Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
1.
Mov Disord ; 38(4): 604-615, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36788297

RESUMEN

BACKGROUND: Epidemiological studies that examined the association between Parkinson's disease (PD) and cancers led to inconsistent results, but they face a number of methodological difficulties. OBJECTIVE: We used results from genome-wide association studies (GWASs) to study the genetic correlation between PD and different cancers to identify common genetic risk factors. METHODS: We used individual data for participants of European ancestry from the Courage-PD (Comprehensive Unbiased Risk Factor Assessment for Genetics and Environment in Parkinson's Disease; PD, N = 16,519) and EPITHYR (differentiated thyroid cancer, N = 3527) consortia and summary statistics of GWASs from iPDGC (International Parkinson Disease Genomics Consortium; PD, N = 482,730), Melanoma Meta-Analysis Consortium (MMAC), Breast Cancer Association Consortium (breast cancer), the Prostate Cancer Association Group to Investigate Cancer Associated Alterations in the Genome (prostate cancer), International Lung Cancer Consortium (lung cancer), and Ovarian Cancer Association Consortium (ovarian cancer) (N comprised between 36,017 and 228,951 for cancer GWASs). We estimated the genetic correlation between PD and cancers using linkage disequilibrium score regression. We studied the association between PD and polymorphisms associated with cancers, and vice versa, using cross-phenotypes polygenic risk score (PRS) analyses. RESULTS: We confirmed a previously reported positive genetic correlation of PD with melanoma (Gcorr = 0.16 [0.04; 0.28]) and reported an additional significant positive correlation of PD with prostate cancer (Gcorr = 0.11 [0.03; 0.19]). There was a significant inverse association between the PRS for ovarian cancer and PD (odds ratio [OR] = 0.89 [0.84; 0.94]). Conversely, the PRS of PD was positively associated with breast cancer (OR = 1.08 [1.06; 1.10]) and inversely associated with ovarian cancer (OR = 0.95 [0.91; 0.99]). The association between PD and ovarian cancer was mostly driven by rs183211 located in an intron of the NSF gene (17q21.31). CONCLUSIONS: We show evidence in favor of a contribution of pleiotropic genes to the association between PD and specific cancers. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society. This article has been contributed to by U.S. Government employees and their work is in the public domain in the USA.


Asunto(s)
Neoplasias Pulmonares , Melanoma , Neoplasias Ováricas , Enfermedad de Parkinson , Neoplasias de la Próstata , Humanos , Masculino , Femenino , Enfermedad de Parkinson/epidemiología , Enfermedad de Parkinson/genética , Estudio de Asociación del Genoma Completo , Predisposición Genética a la Enfermedad/genética , Polimorfismo de Nucleótido Simple/genética , Melanoma/epidemiología , Melanoma/genética , Factores de Riesgo
2.
Mov Disord ; 37(9): 1929-1937, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35810454

RESUMEN

BACKGROUND: Two studies that examined the interaction between HLA-DRB1 and smoking in Parkinson's disease (PD) yielded findings in opposite directions. OBJECTIVE: To perform a large-scale independent replication of the HLA-DRB1 × smoking interaction. METHODS: We genotyped 182 single nucleotide polymorphism (SNPs) associated with smoking initiation in 12 424 cases and 9480 controls to perform a Mendelian randomization (MR) analysis in strata defined by HLA-DRB1. RESULTS: At the amino acid level, a valine at position 11 (V11) in HLA-DRB1 displayed the strongest association with PD. MR showed an inverse association between genetically predicted smoking initiation and PD only in absence of V11 (odds ratio, 0.74, 95% confidence interval, 0.59-0.93, PInteraction  = 0.028). In silico predictions of the influence of V11 and smoking-induced modifications of α-synuclein on binding affinity showed findings consistent with this interaction pattern. CONCLUSIONS: Despite being one of the most robust findings in PD research, the mechanisms underlying the inverse association between smoking and PD remain unknown. Our findings may help better understand this association. © 2022 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.


