetics, vol.10, p.347, 2009.

S. Lesage, A. Dürr, and M. Tazir, LRRK2 G2019S as a Cause of Parkinson's Disease in North African Arabs, New England Journal of Medicine, vol.354, pp.422-423, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00221453

A. Orr-urtreger, C. Shifrin, and U. Rozovski, The LRRK2 G2019S mutation in Ashkenazi Jews with Parkinson disease: is there a gender effect, Neurology, vol.69, pp.1595-1602, 2007.

S. Lesage, P. E. Condroyer, and C. , Parkinson's disease-related LRRK2 G2019S mutation results from independent mutational events in humans, Hum Mol Genet, vol.19, 1998.
URL : https://hal.archives-ouvertes.fr/inserm-00522011

R. N. Alcalay, H. Mejia-santana, and M. X. Tang, Motor phenotype of LRRK2 G2019S carriers in early-onset Parkinson disease, Arch Neurol, vol.66, pp.1517-1522, 2009.

F. Nabli, B. Sassi, S. Amouri, and R. , Motor phenotype of LRRK2-associated Parkinson's disease: a Tunisian longitudinal study, Mov Disord, vol.30, pp.253-258, 2015.

K. Nishioka, M. Kefi, and B. Jasinska-myga, A comparative study of LRRK2, PINK1 and genetically undefined familial Parkinson's disease, J Neurol Neurosurg Psychiatry, vol.81, pp.391-395, 2010.

P. Zhang, L. Yu, and J. Gao, Cloning and Characterization of Human VPS35 and Mouse Vps35 and Mapping of VPS35 to Human Chromosome 16q13-q21, Genomics, vol.70, pp.253-257, 2000.

A. J. Edgar and J. M. Polak, Human Homologues of Yeast Vacuolar Protein Sorting 29 and 35, Biochemical and Biophysical Research Communications, vol.277, pp.622-630, 2000.

C. Vilariño-güell, C. Wider, and O. A. Ross, VPS35 Mutations in Parkinson Disease, The American Journal of Human Genetics, vol.89, pp.162-167, 2011.

A. Zimprich, A. Benet-pagès, and W. Struhal, A Mutation in VPS35, Encoding a Subunit of the Retromer Complex, Causes Late-Onset Parkinson Disease, The American Journal of Human Genetics, vol.89, pp.168-175, 2011.

M. Ando, M. Funayama, and Y. Li, VPS35 mutation in Japanese patients with typical Parkinson's disease, Mov Disord, vol.27, pp.1413-1417, 2012.

S. Lesage, C. Condroyer, and S. Klebe, Identification of VPS35 mutations replicated in French families with Parkinson disease, Neurology, vol.78, pp.1449-1450, 2012.
URL : https://hal.archives-ouvertes.fr/inserm-00807463

K. R. Kumar, A. Weissbach, and M. Heldmann, Frequency of the D620N Mutation in VPS35 in Parkinson Disease, Arch Neurol, vol.69, pp.1360-1364, 2012.

M. Horowitz, S. Wilder, and Z. Horowitz, The human glucocerebrosidase gene and pseudogene: Structure and evolution, Genomics, vol.4, pp.87-96, 1989.

E. Beutler, Gaucher disease: new molecular approaches to diagnosis and treatment, Science, vol.256, pp.794-799, 1992.

H. Dvir, M. Harel, and A. A. Mccarthy, X-ray structure of human acid-?glucosidase, the defective enzyme in Gaucher disease, EMBO Rep, vol.4, pp.704-709, 2003.

E. Beutler, G. ;. Grabowski, W. S. Sly, B. Childs, A. L. Beaudet et al., Glucosylceramide lipidosis-Gaucher disease, The Metabolic and Molecular Bases of Inherited Disease. Scriver, pp.3635-3668, 2001.

S. Tsuji, P. V. Choudary, and B. M. Martin, A mutation in the human glucocerebrosidase gene in neuronopathic Gaucher's disease, N Engl J Med, vol.316, pp.570-575, 1987.

P. Giraldo, J. L. Capablo, and P. Alfonso, Neurological manifestations in patients with Gaucher disease and their relatives, it is just a coincidence?, J Inherit Metab Dis, vol.34, pp.781-787, 2011.

A. Halperin, D. Elstein, and A. Zimran, Increased incidence of Parkinson disease among relatives of patients with Gaucher disease, Molecules, and Diseases, vol.36, pp.426-428, 2006.

O. Neudorfer, N. Giladi, and D. Elstein, Occurrence of Parkinson's syndrome in type I Gaucher disease, QJM, vol.89, pp.691-694, 1996.

N. Tayebi, J. Walker, and B. Stubblefield, Gaucher disease with parkinsonian manifestations: does glucocerebrosidase deficiency contribute to a vulnerability to parkinsonism?, Molecular Genetics and Metabolism, vol.79, pp.104-109, 2003.

O. Goker-alpan, G. Lopez, and J. Vithayathil, The Spectrum of Parkinsonian Manifestations Associated With Glucocerebrosidase Mutations, Arch Neurol, vol.65, pp.1353-1357, 2008.

S. Lesage, M. Anheim, and C. Condroyer, Large-scale screening of the Gaucher's disease-related glucocerebrosidase gene in Europeans with Parkinson's disease, Hum Mol Genet, vol.20, pp.202-210, 2011.

W. C. Nichols, N. Pankratz, and D. K. Marek, Mutations in GBA are associated with familial Parkinson disease susceptibility and age at onset, Neurology, vol.72, pp.310-316, 2009.

M. E. Gegg and A. Schapira, The role of glucocerebrosidase in Parkinson disease pathogenesis, FEBS J Epub ahead of print, 2018.

S. Jesús, I. Huertas, and I. Bernal-bernal, GBA Variants Influence Motor and Non-Motor Features of Parkinson's Disease, PLoS ONE, vol.11, p.167749, 2016.

V. Lythe, D. Athauda, and J. Foley, GBA-Associated Parkinson's Disease: Progression in a Deep Brain Stimulation Cohort, J Parkinsons Dis, vol.7, pp.635-644, 2017.

K. A. Senkevich, I. V. Miliukhina, and M. V. Beletskaia, The clinical features of Parkinson's disease in patients with mutations and polymorphic variants of GBA gene, 2017.

, Zh Nevrol Psikhiatr Im S S Korsakova, vol.117, pp.81-86

S. E. Winder-rhodes, J. R. Evans, and M. Ban, Glucocerebrosidase mutations influence the natural history of Parkinson's disease in a community-based incident cohort, Brain, vol.136, pp.392-399, 2013.

C. Lu, H. Chang, and P. Kuo, The parkinsonian phenotype of spinocerebellar ataxia type 3 in a Taiwanese family, Parkinsonism Relat Disord, vol.10, pp.369-373, 2004.

M. P. Socal, V. E. Emmel, and C. Rieder, Intrafamilial variability of Parkinson phenotype in SCAs: novel cases due to SCA2 and SCA3 expansions, Parkinsonism Relat Disord, vol.15, pp.374-378, 2009.

P. Charles, A. Camuzat, and N. Benammar, Are interrupted SCA2 CAG repeat expansions responsible for parkinsonism, Neurology, vol.69, pp.1970-1975, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00221466

M. Sharma, J. C. Mueller, and A. Zimprich, The sepiapterin reductase gene region reveals association in the PARK3 locus: analysis of familial and sporadic Parkinson's disease in European populations, J Med Genet, vol.43, pp.557-562, 2006.

M. Sharma, D. M. Maraganore, and J. Ioannidis, Role of sepiapterin reductase gene at the PARK3 locus in Parkinson's disease, Neurobiol Aging, vol.32, 2011.