Asunto(s)
Enfermedad de Parkinson , Predisposición Genética a la Enfermedad , Cadenas HLA-DRB1/genética , Humanos , Enfermedad de Parkinson/genética , Polimorfismo de Nucleótido Simple/genética , Fumar/genética
3.
J Parkinsons Dis ; 12(1): 267-282, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34633332

RESUMEN

BACKGROUND: Previous studies showed that lifestyle behaviors (cigarette smoking, alcohol, coffee) are inversely associated with Parkinson's disease (PD). The prodromal phase of PD raises the possibility that these associations may be explained by reverse causation. OBJECTIVE: To examine associations of lifestyle behaviors with PD using two-sample Mendelian randomisation (MR) and the potential for survival and incidence-prevalence biases. METHODS: We used summary statistics from publicly available studies to estimate the association of genetic polymorphisms with lifestyle behaviors, and from Courage-PD (7,369 cases, 7,018 controls; European ancestry) to estimate the association of these variants with PD. We used the inverse-variance weighted method to compute odds ratios (ORIVW) of PD and 95%confidence intervals (CI). Significance was determined using a Bonferroni-corrected significance threshold (p = 0.017). RESULTS: We found a significant inverse association between smoking initiation and PD (ORIVW per 1-SD increase in the prevalence of ever smoking = 0.74, 95%CI = 0.60-0.93, p = 0.009) without significant directional pleiotropy. Associations in participants ≤67 years old and cases with disease duration ≤7 years were of a similar size. No significant associations were observed for alcohol and coffee drinking. In reverse MR, genetic liability toward PD was not associated with smoking or coffee drinking but was positively associated with alcohol drinking. CONCLUSION: Our findings are in favor of an inverse association between smoking and PD that is not explained by reverse causation, confounding, and survival or incidence-prevalence biases. Genetic liability toward PD was positively associated with alcohol drinking. Conclusions on the association of alcohol and coffee drinking with PD are hampered by insufficient statistical power.


Asunto(s)
Café , Enfermedad de Parkinson , Anciano , Consumo de Bebidas Alcohólicas/epidemiología , Consumo de Bebidas Alcohólicas/genética , Estudio de Asociación del Genoma Completo , Humanos , Análisis de la Aleatorización Mendeliana , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/genética , Factores de Riesgo , Fumar/epidemiología
4.
Parkinsonism Relat Disord ; 76: 56-62, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32866938

RESUMEN

BACKGROUND: Subthalamic nucleus deep brain stimulation (STN-DBS) has demonstrated its efficacy on motor complications in advanced Parkinson's disease (PD) but does not modify disease progression. Genetic forms of PD have been associated with different cognitive progression profiles. OBJECTIVE: To assess the effect of PD-related genetic mutations on cognitive outcome after STN-DBS. METHODS: Patients with STN-DBS were screened for LRRK2, GBA, and PRKN mutations at the Pitié-Salpêtrière Hospital between 1997 and 2009. Patients with known monogenetic forms of PD from six other centers were also included. The Mattis Dementia Rating Scale (MDRS) was used to evaluate cognition at baseline and one-year post-surgery. The standardized Unified PD Rating Scale (UPDRS) evaluation On and Off medication/DBS was also administered. A generalized linear model adjusted for sex, ethnicity, age at onset, and disease duration was used to evaluate the effect of genetic factors on MDRS changes. RESULTS: We analyzed 208 patients (131 males, 77 females, 54.3 ± 8.8 years) including 25 GBA, 18 LRRK2, 22 PRKN, and 143 PD patients without mutations. PRKN patients were younger and had a longer disease duration at baseline. A GBA mutation was the only significant genetic factor associated with MDRS change (ß = -2.51, p = 0.009). GBA mutation carriers had a more pronounced post-operative MDRS decline (3.2 ± 5.1) than patients with LRRK2 (0.9 ± 4.8), PRKN (0.5 ± 2.7) or controls (1.4 ± 4.4). The motor response to DBS was similar between groups. CONCLUSION: GBA mutations are associated with early cognitive decline following STN-DBS. Neuropsychological assessment and discussions on the benefit/risk ratio of DBS are particularly important for this population.


Asunto(s)
Disfunción Cognitiva , Estimulación Encefálica Profunda , Progresión de la Enfermedad , Glucosilceramidasa/genética , Enfermedad de Parkinson , Núcleo Subtalámico , Anciano , Disfunción Cognitiva/etiología , Disfunción Cognitiva/genética , Disfunción Cognitiva/fisiopatología , Estimulación Encefálica Profunda/efectos adversos , Femenino , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Masculino , Persona de Mediana Edad , Pruebas Neuropsicológicas , Enfermedad de Parkinson/complicaciones , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/terapia , Estudios Retrospectivos , Núcleo Subtalámico/cirugía , Ubiquitina-Proteína Ligasas/genética
5.
Genet Med ; 22(11): 1851-1862, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32713943