E. Leroy, R. Boyer, and G. Auburger, The ubiquitin pathway in Parkinson's disease, Nature, vol.395, pp.451-452, 1998.

C. Lautier, S. Goldwurm, and A. Dürr, Mutations in the GIGYF2 (TNRC15) Gene at the PARK11 Locus in Familial Parkinson Disease, The American Journal of Human Genetics, vol.82, pp.822-833, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00280725

M. Bonetti, A. Ferraris, and M. Petracca, GIGYF2 variants are not associated with Parkinson's disease in Italy, Mov Disord, vol.24, pp.1867-1868, 2009.

D. Fonzo, A. Fabrizio, E. Thomas, and A. , GIGYF2 mutations are not a frequent cause of familial Parkinson's disease, Parkinsonism & Related Disorders, vol.15, pp.703-705, 2009.

W. C. Nichols, D. K. Kissell, and N. Pankratz, Variation in GIGYF2 is not associated with Parkinson disease, Neurology, vol.72, pp.1886-1892, 2009.

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

R. Krüger, M. Sharma, and O. Riess, A large-scale genetic association study to evaluate the contribution of Omi/HtrA2 (PARK13) to Parkinson's disease, Neurobiol Aging, vol.32, pp.548-557, 2011.

O. A. Ross, A. I. Soto, and C. Vilariño-güell, Genetic variation of Omi/HtrA2 and Parkinson's disease, Parkinsonism & Related Disorders, vol.14, pp.539-543, 2008.

V. Bogaerts, K. Nuytemans, and J. Reumers, Genetic variability in the mitochondrial serine protease HTRA2 contributes to risk for Parkinson disease, Hum Mutat, vol.29, pp.832-840, 2008.

K. Momma, M. Funayama, and Y. Li, A new mutation in the GCH1 gene presents as early-onset Parkinsonism, Parkinsonism & Related Disorders, vol.15, pp.160-161, 2009.

M. Baker, I. R. Mackenzie, and S. M. Pickering-brown, Mutations in progranulin cause tau-negative frontotemporal dementia linked to chromosome 17, Nature, vol.442, pp.916-919, 2006.

B. J. Kelley, W. Haidar, and B. F. Boeve, Prominent phenotypic variability associated with mutations in Progranulin, Neurobiology of Aging, vol.30, pp.739-751, 2009.

M. Quadri, G. Cossu, and V. Saddi, Broadening the phenotype of <Emphasis Type="Italic">TARDBP</Emphasis> mutations: the <Emphasis Type="Italic">TARDBP</Emphasis> Ala382Thr mutation and Parkinson's disease in Sardinia, Neurogenetics, vol.12, pp.203-209, 2011.

M. Hutton, C. L. Lendon, and P. Rizzu, Association of missense and 5?-splicesite mutations in tau with the inherited dementia FTDP-17, Nature, vol.393, pp.702-705, 1998.

E. Naro?a?ska, B. Jasi?ska-myga, and E. J. Sitek, Frontotemporal dementia and parkinsonism linked to chromosome 17 -the first Polish family, European Journal of Neurology, vol.18, pp.535-537, 2011.

J. Van-swieten and M. G. Spillantini, Hereditary Frontotemporal Dementia Caused by Tau Gene Mutations, Brain Pathology, vol.17, pp.63-73, 2007.

G. Davidzon, P. Greene, and M. Mancuso, Early-onset familial parkinsonism due to POLG mutations, Ann Neurol, vol.59, pp.859-862, 2006.

G. Hudson, A. M. Schaefer, and R. W. Taylor, Mutation of the Linker Region of the Polymerase ?-1 (POLG1Gene Associated With Progressive External Ophthalmoplegia and Parkinsonism, Arch Neurol, vol.64, pp.553-557, 2007.

A. M. Remes, R. Hinttala, and M. Kärppä, Parkinsonism associated with the homozygous W748S mutation in the POLG1 gene, Parkinsonism & Related Disorders, vol.14, pp.652-654, 2008.

M. Chartier-harlin, J. C. Dachsel, and C. Vilariño-güell, Translation Initiator EIF4G1 Mutations in Familial Parkinson Disease, The American Journal of Human Genetics, vol.89, pp.398-406, 2011.

S. Asakawa, K. Tsunematsu, and A. Takayanagi, The Genomic Structure and Promoter Region of the Human Parkin Gene, Biochemical and Biophysical Research Communications, vol.286, pp.863-868, 2001.

S. R. Yoshii, C. Kishi, N. Ishihara, and N. Mizushima, Parkin Mediates Proteasomedependent Protein Degradation and Rupture of the Outer Mitochondrial Membrane, J Biol Chem, vol.286, pp.19630-19640, 2011.

N. C. Chan and D. C. Chan, Parkin uses the UPS to ship off dysfunctional mitochondria, Autophagy, vol.7, pp.771-772, 2011.

T. Kitada, S. Asakawa, and N. Hattori, Mutations in the parkin gene cause autosomal recessive juvenile parkinsonism, Nature, vol.392, pp.605-608, 1998.

C. B. Lücking, A. Dürr, and V. Bonifati, Association between early-onset Parkinson's disease and mutations in the parkin gene, N Engl J Med, vol.342, pp.1560-1567, 2000.

M. R. Cookson, P. J. Lockhart, and C. Mclendon, RING finger 1 mutations in Parkin produce altered localization of the protein, Hum Mol Genet, vol.12, pp.2957-2965, 2003.

W. Gu, O. Corti, and F. Araujo, The C289G and C418R missense mutations cause rapid sequestration of human Parkin into insoluble aggregates, Neurobiol Dis, vol.14, pp.357-364, 2003.

C. Wang, J. Tan, and M. Ho, Alterations in the solubility and intracellular localization of parkin by several familial Parkinson's disease-linked point mutations, J Neurochem, vol.93, pp.422-431, 2005.

J. J. Palacino, D. Sagi, and M. S. Goldberg, Mitochondrial dysfunction and oxidative damage in parkin-deficient mice, J Biol Chem, vol.279, pp.18614-18622, 2004.

E. Lohmann, M. Periquet, and V. Bonifati, How much phenotypic variation can be attributed to parkin genotype, Ann Neurol, vol.54, pp.176-185, 2003.

E. M. Valente, A. R. Bentivoglio, and P. H. Dixon, Localization of a Novel Locus for Autosomal Recessive Early-Onset Parkinsonism, PARK6, on Human Chromosome 1p35-p36, Am J Hum Genet, vol.68, pp.895-900, 2001.

E. M. Valente, P. M. Abou-sleiman, and V. Caputo, Hereditary Early-Onset Parkinson's Disease Caused by Mutations in PINK1, Science, vol.304, pp.1158-1160, 2004.

A. C. Poole, R. E. Thomas, and L. A. Andrews, The PINK1/Parkin pathway regulates mitochondrial morphology, PNAS, vol.105, pp.1638-1643, 2008.

M. Lazarou, D. A. Sliter, and L. A. Kane, The ubiquitin kinase PINK1 recruits autophagy receptors to induce mitophagy, Nature, vol.524, pp.309-314, 2015.

A. M. Pickrell and R. J. Youle, The Roles of PINK1, Parkin and Mitochondrial Fidelity in Parkinson's Disease, Neuron, vol.85, pp.257-273, 2015.

N. Exner, B. Treske, and D. Paquet, Loss-of-Function of Human PINK1 Results in Mitochondrial Pathology and Can Be Rescued by Parkin, J Neurosci, vol.27, pp.12413-12418, 2007.

L. Zhang, M. Shimoji, and B. Thomas, Mitochondrial localization of the Parkinson's disease related protein DJ-1: implications for pathogenesis, Hum Mol Genet, vol.14, pp.2063-2073, 2005.

T. Taira, Y. Saito, and T. Niki, DJ-1 has a role in antioxidative stress to prevent cell death, EMBO Rep, vol.5, pp.213-218, 2004.

R. M. Canet-avilés, M. A. Wilson, and D. W. Miller, The Parkinson's disease protein DJ-1 is neuroprotective due to cysteine-sulfinic acid-driven mitochondrial localization, Proc Natl Acad Sci U S A, vol.101, pp.9103-9108, 2004.

V. Bonifati, P. Rizzu, and M. J. Van-baren, Mutations in the DJ-1 gene associated with autosomal recessive early-onset parkinsonism, Science, vol.299, pp.256-259, 2003.

G. Annesi, G. Savettieri, and P. Pugliese, DJ-1 mutations and parkinsonismdementia-amyotrophic lateral sclerosis complex, Ann Neurol, vol.58, pp.803-807, 2005.

A. Ramirez, A. Heimbach, and J. Gründemann, Hereditary parkinsonism with dementia is caused by mutations in ATP13A2, encoding a lysosomal type 5 P-type ATPase, Nat Genet, vol.38, pp.1184-1191, 2006.

D. R. Williams, A. Hadeed, and A. Din, Kufor Rakeb disease: autosomal recessive, levodopa-responsive parkinsonism with pyramidal degeneration, supranuclear gaze palsy, and dementia, Mov Disord, vol.20, pp.1264-1271, 2005.

P. Coro, B. Kailash, P. , and L. Abi, Characterization of PLA2G6 as a locus for dystonia-parkinsonism, Annals of Neurology, vol.65, pp.19-23, 2009.

S. Shojaee, F. Sina, and S. S. Banihosseini, 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.

S. Edvardson, Y. Cinnamon, and A. Ta-shma, A Deleterious Mutation in DNAJC6 Encoding the Neuronal-Specific Clathrin-Uncoating Co-Chaperone Auxilin, Is Associated with Juvenile Parkinsonism. PLOS ONE, vol.7, p.36458, 2012.

Ç. Köro?lu, L. Baysal, and M. Cetinkaya, DNAJC6 is responsible for juvenile parkinsonism with phenotypic variability, Parkinsonism & Related Disorders, vol.19, pp.320-324, 2013.

O. Simone, Q. Marialuisa, and F. Mingyan, DNAJC6 Mutations Associated With Early-Onset Parkinson's Disease, vol.79, pp.244-256, 2016.

C. E. Krebs, S. Karkheiran, and J. C. Powell, The Sac1 domain of SYNJ1 identified mutated in a family with early-onset progressive Parkinsonism with generalized seizures, Hum Mutat, vol.34, pp.1200-1207, 2013.

M. Quadri, M. Fang, and M. Picillo, Mutation in the SYNJ1 gene associated with autosomal recessive, early-onset Parkinsonism, Hum Mutat, vol.34, pp.1208-1215, 2013.

K. Hardies, Y. Cai, and C. Jardel, Loss of SYNJ1 dual phosphatase activity leads to early onset refractory seizures and progressive neurological decline, Brain, vol.139, pp.2420-2430, 2016.

G. Arianna, P. Carla, S. Filippo, and M. , Novel mutations in SPG11 cause hereditary spastic paraplegia associated with early-onset levodopa-responsive Parkinsonism, Movement Disorders, vol.26, pp.553-556, 2011.

S. Lesage, V. Drouet, and E. Majounie, 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

M. Anheim, C. Lagier-tourenne, and G. Stevanin, SPG11 spastic paraplegia. A new cause of juvenile parkinsonism, J Neurol, vol.256, pp.104-108, 2009.
URL : https://hal.archives-ouvertes.fr/inserm-00370093

,

H. Büeler, Mitochondrial dynamics, cell death and the pathogenesis of Parkinson's disease, Apoptosis, vol.15, pp.1336-1353, 2010.

A. Camilleri and N. Vassallo, The centrality of mitochondria in the pathogenesis and treatment of Parkinson's disease, CNS Neurosci Ther, vol.20, pp.591-602, 2014.

I. Celardo, L. M. Martins, and S. Gandhi, Unravelling mitochondrial pathways to Parkinson's disease, Br J Pharmacol, vol.171, pp.1943-1957, 2014.

L. Zuo and M. S. Motherwell, The impact of reactive oxygen species and genetic mitochondrial mutations in Parkinson's disease, Gene, vol.532, pp.18-23, 2013.

D. T. Dexter and P. Jenner, Parkinson disease: from pathology to molecular disease mechanisms, Free Radic Biol Med, vol.62, pp.132-144, 2013.

J. Trinh and M. Farrer, Advances in the genetics of Parkinson disease, Nat Rev Neurol, vol.9, pp.445-454, 2013.

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.

J. M. Cedarbaum and F. H. Mcdowell, Sixteen-year follow-up of 100 patients begun on levodopa in 1968: emerging problems, Adv Neurol, vol.45, pp.469-472, 1987.

D. Abbassi and R. Ilbert, Entre Bourguiba et Hannibal: identité tunisienne et histoire depuis l'indépendance. Karthala, 2005.

R. Collignon, Bulletins et Mémoires de la Société d'anthropologie de Paris, 1915.

L. Cherni, A. J. Pakstis, and S. Boussetta, Genetic variation in Tunisia in the context of human diversity worldwide, American Journal of Physical Anthropology, vol.161, p.62, 2016.

K. Fadhlaoui-zid, B. Martinez-cruz, and H. Khodjet-el-khil, Genetic structure of Tunisian ethnic groups revealed by paternal lineages, Am J Phys Anthropol, vol.146, pp.271-280, 2011.

S. Chaker, Langue et littérature berbères, vol.3, 2004.

B. T. Lahn and D. C. Page, Functional coherence of the human Y chromosome, Science, vol.278, pp.675-680, 1997.

P. A. Underhill, P. Shen, and A. A. Lin, Y chromosome sequence variation and the history of human populations, Nat Genet, vol.26, pp.358-361, 2000.

P. A. Underhill, G. Passarino, and A. A. Lin, The phylogeography of Y chromosome binary haplotypes and the origins of modern human populations, 2001.

, Ann Hum Genet, vol.65, pp.43-62

T. M. Karafet, F. L. Mendez, and M. B. Meilerman, New binary polymorphisms reshape and increase resolution of the human Y chromosomal haplogroup tree, 2008.

, Genome Res, vol.18, pp.830-838

F. Cruciani, B. Trombetta, and A. Massaia, A Revised Root for the Human Y Chromosomal Phylogenetic Tree: The Origin of Patrilineal Diversity in Africa, The American Journal of Human Genetics, vol.88, pp.814-818, 2011.

H. Zhong, H. Shi, and X. Qi, Global distribution of Y-chromosome haplogroup C reveals the prehistoric migration routes of African exodus and early settlement in East Asia, Journal of Human Genetics, vol.55, pp.428-435, 2010.

C. Cinnio?lu, R. King, and T. Kivisild, Excavating Y-chromosome haplotype strata in Anatolia, Hum Genet, vol.114, pp.127-148, 2004.

S. Rootsi, C. Magri, and T. Kivisild, Phylogeography of Y-chromosome haplogroup I reveals distinct domains of prehistoric gene flow in europe, Am J Hum Genet, vol.75, pp.128-137, 2004.

O. Semino, C. Magri, and G. Benuzzi, Origin, diffusion, and differentiation of Y-chromosome haplogroups E and J: inferences on the neolithization of Europe and later migratory events in the Mediterranean area, Am J Hum Genet, vol.74, pp.1023-1034, 2004.

L. Scheinfeldt, F. Friedlaender, and J. Friedlaender, Unexpected NRY chromosome variation in Northern Island Melanesia, Mol Biol Evol, vol.23, pp.1628-1641, 2006.