RESUMEN

PURPOSE: Pathogenic variants in STUB1 were initially described in autosomal recessive spinocerebellar ataxia type 16 and dominant cerebellar ataxia with cerebellar cognitive dysfunction (SCA48). METHODS: We analyzed a large series of 440 index cerebellar ataxia cases, mostly with dominant inheritance. RESULTS: STUB1 variants were detected in 50 patients. Age at onset and severity were remarkably variable. Cognitive impairment, predominantly frontal syndrome, was observed in 54% of STUB1 variant carriers, including five families with Huntington or frontotemporal dementia disease-like phenotypes associated with ataxia, while no STUB1 variant was found in 115 patients with frontotemporal dementia. We report neuropathological findings of a STUB1 heterozygous patient, showing massive loss of Purkinje cells in the vermis and major loss in the cerebellar hemispheres without atrophy of the pons, hippocampus, or cerebral cortex. This screening of STUB1 variants revealed new features: (1) the majority of patients were women (70%) and (2) "second hits" in AFG3L2, PRKCG, and TBP were detected in three families suggesting synergic effects. CONCLUSION: Our results reveal an unexpectedly frequent (7%) implication of STUB1 among dominantly inherited cerebellar ataxias, and suggest that the penetrance of STUB1 variants could be modulated by other factors, including sex and variants in other ataxia-related genes.


Asunto(s)
Ataxia Cerebelosa , Disfunción Cognitiva , Ataxias Espinocerebelosas , Proteasas ATP-Dependientes , ATPasas Asociadas con Actividades Celulares Diversas , Ataxia , Ataxia Cerebelosa/genética , Femenino , Humanos , Masculino , Ataxias Espinocerebelosas/genética , Ubiquitina-Proteína Ligasas
6.
Lancet Neurol ; 18(12): 1091-1102, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31701892

RESUMEN

BACKGROUND: Genome-wide association studies (GWAS) in Parkinson's disease have increased the scope of biological knowledge about the disease over the past decade. We aimed to use the largest aggregate of GWAS data to identify novel risk loci and gain further insight into the causes of Parkinson's disease. METHODS: We did a meta-analysis of 17 datasets from Parkinson's disease GWAS available from European ancestry samples to nominate novel loci for disease risk. These datasets incorporated all available data. We then used these data to estimate heritable risk and develop predictive models of this heritability. We also used large gene expression and methylation resources to examine possible functional consequences as well as tissue, cell type, and biological pathway enrichments for the identified risk factors. Additionally, we examined shared genetic risk between Parkinson's disease and other phenotypes of interest via genetic correlations followed by Mendelian randomisation. FINDINGS: Between Oct 1, 2017, and Aug 9, 2018, we analysed 7·8 million single nucleotide polymorphisms in 37 688 cases, 18 618 UK Biobank proxy-cases (ie, individuals who do not have Parkinson's disease but have a first degree relative that does), and 1·4 million controls. We identified 90 independent genome-wide significant risk signals across 78 genomic regions, including 38 novel independent risk signals in 37 loci. These 90 variants explained 16-36% of the heritable risk of Parkinson's disease depending on prevalence. Integrating methylation and expression data within a Mendelian randomisation framework identified putatively associated genes at 70 risk signals underlying GWAS loci for follow-up functional studies. Tissue-specific expression enrichment analyses suggested Parkinson's disease loci were heavily brain-enriched, with specific neuronal cell types being implicated from single cell data. We found significant genetic correlations with brain volumes (false discovery rate-adjusted p=0·0035 for intracranial volume, p=0·024 for putamen volume), smoking status (p=0·024), and educational attainment (p=0·038). Mendelian randomisation between cognitive performance and Parkinson's disease risk showed a robust association (p=8·00 × 10-7). INTERPRETATION: These data provide the most comprehensive survey of genetic risk within Parkinson's disease to date, to the best of our knowledge, by revealing many additional Parkinson's disease risk loci, providing a biological context for these risk factors, and showing that a considerable genetic component of this disease remains unidentified. These associations derived from European ancestry datasets will need to be followed-up with more diverse data. FUNDING: The National Institute on Aging at the National Institutes of Health (USA), The Michael J Fox Foundation, and The Parkinson's Foundation (see appendix for full list of funding sources).