S. L. Zegura, T. M. Karafet, L. A. Zhivotovsky, and M. F. Hammer, High-resolution SNPs and microsatellite haplotypes point to a single, recent entry of Native American Y chromosomes into the Americas, Mol Biol Evol, vol.21, pp.164-175, 2004.

H. Genséric, L. Génétiquement, and . Arabes, , 2013.

, Accessed, vol.3, 2018.

K. Fadhlaoui-zid, J. Dugoujon, and A. Elgaaied, Genetic diversity in Tunisia: a study based on the GM polymorphism of human immunoglobulins, Hum Biol, vol.76, pp.559-567, 2004.

Y. Loveslati, B. Sanchez-mazas, A. Ennafaa, and H. , A study of Gm allotypes and immunoglobulin heavy gamma IGHG genes in Berbers, Arabs and sub-Saharan Africans from Jerba Island, European Journal of Immunogenetics, vol.28, pp.531-538, 2001.

A. Guenounou, B. Loueslati, B. Y. Buhler, and S. , HLA class II genetic diversity in southern Tunisia and the Mediterranean area, International Journal of Immunogenetics, vol.33, pp.93-103, 2006.

K. Fadhlaoui-zid, S. Buhler, and A. Dridi, Polymorphism of HLA class II genes in Berbers from Southern Tunisia, Tissue Antigens, vol.76, pp.416-420, 2010.

E. Bosch, J. Clarimón, A. Pérez-lezaun, and F. Calafell, STR data for 21 loci in northwestern Africa, Forensic Science International, vol.116, pp.41-51, 2001.

H. Khodjet-el-khil, K. Fadhlaoui-zid, and L. Gusmão, Substructure of a Tunisian Berber Population as Inferred from 15 Autosomal Short Tandem Repeat Loci, Human Biology, vol.80, pp.435-448, 2008.

H. Ennafaa, M. B. Amor, and B. Yacoubi-loueslati, Alu polymorphisms in Jerba Island population (Tunisia): comparative study in Arab and Berber groups, 2006.

, Ann Hum Biol, vol.33, pp.634-640

S. Frigi, H. Ennafaa, B. Amor, and M. , Assessing human genetic diversity in Tunisian Berber populations by Alu insertion polymorphisms, Ann Hum Biol, vol.38, pp.53-58, 2011.

K. El-khil, H. Marrakchi, R. T. Loueslati, and B. Y. , Distribution of Y chromosome lineages in Jerba island population, Forensic Sci Int, vol.148, pp.211-218, 2005.

V. Onofri, F. Alessandrini, and C. Turchi, Y-chromosome markers distribution in Northern Africa: High-resolution SNP and STR analysis in Tunisia and Morocco populations, Forensic Science International: Genetics Supplement Series, vol.1, pp.235-236, 2008.

R. F. Uhlmann and E. B. Larson, Effect of education on the mini-mental state examination as a screening test for dementia, J Am Geriatr Soc, vol.39, pp.876-880, 1991.

Y. Wang and C. Gorenstein, Assessment of depression in medical patients: A systematic review of the utility of the Beck Depression Inventory-II, Clinics (Sao Paulo), vol.68, pp.1274-1287, 2013.

C. L. Tomlinson, R. Stowe, and S. Patel, Systematic review of levodopa dose equivalency reporting in Parkinson's disease, Mov Disord, vol.25, pp.2649-2653, 2010.

J. Jankovic, M. Mcdermott, and J. Carter, Variable expression of Parkinson's disease: a base-line analysis of the DATATOP cohort. The Parkinson Study Group, Neurology, vol.40, pp.1529-1534, 1990.

H. Ashkenazy, E. Erez, and E. Martz, ConSurf 2010: calculating evolutionary conservation in sequence and structure of proteins and nucleic acids, Nucleic Acids Res, vol.38, pp.529-533, 2010.

E. Mathe, M. Olivier, and S. Kato, Computational approaches for predicting the biological effect of p53 missense mutations: a comparison of three sequence analysis based methods, Nucleic Acids Res, vol.34, pp.1317-1325, 2006.

Y. Choi and A. P. Chan, PROVEAN web server: a tool to predict the functional effect of amino acid substitutions and indels, Bioinformatics, vol.31, pp.2745-2747, 2015.

I. Adzhubei, D. M. Jordan, and S. R. Sunyaev, Predicting functional effect of human missense mutations using PolyPhen-2, Curr Protoc Hum Genet Chapter, p.20, 2013.

C. Ferrer-costa, M. Orozco, and X. De-la-cruz, Characterization of diseaseassociated single amino acid polymorphisms in terms of sequence and structure properties, J Mol Biol, vol.315, pp.771-786, 2002.

E. Capriotti, R. Calabrese, and R. Casadio, Predicting the insurgence of human genetic diseases associated to single point protein mutations with support vector machines and evolutionary information, Bioinformatics, vol.22, pp.2729-2734, 2006.

R. Calabrese, E. Capriotti, and P. Fariselli, Functional annotations improve the predictive score of human disease-related mutations in proteins, Hum Mutat, vol.30, pp.1237-1244, 2009.

J. M. Schwarz, D. N. Cooper, M. Schuelke, and D. Seelow, MutationTaster2: mutation prediction for the deep-sequencing age, Nat Methods, vol.11, pp.361-362, 2014.

B. Li, V. G. Krishnan, and M. E. Mort, Automated inference of molecular mechanisms of disease from amino acid substitutions, Bioinformatics, vol.25, pp.2744-2750, 2009.

E. Capriotti, P. Fariselli, and R. Casadio, I-Mutant2.0: predicting stability changes upon mutation from the protein sequence or structure, Nucleic Acids Res, vol.33, pp.306-310, 2005.

J. P. Schouten, C. J. Mcelgunn, and R. Waaijer, Relative quantification of 40 nucleic acid sequences by multiplex ligation-dependent probe amplification, Nucleic Acids Res, vol.30, p.57, 2002.

S. De-boer and S. J. White, Genotyping Multiallelic Copy Number Variation with Multiplex Ligation-Dependent Probe Amplification (MLPA), Methods Mol Biol, vol.1492, pp.147-153, 2017.

J. Trinh, E. K. Gustavsson, and C. Vilariño-güell, DNM3 and genetic modifiers of age of onset in LRRK2 Gly2019Ser parkinsonism: a genome-wide linkage and association study, The Lancet Neurology, vol.15, pp.1248-1256, 2016.

R. Higuchi, C. Fockler, G. Dollinger, and R. Watson, Kinetic PCR analysis: real-time monitoring of DNA amplification reactions, Biotechnology (NY), vol.11, pp.1026-1030, 1993.

E. Poitras and A. Houde, La PCR en temps réel: principes et applications, Reviews in Biology and Biotechnology, vol.2, pp.2-11, 2002.

V. Venegas, J. Wang, D. Dimmock, and L. Wong, Real-time quantitative PCR analysis of mitochondrial DNA content, Curr Protoc Hum Genet Chapter, 2011.

D. Dimmock, L. Tang, E. S. Schmitt, and L. Wong, Quantitative Evaluation of the Mitochondrial DNA Depletion Syndrome, Clinical Chemistry, vol.56, pp.1119-1127, 2010.

A. B. West, D. J. Moore, and S. Biskup, Parkinson's disease-associated mutations in leucine-rich repeat kinase 2 augment kinase activity, Proc Natl Acad Sci U S A, vol.102, pp.16842-16847, 2005.

M. Jaleel, R. J. Nichols, and M. Deak, LRRK2 phosphorylates moesin at threonine-558: characterization of how Parkinson's disease mutants affect kinase activity, Biochem J, vol.405, pp.307-317, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00478755

S. Lesage, P. Ibanez, and E. Lohmann, G2019S LRRK2 mutation in French and North African families with Parkinson's disease, Annals of Neurology, vol.58, pp.784-787, 2005.

Z. Landoulsi, S. Benromdhan, B. Djebara, and M. , Using KASP technique to screen LRRK2 G2019S mutation in a large Tunisian cohort, BMC Med Genet, vol.18, 2017.
URL : https://hal.archives-ouvertes.fr/pasteur-01639043

D. Fonzo, A. Rohé, C. F. Ferreira, and J. , A frequent LRRK2 gene mutation associated with autosomal dominant Parkinson's disease, Lancet, vol.365, pp.412-415, 2005.

W. C. Nichols, N. Pankratz, and D. Hernandez, Genetic screening for a single common LRRK2 mutation in familial Parkinson's disease, The Lancet, vol.365, pp.410-412, 2005.

W. P. Gilks, P. M. Abou-sleiman, and S. Gandhi, A common LRRK2 mutation in idiopathic Parkinson's disease, Lancet, vol.365, pp.415-416, 2005.

B. Vijayan, S. Gopala, and A. Kishore, LRRK2 G2019S mutation does not contribute to Parkinson's disease in South India, Neurology India, vol.59, p.157, 2011.

M. Cornejo-olivas, L. Torres, and M. R. Velit-salazar, Variable frequency of LRRK2 variants in the Latin American research consortium on the genetics of Parkinson's disease (LARGE-PD), a case of ancestry, NPJ Parkinsons Dis, vol.3, 2017.

A. Thaler, E. Ash, and Z. Gan-or, The LRRK2 G2019S mutation as the cause of Parkinson's disease in Ashkenazi Jews, J Neural Transm, vol.116, pp.1473-1482, 2009.

L. J. Ozelius, G. Senthil, and R. Saunders-pullman, LRRK2 G2019S as a cause of Parkinson's disease in Ashkenazi Jews, N Engl J Med, vol.354, pp.424-425, 2006.

D. G. Healy, M. Falchi, and S. S. O&apos;sullivan, Phenotype, genotype, and worldwide genetic penetrance of LRRK2-associated Parkinson's disease: a casecontrol study, The Lancet Neurology, vol.7, pp.583-590, 2008.

N. Change, G. Mercier, and G. Lucotte, Genetic screening of the G2019S mutation of the LRRK2 gene in Southwest European, North African, and Sephardic Jewish subjects, Genet Test, vol.12, pp.333-339, 2008.

L. Warren, R. Gibson, and L. Ishihara, A founding LRRK2 haplotype shared by Tunisian, US, European and Middle Eastern families with Parkinson's disease, Parkinsonism & Related Disorders, vol.14, pp.77-80, 2008.

S. Goldwurm, A. Fonzo, and E. Simons, The G6055A (G2019S) mutation in LRRK2 is frequent in both early and late onset Parkinson's disease and originates from a common ancestor, J Med Genet, vol.42, p.65, 2005.

J. Kachergus, I. F. Mata, and M. Hulihan, Identification of a Novel LRRK2 Mutation Linked to Autosomal Dominant Parkinsonism: Evidence of a Common Founder across European Populations, Am J Hum Genet, vol.76, pp.672-680, 2005.

S. Lesage, A. Leutenegger, and P. Ibanez, LRRK2 Haplotype Analyses in European and North African Families with Parkinson Disease: A Common Founder for the G2019S Mutation Dating from the 13th Century, Am J Hum Genet, vol.77, pp.330-332, 2005.

C. P. Zabetian, C. M. Hutter, and D. Yearout, LRRK2 G2019S in Families with Parkinson Disease Who Originated from Europe and the Middle East: Evidence of Two Distinct Founding Events Beginning Two Millennia Ago, Am J Hum Genet, vol.79, pp.752-758, 2006.

C. P. Zabetian, H. Morino, and H. Ujike, Identification and haplotype analysis of LRRK2 G2019S in Japanese patients with Parkinson disease, Neurology, vol.67, pp.697-699, 2006.

H. Tomiyama, Y. Li, and M. Funayama, Clinicogenetic study of mutations in LRRK2 exon 41 in Parkinson's disease patients from 18 countries, Mov Disord, vol.21, pp.1102-1108, 2006.

A. Bar-shira, C. M. Hutter, and N. Giladi, Ashkenazi Parkinson's disease patients with the LRRK2 G2019S mutation share a common founder dating from the second to fifth centuries, neurogenetics, vol.10, pp.355-358, 2009.

B. E. Haj, R. Salmi, A. Regragui, and W. , Evidence for prehistoric origins of the G2019S mutation in the North African Berber population, PLoS One, vol.12, 2017.

G. Lucotte, D. David, and N. Change, New contribution on the LRRK2 G2019S mutation associated to Parkinson's disease: age estimation of a common founder event of old age in Moroccan Berbers, International Journal of Modern Anthropology, vol.1, pp.11-22, 2012.

B. R. Al-mubarak, S. A. Bohlega, and T. S. Alkhairallah, Parkinson's Disease in Saudi Patients: A Genetic Study, PLoS One, vol.10, 2015.

D. I. Hashad, A. A. Abou-zeid, and G. A. Achmawy, G2019S mutation of the leucine-rich repeat kinase 2 gene in a cohort of Egyptian patients with Parkinson's disease, Genet Test Mol Biomarkers, vol.15, pp.861-866, 2011.

E. Desoky, E. Khedr, M. Khalil, and T. Gasser, Genetic Analysis of Leucin-Rich Repeat Kinase 2 (LRRK2) G2019S Mutation in a Sample of Egyptian Patients with Références Parkinson's Disease, a Pilot Study, British Journal of Medicine and Medical Research, vol.5, pp.404-408, 2015.

F. Cruciani, L. Fratta, R. Santolamazza, and P. , Phylogeographic Analysis of Haplogroup E3b (E-M215) Y Chromosomes Reveals Multiple Migratory Events Within and Out Of Africa, Am J Hum Genet, vol.74, pp.1014-1022, 2004.

M. Kestenbaum and R. N. Alcalay, Clinical Features of LRRK2 Carriers with Parkinson's Disease, Leucine-Rich Repeat Kinase 2 (LRRK2), pp.31-48, 2017.

S. Luciano, M. Wang, C. Ortega, and R. A. , Sex differences in LRRK2 G2019S and idiopathic Parkinson's Disease, Ann Clin Transl Neurol, vol.4, pp.801-810, 2017.

K. Marder, Y. Wang, and R. N. Alcalay, Age-specific penetrance of LRRK2 G2019S in the Michael J. Fox Ashkenazi Jewish LRRK2 Consortium, Neurology, vol.85, pp.89-95, 2015.

G. Yahalom, Y. Orlev, and O. S. Cohen, Motor progression of Parkinson's disease with the leucine-rich repeat kinase 2 G2019S mutation, Movement Disorders, vol.29, pp.1057-1060, 2014.

D. G. Healy, M. Falchi, and S. S. O&apos;sullivan, Phenotype, genotype, and worldwide genetic penetrance of LRRK2-associated Parkinson's disease: a casecontrol study, Lancet Neurol, vol.7, pp.583-590, 2008.

C. Marras, B. Schuele, and R. P. Munhoz, Phenotype in parkinsonian and nonparkinsonian LRRK2 G2019S mutation carriers, Neurology, vol.77, pp.325-333, 2011.

R. N. Alcalay, A. Mirelman, and R. Saunders-pullman, Parkinson Disease Phenotype in Ashkenazi Jews with and without LRRK2 G2019S mutations, Mov Disord, vol.28, 2013.