Asunto(s)
Bases de Datos Genéticas , Sitios Genéticos/genética , Predisposición Genética a la Enfermedad/genética , Estudio de Asociación del Genoma Completo/métodos , Enfermedad de Parkinson/genética , Predisposición Genética a la Enfermedad/epidemiología , Humanos , Enfermedad de Parkinson/diagnóstico , Enfermedad de Parkinson/epidemiología , Factores de Riesgo
7.
Dis Model Mech ; 12(1)2019 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-30559154

RESUMEN

Perturbation of protein homeostasis and aggregation of misfolded proteins is a major cause of many human diseases. A hallmark of the neurodegenerative disease spinocerebellar ataxia type 7 (SCA7) is the intranuclear accumulation of mutant, misfolded ataxin-7 (polyQ-ATXN7). Here, we show that endogenous ATXN7 is modified by SUMO proteins, thus also suggesting a physiological role for this modification under conditions of proteotoxic stress caused by the accumulation of polyQ-ATXN7. Co-immunoprecipitation experiments, immunofluorescence microscopy and proximity ligation assays confirmed the colocalization and interaction of polyQ-ATXN7 with SUMO2 in cells. Moreover, upon inhibition of the proteasome, both endogenous SUMO2/3 and the RNF4 ubiquitin ligase surround large polyQ-ATXN7 intranuclear inclusions. Overexpression of RNF4 and/or SUMO2 significantly decreased levels of polyQ-ATXN7 and, upon proteasomal inhibition, led to a marked increase in the polyubiquitination of polyQ-ATXN7. This provides a mechanism for the clearance of polyQ-ATXN7 from affected cells that involves the recruitment of RNF4 by SUMO2/3-modified polyQ-ATXN7, thus leading to its ubiquitination and proteasomal degradation. In a SCA7 knock-in mouse model, we similarly observed colocalization of SUMO2/3 with polyQ-ATXN7 inclusions in the cerebellum and retina. Furthermore, we detected accumulation of SUMO2/3 high-molecular-mass species in the cerebellum of SCA7 knock-in mice, compared with their wild-type littermates, and changes in SUMO-related transcripts. Immunohistochemical analysis showed the accumulation of SUMO proteins and RNF4 in the cerebellum of SCA7 patients. Taken together, our results show that the SUMO pathway contributes to the clearance of aggregated ATXN7 and suggest that its deregulation might be associated with SCA7 disease progression.


Asunto(s)
Ataxina-7/metabolismo , Proteínas Nucleares/metabolismo , Pliegue de Proteína , Proteolisis , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Ataxias Espinocerebelosas/metabolismo , Sumoilación , Factores de Transcripción/metabolismo , Animales , Cerebelo/metabolismo , Niño , Modelos Animales de Enfermedad , Células HEK293 , Células HeLa , Humanos , Cuerpos de Inclusión/metabolismo , Células MCF-7 , Ratones , Persona de Mediana Edad , Mutación/genética , Proteína de la Leucemia Promielocítica/metabolismo , Inhibidores de Proteasoma/farmacología , Agregado de Proteínas/efectos de los fármacos , Pliegue de Proteína/efectos de los fármacos , Proteolisis/efectos de los fármacos , Ataxias Espinocerebelosas/patología , Sumoilación/efectos de los fármacos , Ubiquitina/metabolismo
8.
Glia ; 66(8): 1736-1751, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29665074

RESUMEN

Neuroinflammation and mitochondrial dysfunction, key mechanisms in the pathogenesis of Parkinson's disease (PD), are usually explored independently. Loss-of-function mutations of PARK2 and PARK6, encoding the E3 ubiquitin protein ligase Parkin and the mitochondrial serine/threonine kinase PINK1, account for a large proportion of cases of autosomal recessive early-onset PD. PINK1 and Parkin regulate mitochondrial quality control and have been linked to the modulation of innate immunity pathways. We report here an exacerbation of NLRP3 inflammasome activation by specific inducers in microglia and bone marrow-derived macrophages from Park2-/- and Pink1-/- mice. The caspase 1-dependent release of IL-1ß and IL-18 was, therefore, enhanced in Park2-/- and Pink1-/- cells. This defect was confirmed in blood-derived macrophages from patients with PARK2 mutations and was reversed by MCC950, which specifically inhibits NLRP3 inflammasome complex formation. Enhanced NLRP3 signaling in Parkin-deficient cells was accompanied by a lack of induction of A20, a well-known negative regulator of the NF-κB pathway recently shown to attenuate NLRP3 inflammasome activity. We also found an inverse correlation between A20 abundance and IL-1ß release, in human macrophages challenged with NLRP3 inflammasome inducers. Overall, our observations suggest that the A20/NLRP3-inflammasome axis participates in the pathogenesis of PARK2-linked PD, paving the way for the exploration of its potential as a biomarker and treatment target.