J. Trinh, R. Amouri, and J. E. Duda, A comparative study of Parkinson's disease and leucine-rich repeat kinase 2 p.G2019S parkinsonism, Neurobiology of Aging, vol.35, pp.1125-1131, 2014.

A. Bouhouche, H. Tibar, B. E. Haj, and R. , LRRK2 G2019S Mutation: Prevalence and Clinical Features in Moroccans with Parkinson's Disease, Parkinsons Dis, 2017.

L. Ishihara, R. A. Gibson, and L. Warren, Screening for Lrrk2 G2019S and clinical comparison of Tunisian and North American Caucasian Parkinson's disease families, Movement Disorders, vol.22, pp.55-61, 2007.

C. Marras, R. N. Alcalay, and C. Caspell-garcia, Motor and nonmotor heterogeneity of LRRK2-related and idiopathic Parkinson's disease, Mov Disord, vol.31, pp.1192-1202, 2016.

A. Mirelman, T. Heman, and K. Yasinovsky, Fall risk and gait in Parkinson's disease: The role of the LRRK2 G2019S mutation, Movement Disorders, vol.28, pp.1683-1690, 2013.

R. N. Alcalay, H. Mejia-santana, and A. Mirelman, Neuropsychological performance in LRRK2 G2019S carriers with Parkinson's disease, Parkinsonism Relat Disord, vol.21, pp.106-110, 2015.

R. Saunders-pullman, A. Mirelman, and R. N. Alcalay, Progression in the LRRK2 -Associated Parkinson Disease Population, JAMA Neurology, vol.75, p.312, 2018.

J. H. Somme, M. Salazar, A. Gonzalez, and A. , Cognitive and behavioral symptoms in Parkinson's disease patients with the G2019S and R1441G mutations of the LRRK2 gene, Parkinsonism & Related Disorders, vol.21, pp.494-499, 2015.

S. Srivatsal, B. Cholerton, and J. B. Leverenz, Cognitive Profile of LRRK2-related Parkinson's Disease, Mov Disord, vol.30, pp.728-733, 2015.

B. Sassi, S. Nabli, F. Hentati, and E. , Cognitive dysfunction in Tunisian LRRK2 associated Parkinson's disease, Parkinsonism & Related Disorders, vol.18, pp.243-246, 2012.

R. Cilia, C. Siri, and D. Rusconi, LRRK2 mutations in Parkinson's disease: Confirmation of a gender effect in the Italian population, Parkinsonism Relat Disord, vol.20, pp.911-914, 2014.

Z. Gan-or, C. S. Leblond, and V. Mallett, LRRK2 mutations in Parkinson disease; a sex effect or lack thereof? A meta-analysis, Parkinsonism & Related Disorders, vol.21, pp.778-782, 2015.

D. Georgiev, K. Hamberg, and M. Hariz, Gender differences in Parkinson's disease: A clinical perspective, Acta Neurol Scand, vol.136, pp.570-584, 2017.

L. Ishihara, L. Warren, and R. Gibson, Clinical Features of Parkinson Disease Patients With Homozygous Leucine-Rich Repeat Kinase 2 G2019S Mutations, Archives of Neurology, vol.63, p.1250, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00221455

M. Mettlen, T. Pucadyil, R. Ramachandran, and S. L. Schmid, Dissecting dynamin's role in clathrin-mediated endocytosis, Biochem Soc Trans, vol.37, pp.1022-1026, 2009.

K. Stafa, E. Tsika, and R. Moser, Functional interaction of Parkinson's disease-associated LRRK2 with members of the dynamin GTPase superfamily, Hum Mol Genet, vol.23, pp.2055-2077, 2014.

R. Fernández-santiago, A. Garrido, and J. Infante, ) ?-synuclein (SNCA) but not dynamin 3 (DNM3) influences age at onset of leucine-rich repeat kinase 2 (LRRK2) Parkinson's disease in Spain, Mov Disord, vol.33, pp.637-641, 2018.

A. Rosenbaum, H. Gershoni-baruch, and R. , Mutations in the Glucocerebrosidase Gene and Parkinson's Disease in Ashkenazi Jews, New England Journal of Medicine, vol.351, pp.1972-1977, 2004.

M. J. Eblan, J. M. Walker, and E. Sidransky, The glucocerebrosidase gene and Parkinson's disease in Ashkenazi Jews, N Engl J Med, vol.352, pp.728-731, 2005.

P. Alfonso, J. C. Rodríguez-rey, and A. Gañán, Expression and functional characterization of mutated glucocerebrosidase alleles causing Gaucher disease in Spanish patients, Blood Cells Mol Dis, vol.32, pp.218-225, 2004.

M. Pasmanik-chor, L. Madar-shapiro, and E. O. Stein, Expression of mutated glucocerebrosidase alleles in human cells, Hum Mol Genet, vol.6, pp.887-895, 1997.

Y. N. Blech-hermoni, S. G. Ziegler, and K. S. Hruska, In silico and functional studies of the regulation of the glucocerebrosidase gene, Mol Genet Metab, vol.99, pp.275-282, 2010.

M. Manickam, P. Ravanan, P. Singh, and P. Talwar, In silico identification of genetic variants in glucocerebrosidase (GBA) gene involved in Gaucher's disease using multiple software tools, Front Genet, vol.5, p.148, 2014.

V. Koprivica, D. L. Stone, and J. K. Park, Analysis and classification of 304 mutant alleles in patients with type 1 and type 3 Gaucher disease, Am J Hum Genet, vol.66, pp.1777-1786, 2000.

Z. Gan-or, I. Amshalom, and L. L. Kilarski, Differential effects of severe vs mild GBA mutations on Parkinson disease, Neurology, vol.84, pp.880-887, 2015.

A. Thaler, N. Bregman, and T. Gurevich, Parkinson's disease phenotype is influenced by the severity of the mutations in the GBA gene, Parkinsonism Relat Disord, 2018.

. Références,

P. Suwannarat, S. Keeratichamroen, and D. Wattanasirichaigoon, Molecular characterization of type 3 (neuronopathic) Gaucher disease in Thai patients, Molecules, and Diseases, vol.39, pp.348-352, 2007.

K. Nuytemans, B. Meeus, and D. Crosiers, Relative contribution of simple mutations vs. copy number variations in five Parkinson disease genes in the Belgian population, Human Mutation, vol.30, pp.1054-1061

O. A. Ross, A. I. Soto-ortolaza, and M. G. Heckman, LRRK2 exonic variants and susceptibility to Parkinson's disease, Lancet Neurol, vol.10, pp.898-908, 2011.

D. Fonzo, A. Tassorelli, C. , D. Mari, and M. , Comprehensive analysis of the LRRK2 gene in sixty families with Parkinson's disease, Eur J Hum Genet, vol.14, pp.322-331, 2006.

S. Lesage, C. Condroyer, and A. Lannuzel, Molecular analyses of the LRRK2 gene in European and North African autosomal dominant Parkinson's disease, J Med Genet, vol.46, pp.458-464, 2009.

C. Paisán-ruíz, P. Nath, and N. Washecka, Comprehensive analysis of LRRK2 in publicly available Parkinson's disease cases and neurologically normal controls, Human Mutation, vol.29, pp.485-490

C. Paisán-ruíz, A. E. Lang, and T. Kawarai, LRRK2 gene in Parkinson disease: mutation analysis and case control association study, Neurology, vol.65, pp.696-700, 2005.

B. A. Benitez, A. A. Davis, and S. C. Jin, Resequencing analysis of five Mendelian genes and the top genes from genome-wide association studies in Parkinson's Disease, Molecular Neurodegeneration, vol.11, p.29, 2016.

J. N. Foo, L. C. Tan, and H. Liany, Analysis of non-synonymous-coding variants of Parkinson's disease-related pathogenic and susceptibility genes in East Asian populations, Hum Mol Genet, vol.23, pp.3891-3897, 2014.

N. Pankratz, D. K. Kissell, and M. W. Pauciulo, Parkin dosage mutations have greater pathogenicity in familial PD than simple sequence mutations, Neurology, vol.73, pp.279-286, 2009.

I. F. Mata, V. Alvarez, and E. Coto, Homozygous partial genomic triplication of the parkin gene in early-onset parkinsonism, Neuroscience Letters, vol.380, pp.257-259, 2005.

K. Hedrich, M. Kann, and A. J. Lanthaler, The importance of gene dosage studies: mutational analysis of the parkin gene in early-onset parkinsonism, Hum Mol Genet, vol.10, pp.1649-1656, 2001.

T. Foroud, S. K. Uniacke, and L. Liu, Heterozygosity for a mutation in the parkin gene leads to later onset Parkinson disease, Neurology, vol.60, pp.796-801, 2003.

S. N. Illarioshkin, M. Periquet, and N. Rawal, Mutation analysis of the parkin gene in Russian families with autosomal recessive juvenile parkinsonism, Mov Disord, vol.18, pp.914-919, 2003.

A. Djarmati, K. Hedrich, and M. Svetel, Detection of Parkin (PARK2) and DJ1 (PARK7) mutations in early-onset Parkinson disease: Parkin mutation frequency depends on ethnic origin of patients, Hum Mutat, vol.23, p.525, 2004.

W. Shyu, S. Lin, and M. Chiang, Early-onset Parkinson's disease in a Chinese population: 99mTc-TRODAT-1 SPECT, Parkin gene analysis and clinical study, Parkinsonism Relat Disord, vol.11, pp.173-180, 2005.

R. Inzelberg, N. Hattori, and Y. Mizuno, Dopaminergic dysfunction in unrelated, asymptomatic carriers of a single parkin mutation, Neurology, vol.65, p.1843, 2005.

S. Chaudhary, M. Behari, and M. Dihana, Parkin mutations in familial and sporadic Parkinson's disease among Indians, Parkinsonism Relat Disord, vol.12, pp.239-245, 2006.

E. J. Chung, C. Ki, and W. Y. Lee, Clinical features and gene analysis in Korean patients with early-onset Parkinson disease, Arch Neurol, vol.63, pp.1170-1174, 2006.

M. Mani, S. Y. Lee, and L. Lucast, The dual phosphatase activity of synaptojanin1 is required for both efficient synaptic vesicle endocytosis and reavailability at nerve terminals, Neuron, vol.56, pp.1004-1018, 2007.

P. S. Mcpherson, E. P. Garcia, and V. I. Slepnev, A presynaptic inositol-5-phosphatase, Nature, vol.379, pp.353-357, 1996.

D. Paolo, G. , D. Camilli, and P. , Phosphoinositides in cell regulation and membrane dynamics, Nature, vol.443, pp.651-657, 2006.

R. M. Perera, R. Zoncu, and L. Lucast, Two synaptojanin 1 isoforms are recruited to clathrin-coated pits at different stages, Proc Natl Acad Sci, vol.103, pp.19332-19337, 2006.

Y. Tsujishita, S. Guo, and L. E. Stolz, Specificity determinants in phosphoinositide dephosphorylation: crystal structure of an archetypal inositol polyphosphate 5-phosphatase, Cell, vol.105, pp.379-389, 2001.

K. Soda, D. M. Balkin, and S. M. Ferguson, Role of dynamin, synaptojanin, and endophilin in podocyte foot processes, J Clin Invest, vol.122, pp.4401-4411, 2012.

M. Krauß and V. Haucke, Phosphoinositide-metabolizing enzymes at the interface between membrane traffic and cell signalling, EMBO reports, vol.8, pp.241-246, 2007.

G. J. Praefcke, M. G. Ford, and E. M. Schmid, Evolving nature of the AP2 ?-appendage hub during clathrin-coated vesicle endocytosis, The EMBO Journal, vol.23, pp.4371-4383, 2004.

C. Haffner, K. Takei, and H. Chen, Synaptojanin 1: localization on coated endocytic intermediates in nerve terminals and interaction of its 170 kDa isoform with Eps15, FEBS Letters, vol.419, pp.175-180, 1997.

M. Mettlen, M. Stoeber, and D. Loerke, Endocytic Accessory Proteins Are Functionally Distinguished by Their Differential Effects on the Maturation of Clathrin-coated Pits, Mol Biol Cell, vol.20, pp.3251-3260, 2009.

E. M. Schmid, M. Ford, and A. Burtey, Role of the AP2 ?-Appendage Hub in Recruiting Partners for Clathrin-Coated Vesicle Assembly, PLoS Biol, vol.4, 2006.

S. Olgiati, D. Rosa, A. Quadri, and M. , PARK20 caused by SYNJ1 homozygous Arg258Gln mutation in a new Italian family, Neurogenetics, vol.15, pp.183-188, 2014.

L. Kirola, M. Behari, C. Shishir, and B. K. Thelma, Identification of a novel homozygous mutation Arg459Pro in SYNJ1 gene of an Indian family with autosomal recessive juvenile Parkinsonism, Parkinsonism Relat Disord, vol.31, pp.124-128, 2016.

D. A. Dyment, A. C. Smith, and P. Humphreys, Homozygous nonsense mutation in SYNJ1 associated with intractable epilepsy and tau pathology, Neurobiol Aging, vol.36, 2015.

S. Taghavi, R. Chaouni, and A. Tafakhori, A Clinical and Molecular Genetic Study of 50 Families with Autosomal Recessive Parkinsonism Revealed Known and Novel Gene Mutations, Mol Neurobiol, 2017.

M. Picillo, A. Ranieri, and G. Orefice, Clinical progression of SYNJ1-related early onset atypical parkinsonism: 3-year follow up of the original Italian family, J Neurol, vol.261, pp.823-824, 2014.

L. Ishihara-paul, M. M. Hulihan, and J. Kachergus, mutations and parkinsonism, Neurology, vol.71, pp.896-902, 2008.

V. Bonifati, C. F. Rohé, and G. J. Breedveld, Early-onset parkinsonism associated with PINK1 mutations: frequency, genotypes, and phenotypes, Neurology, vol.65, pp.87-95, 2005.

Y. Weng, Y. Chou, and W. Wu, PINK1 mutation in Taiwanese earlyonset parkinsonism : clinical, genetic, and dopamine transporter studies, J Neurol, vol.254, pp.1347-1355, 2007.

C. Koros, A. Simitsi, and L. Stefanis, Genetics of Parkinson's Disease, International Review of Neurobiology, pp.197-231, 2017.

C. Y. Kim and R. N. Alcalay, Genetic Forms of Parkinson's Disease, Semin Neurol, vol.37, pp.135-146, 2017.

M. Kasten, C. Hartmann, and J. Hampf, Genotype-Phenotype Relations for the Parkinson's Disease Genes Parkin, PINK1, DJ1: MDSGene Systematic Review, Movement Disorders, vol.33, pp.730-741, 2018.

B. L. Cantarel, P. M. Coutinho, and C. Rancurel, The Carbohydrate-Active EnZymes database (CAZy): an expert resource for Glycogenomics, Nucleic Acids Res, vol.37, pp.233-238, 2009.

A. C. Crawley, Developmental Analysis of CNS Pathology in the Lysosomal Storage Disease >-Mannosidosis, J Neuropathol Exp Neurol, vol.66, p.11, 2007.

D. R. Tulsiani and O. Touster, Swainsonine, a potent mannosidase inhibitor, elevates rat liver and brain lysosomal alpha-D-mannosidase, decreases Golgi alpha-D-mannosidase II, and increases the plasma levels of several acid hydrolases, Arch Biochem Biophys, vol.224, pp.594-600, 1983.