Asunto(s)
Retroalimentación Fisiológica/fisiología , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Ubiquitina-Proteína Ligasas/deficiencia , Adulto , Humanos , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Microglía/metabolismo , Persona de Mediana Edad , Mitocondrias/metabolismo , FN-kappa B/metabolismo
9.
Neurol Genet ; 4(2): e223, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29577077

RESUMEN

OBJECTIVE: To identify the clinical characteristics and genetic etiology of a family affected with hereditary spastic paraplegia (HSP). METHODS: Clinical, genetic, and functional analyses involving genome-wide linkage coupled to whole-exome sequencing in a consanguineous family with complicated HSP. RESULTS: A homozygous missense mutation was identified in the ACO2 gene (c.1240T>G p.Phe414Val) that segregated with HSP complicated by intellectual disability and microcephaly. Lymphoblastoid cell lines of homozygous carrier patients revealed significantly decreased activity of the mitochondrial aconitase enzyme and defective mitochondrial respiration. ACO2 encodes mitochondrial aconitase, an essential enzyme in the Krebs cycle. Recessive mutations in this gene have been previously associated with cerebellar ataxia. CONCLUSIONS: Our findings nominate ACO2 as a disease-causing gene for autosomal recessive complicated HSP and provide further support for the central role of mitochondrial defects in the pathogenesis of HSP.

10.
Neuropathology ; 38(2): 113-124, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29218765

RESUMEN

Autophagy, the major pathway for protein turnover, is critical to maintain cellular homeostasis and has been implicated in neurodegenerative diseases. The aim of this research was to analyze the expression of autophagy markers in postmortem brains from Machado-Joseph disease (MJD) patients. The expression of autophagy markers in the cerebellum and the oculomotor nucleus from MJD patients and age-matched controls with no signs of neuropathology was inspected postmortem by immunohistochemistry (IHC) and Western blot. Furthermore, autophagy was examined by means of transmission electron microscopy (TEM). Western blot and IHC revealed nuclear accumulation of misfolded ataxin-3 (ATXN3) and the presence of ubiquitin- and p62-positive aggregates in MJD patients as compared to controls. Moreover, the autophagic proteins, autophagy-related gene (Atg) protein (ATG)-7, ATG-12, ATG16L2 and autophagosomal microtubule-associated protein light chain 3 (LC3) were significantly increased in MJD brains relative to controls, while beclin-1 levels were reduced in MJD patients. Increase in the levels of lysosomal-associated membrane protein 2 (LAMP-2) and of the endosomal markers (Rab7 and Rab1A) were observed in MJD patients relatively to controls. In addition, these findings were further confirmed by TEM in brain tissue where large vesicles accumulating electron-dense materials were highly enriched in MJD patients. Postmortem brains with MJD exhibit increased markers of autophagy relative to age-matched control brains, therefore suggesting strong dysregulation of autophagy that may have an important role in the course of MJD pathogenesis.


Asunto(s)
Autofagia , Cerebelo/metabolismo , Enfermedad de Machado-Joseph/metabolismo , Complejo Nuclear Oculomotor/metabolismo , Adulto , Ataxina-3/metabolismo , Beclina-1/metabolismo , Biomarcadores/metabolismo , Moléculas de Adhesión Celular Neuronal/metabolismo , Endosomas/metabolismo , Femenino , Proteínas Ligadas a GPI/metabolismo , Humanos , Lisosomas/metabolismo , Masculino , Proteínas Asociadas a Microtúbulos/metabolismo , Persona de Mediana Edad , Proteínas Proto-Oncogénicas c-myc/metabolismo , Sirolimus/metabolismo , Ubiquitina/metabolismo
11.
J Clin Invest ; 127(11): 3923-3936, 2017 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-28945198

RESUMEN

Netrin-1 is a secreted protein that was first identified 20 years ago as an axon guidance molecule that regulates midline crossing in the CNS. It plays critical roles in various tissues throughout development and is implicated in tumorigenesis and inflammation in adulthood. Despite extensive studies, no inherited human disease has been directly associated with mutations in NTN1, the gene coding for netrin-1. Here, we have identified 3 mutations in exon 7 of NTN1 in 2 unrelated families and 1 sporadic case with isolated congenital mirror movements (CMM), a disorder characterized by involuntary movements of one hand that mirror intentional movements of the opposite hand. Given the diverse roles of netrin-1, the absence of manifestations other than CMM in NTN1 mutation carriers was unexpected. Using multimodal approaches, we discovered that the anatomy of the corticospinal tract (CST) is abnormal in patients with NTN1-mutant CMM. When expressed in HEK293 or stable HeLa cells, the 3 mutated netrin-1 proteins were almost exclusively detected in the intracellular compartment, contrary to WT netrin-1, which is detected in both intracellular and extracellular compartments. Since netrin-1 is a diffusible extracellular cue, the pathophysiology likely involves its loss of function and subsequent disruption of axon guidance, resulting in abnormal decussation of the CST.


Asunto(s)
Trastornos del Movimiento/genética , Netrina-1/genética , Anciano de 80 o más Años , Secuencia de Aminoácidos , Animales , Secuencia Conservada , Femenino , Frecuencia de los Genes , Estudios de Asociación Genética , Células HEK293 , Células HeLa , Heterocigoto , Humanos , Masculino , Ratones , Mutación Missense , Linaje , Eliminación de Secuencia
12.
JAMA Neurol ; 74(7): 780-792, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28586827

RESUMEN

Importance: Recent genome-wide association studies (GWAS) and pathway analyses supported long-standing observations of an association between immune-mediated diseases and Parkinson disease (PD). The post-GWAS era provides an opportunity for cross-phenotype analyses between different complex phenotypes. Objectives: To test the hypothesis that there are common genetic risk variants conveying risk of both PD and autoimmune diseases (ie, pleiotropy) and to identify new shared genetic variants and their pathways by applying a novel statistical framework in a genome-wide approach. Design, Setting, and Participants: Using the conjunction false discovery rate method, this study analyzed GWAS data from a selection of archetypal autoimmune diseases among 138 511 individuals of European ancestry and systemically investigated pleiotropy between PD and type 1 diabetes, Crohn disease, ulcerative colitis, rheumatoid arthritis, celiac disease, psoriasis, and multiple sclerosis. NeuroX data (6927 PD cases and 6108 controls) were used for replication. The study investigated the biological correlation between the top loci through protein-protein interaction and changes in the gene expression and methylation levels. The dates of the analysis were June 10, 2015, to March 4, 2017. Main Outcomes and Measures: The primary outcome was a list of novel loci and their pathways involved in PD and autoimmune diseases. Results: Genome-wide conjunctional analysis identified 17 novel loci at false discovery rate less than 0.05 with overlap between PD and autoimmune diseases, including known PD loci adjacent to GAK, HLA-DRB5, LRRK2, and MAPT for rheumatoid arthritis, ulcerative colitis and Crohn disease. Replication confirmed the involvement of HLA, LRRK2, MAPT, TRIM10, and SETD1A in PD. Among the novel genes discovered, WNT3, KANSL1, CRHR1, BOLA2, and GUCY1A3 are within a protein-protein interaction network with known PD genes. A subset of novel loci was significantly associated with changes in methylation or expression levels of adjacent genes. Conclusions and Relevance: The study findings provide novel mechanistic insights into PD and autoimmune diseases and identify a common genetic pathway between these phenotypes. The results may have implications for future therapeutic trials involving anti-inflammatory agents.


Asunto(s)
Enfermedades Autoinmunes/genética , Pleiotropía Genética , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Enfermedad de Parkinson/genética , Artritis Reumatoide/genética , Enfermedad Celíaca/genética , Colitis Ulcerosa/genética , Enfermedad de Crohn/genética , Diabetes Mellitus Tipo 1/genética , Sitios Genéticos , Humanos , Esclerosis Múltiple/genética , Psoriasis/genética , Factores de Riesgo
13.
Nat Genet ; 49(4): 511-514, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28250454

RESUMEN

Brain malformations involving the corpus callosum are common in children with developmental disabilities. We identified DCC mutations in four families and five sporadic individuals with isolated agenesis of the corpus callosum (ACC) without intellectual disability. DCC mutations result in variable dominant phenotypes with decreased penetrance, including mirror movements and ACC associated with a favorable developmental prognosis. Possible phenotypic modifiers include the type and location of mutation and the sex of the individual.