P. M. Novikoff, N. Ab, and T. Dp, Effects of swainsonine on rat liver and kidney: biochemical and morphological studies, J Cell Biol, vol.101, pp.339-349, 1985.

A. C. Crawley and S. U. Walkley, Developmental Analysis of CNS Pathology in the Lysosomal Storage Disease ?-Mannosidosis, J Neuropathol Exp Neurol, vol.66, pp.687-697, 2007.

D. A. Siegel and S. U. Walkley, Growth of ectopic dendrites on cortical pyramidal neurons in neuronal storage diseases correlates with abnormal accumulation of GM2 ganglioside, J Neurochem, vol.62, pp.1852-1862, 1994.

L. A. Goodman, P. O. Livingston, and S. U. Walkley, Ectopic dendrites occur only on cortical pyramidal cells containing elevated GM2 ganglioside in alphamannosidosis, Proc Natl Acad Sci, vol.88, pp.11330-11334, 1991.

M. Vandevelde, R. Fankhauser, and P. Bichsel, Hereditary neurovisceral mannosidosis associated with alpha-mannosidase deficiency in a family of Persian cats, Acta Neuropathol, vol.58, pp.64-68, 1982.

C. H. Vite, J. C. Mcgowan, and K. G. Braund, Histopathology, electrodiagnostic testing, and magnetic resonance imaging show significant peripheral and central nervous system myelin abnormalities in the cat model of alpha-mannosidosis, J Neuropathol Exp Neurol, vol.60, pp.817-828, 2001.

V. K. Mootha, J. Bunkenborg, and J. V. Olsen, Integrated analysis of protein composition, tissue diversity, and gene regulation in mouse mitochondria, Cell, vol.115, pp.629-640, 2003.

B. Gilquin, E. Taillebourg, and N. Cherradi, The AAA+ ATPase ATAD3A Controls Mitochondrial Dynamics at the Interface of the Inner and Outer Membranes, Mol Cell Biol, vol.30, 1984.
URL : https://hal.archives-ouvertes.fr/hal-02459107

H. Fang, C. Chang, and S. Hsu, ATPase family AAA domain-containing 3A is a novel anti-apoptotic factor in lung adenocarcinoma cells, J Cell Sci, vol.123, pp.1171-1180, 2010.

M. Hoffmann, N. Bellance, and R. Rossignol, C. elegans ATAD-3 Is Essential for Mitochondrial Activity and Development, PLOS ONE, vol.4, p.7644, 2009.
URL : https://hal.archives-ouvertes.fr/in2p3-00025853

T. Harel, W. H. Yoon, and C. Garone, Recurrent De Novo and Biallelic Variation of ATAD3A, Encoding a Mitochondrial Membrane Protein, Results in Distinct Neurological Syndromes, The American Journal of Human Genetics, vol.99, pp.831-845, 2016.

H. M. Cooper, Y. Yang, and E. Ylikallio, ATPase-deficient mitochondrial inner membrane protein ATAD3A disturbs mitochondrial dynamics in dominant hereditary spastic paraplegia, Hum Mol Genet, vol.26, pp.1432-1443, 2017.

A. Rötig and J. Poulton, Genetic causes of mitochondrial DNA depletion in humans, Biochim Biophys Acta, vol.1792, pp.1103-1108, 2009.

W. Q. Qiu, D. M. Walsh, and Z. Ye, Insulin-degrading Enzyme Regulates Extracellular Levels of Amyloid ?-Protein by Degradation, J Biol Chem, vol.273, pp.32730-32738, 1998.

K. Vekrellis, Z. Ye, and W. Q. Qiu, Neurons regulate extracellular levels of amyloid beta-protein via proteolysis by insulin-degrading enzyme, J Neurosci, vol.20, pp.1657-1665, 2000.

E. Malito, L. A. Ralat, and M. Manolopoulou, Molecular bases for the recognition of short peptide substrates and cysteine-directed modifications of human insulin-degrading enzyme, Biochemistry, vol.47, pp.12822-12834, 2008.

B. C. Miller, E. A. Eckman, and K. Sambamurti, Amyloid-? peptide levels in brain are inversely correlated with insulysin activity levels in vivo, PNAS, vol.100, pp.6221-6226, 2003.

M. A. Leissring, W. Farris, and A. Y. Chang, Enhanced Proteolysis of ?-Amyloid in APP Transgenic Mice Prevents Plaque Formation, Secondary Pathology, and Premature Death, Neuron, vol.40, pp.1087-1093, 2003.

W. Farris, S. Mansourian, and Y. Chang, Insulin-degrading enzyme regulates the levels of insulin, amyloid -protein, and the -amyloid precursor protein intracellular domain in vivo, Proceedings of the National Academy of Sciences, vol.100, pp.4162-4167, 2003.

M. Hiltunen, T. Van-groen, and J. Jolkkonen, Functional Roles of Amyloid-? Protein Precursor and Amyloid-? Peptides: Evidence from Experimental Studies, Journal of Alzheimer's Disease, vol.18, pp.401-412, 2009.

S. S. Sisodia and D. L. Price, Role of the beta-amyloid protein in Alzheimer's disease, FASEB J, vol.9, pp.366-370, 1995.

F. M. Laferla, B. T. Tinkle, and C. J. Bieberich, The Alzheimer's A beta peptide induces neurodegeneration and apoptotic cell death in transgenic mice, Nat Genet, vol.9, pp.21-30, 1995.

R. D. Terry, Alzheimer's disease and the aging brain, J Geriatr Psychiatry Neurol, vol.19, pp.125-128, 2006.

R. W. Carrell, Cell toxicity and conformational disease, Trends in Cell Biology, vol.15, pp.574-580, 2005.

I. F. Tsigelny, L. Crews, and P. Desplats, Mechanisms of Hybrid Oligomer Formation in the Pathogenesis of Combined Alzheimer's and Parkinson's Diseases, PLoS One, vol.3, 2008.

J. Oláh, O. Vincze, and D. Virók, Interactions of pathological hallmark proteins: tubulin polymerization promoting protein/p25, beta-amyloid, and alpha-synuclein, J Biol Chem, vol.286, pp.34088-34100, 2011.

X. Hu, J. A. Kim, and A. Castillo, NBA1/MERIT40 and BRE Interaction Is Required for the Integrity of Two Distinct Deubiquitinating Enzyme BRCC36-containing Complexes, J Biol Chem, vol.286, pp.11734-11745, 2011.

L. Feng, J. Huang, and J. Chen, MERIT40 facilitates BRCA1 localization and DNA damage repair, Genes Dev, vol.23, pp.719-728, 2009.

K. Gwinn-hardy, A. Singleton, and P. O&apos;suilleabhain, Spinocerebellar ataxia type 3 phenotypically resembling parkinson disease in a black family, Arch Neurol, vol.58, pp.296-299, 2001.

U. Gulsuner, H. Gulsuner, S. Mercan, and F. N. , Mitochondrial serine protease HTRA2 p.G399S in a kindred with essential tremor and Parkinson disease, Proceedings of the National Academy of Sciences, vol.111, pp.18285-18290, 2014.

M. Caillet-boudin, L. Buée, N. Sergeant, and B. Lefebvre, Regulation of human MAPT gene expression, Molecular Neurodegeneration, vol.10, p.28, 2015.

G. Lindwall and R. D. Cole, Phosphorylation affects the ability of tau protein to promote microtubule assembly, J Biol Chem, vol.259, pp.5301-5305, 1984.

S. Wray and P. A. Lewis, A Tangled Web -Tau and Sporadic Parkinson's, Disease. Front Psychiatry, vol.1, 2010.

, Annexes