Asunto(s)
Agenesia del Cuerpo Calloso/genética , Discapacidades del Desarrollo/genética , Mutación/genética , Receptores de Superficie Celular/genética , Proteínas Supresoras de Tumor/genética , Anomalías Múltiples/genética , Encéfalo/patología , Cuerpo Calloso/patología , Receptor DCC , Familia , Femenino , Humanos , Masculino , Malformaciones del Sistema Nervioso/genética , Células-Madre Neurales/patología , Penetrancia , Fenotipo
14.
Neurology ; 88(12): 1114-1119, 2017 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-28202696

RESUMEN

OBJECTIVE: Polyglutamine (PolyQ) diseases are dominantly transmitted neurologic disorders, caused by coding and expanded CAG trinucleotide repeats. Cancer was reported retrospectively to be rare in patients with PolyQ diseases and we aimed to investigate its prevalence in France. METHODS: Consecutive patients with Huntington disease (HD) and spinocerebellar ataxia (SCA) were questioned about cancer, cardiovascular diseases, and related risk factors in 4 university hospitals in Paris, Toulouse, Strasbourg, and Montpellier. Standardized incidence ratios (SIR), based on age- and sex-adjusted rate of the French population, were assessed for different types of cancer. RESULTS: We questioned 372 patients with HD and 134 patients with SCA. SIR showed significantly reduced risk of cancer in HD: 23 observed cases vs 111.05 expected ones (SIR 0.21, 95% confidence interval [CI] 0.13-0.31), as well as in SCA: 7 observed cases vs 34.73 expected (SIR 0.23, 95% CI 0.08-0.42). This was surprising since risk behavior for cancer was increased in these patients, with significantly greater tobacco and alcohol consumption in patients with HD vs patients with SCA (p < 0.0056). There was no association between CAG repeat size and cancer or cardiovascular disease. However, in patients with HD, skin cancers were more frequent than expected (5 vs 0.98, SIR 5.11, 95% CI 1.65-11.95). CONCLUSIONS: There was a decreased cancer rate in PolyQ diseases despite high incidence of risk factors. Intriguingly, skin cancer incidence was higher, suggesting a crosstalk between neurodegeneration and skin tumorigenesis.


Asunto(s)
Predisposición Genética a la Enfermedad/epidemiología , Enfermedad de Huntington/epidemiología , Neoplasias/epidemiología , Péptidos/genética , Ataxias Espinocerebelosas/epidemiología , Europa (Continente)/epidemiología , Femenino , Humanos , Enfermedad de Huntington/genética , Masculino , Persona de Mediana Edad , Neoplasias/genética , Estudios Retrospectivos , Factores de Riesgo , Ataxias Espinocerebelosas/genética
15.
Cell Rep ; 16(1): 79-91, 2016 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-27320912

RESUMEN

A CAPN1 missense mutation in Parson Russell Terrier dogs is associated with spinocerebellar ataxia. We now report that homozygous or heterozygous CAPN1-null mutations in humans result in cerebellar ataxia and limb spasticity in four independent pedigrees. Calpain-1 knockout (KO) mice also exhibit a mild form of ataxia due to abnormal cerebellar development, including enhanced neuronal apoptosis, decreased number of cerebellar granule cells, and altered synaptic transmission. Enhanced apoptosis is due to absence of calpain-1-mediated cleavage of PH domain and leucine-rich repeat protein phosphatase 1 (PHLPP1), which results in inhibition of the Akt pro-survival pathway in developing granule cells. Injection of neonatal mice with the indirect Akt activator, bisperoxovanadium, or crossing calpain-1 KO mice with PHLPP1 KO mice prevented increased postnatal cerebellar granule cell apoptosis and restored granule cell density and motor coordination in adult mice. Thus, mutations in CAPN1 are an additional cause of ataxia in mammals, including humans.


Asunto(s)
Calpaína/genética , Ataxia Cerebelosa/genética , Cerebelo/embriología , Cerebelo/metabolismo , Envejecimiento/metabolismo , Secuencia de Aminoácidos , Animales , Animales Recién Nacidos , Apoptosis , Calpaína/química , Calpaína/metabolismo , Recuento de Células , Ataxia Cerebelosa/patología , Ataxia Cerebelosa/fisiopatología , Cerebelo/patología , Cerebelo/fisiopatología , Activación Enzimática , Femenino , Humanos , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Discapacidad Intelectual/fisiopatología , Masculino , Ratones Noqueados , Actividad Motora , Espasticidad Muscular/genética , Espasticidad Muscular/patología , Espasticidad Muscular/fisiopatología , Mutación/genética , Proteínas Nucleares/metabolismo , Atrofia Óptica/genética , Atrofia Óptica/patología , Atrofia Óptica/fisiopatología , Fosfoproteínas Fosfatasas/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Células de Purkinje/patología , Ataxias Espinocerebelosas/genética , Ataxias Espinocerebelosas/patología , Ataxias Espinocerebelosas/fisiopatología , Transmisión Sináptica
16.
Brain ; 139(Pt 6): 1723-34, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27016404

RESUMEN

The most common form of autosomal recessive hereditary spastic paraplegia is caused by mutations in the SPG11/KIAA1840 gene on chromosome 15q. The nature of the vast majority of SPG11 mutations found to date suggests a loss-of-function mechanism of the encoded protein, spatacsin. The SPG11 phenotype is, in most cases, characterized by a progressive spasticity with neuropathy, cognitive impairment and a thin corpus callosum on brain MRI. Full neuropathological characterization has not been reported to date despite the description of >100 SPG11 mutations. We describe here the clinical and pathological features observed in two unrelated females, members of genetically ascertained SPG11 families originating from Belgium and Italy, respectively. We confirm the presence of lesions of motor tracts in medulla oblongata and spinal cord associated with other lesions of the central nervous system. Interestingly, we report for the first time pathological hallmarks of SPG11 in neurons that include intracytoplasmic granular lysosome-like structures mainly in supratentorial areas, and others in subtentorial areas that are partially reminiscent of those observed in amyotrophic lateral sclerosis, such as ubiquitin and p62 aggregates, except that they are never labelled with anti-TDP-43 or anti-cystatin C. The neuropathological overlap with amyotrophic lateral sclerosis, associated with some shared clinical manifestations, opens up new fields of investigation in the physiopathological continuum of motor neuron degeneration.


Asunto(s)
Esclerosis Amiotrófica Lateral/patología , Neuronas Motoras/patología , Degeneración Nerviosa/patología , Paraplejía Espástica Hereditaria/patología , Adulto , Encéfalo/patología , Femenino , Ganglios Espinales/patología , Humanos , Lisosomas/ultraestructura , Masculino , Bulbo Raquídeo/patología , Persona de Mediana Edad , Mutación , Proteínas/genética , Paraplejía Espástica Hereditaria/diagnóstico , Paraplejía Espástica Hereditaria/diagnóstico por imagen , Médula Espinal/patología
17.
BMC Neurosci ; 17: 6, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26831335

RESUMEN

BACKGROUND: Bilateral subthalamic nucleus deep brain stimulation (STN-DBS) of parkinson's disease (PD) patients has demonstrated to improve motor performance and to reduce dopa-induced dyskinesia. An association between the occurrence of dyskinesias and LRRK2 (leucine-rich repeat kinase 2) G2019S gene mutations has recently been suggested. The aim of this study is to discover the impact of the G2019S mutation (with high incidence in the authors' native Algeria) on the symptom response of PD in patients who underwent STN-DBS. METHODS: We carried out a comparative statistical study for the clinical evaluation and neuropsychological assessment of 27 Algerian PD STN-DBS patients, both G2019S mutation carriers (MC) and non-carriers (NC). A multiple correspondence analysis (MCA) was then conducted to compare the results with those from groups of individuals with similar modalities. RESULTS: The MCA revealed that MC and NC PD patients showed two different patterns of clinical evaluations. The group of idiopathic patients showed some differences compared to the clinical evaluations, depending on gender. No association was found between the G2019S mutation and the Mini Mental State Examination scores (MMSE), and MC patients appeared more susceptible to dyskinesia than NC patients. In NC patients, we found two cases with Parkin mutations who had a different "honeymoon" period and different initial symptoms. The results showed considerable improvement of motor unified parkinson's disease rating scale III (UPDRS-III) in a situation of stimulation without medication in the MC patients with a percentage of improvement (51.1 %) over the required 30 % compared to the NC patients (25.5 %). The same result was observed for the Schwab and England's activities of daily living scale (S and E scale), which thus demonstrated a greater effectiveness of DBS for MC patients than for NC patients. However, the Hoehn and Yahr scale (H and Y Scale) showed the same significance in a situation of stimulation for MC and NC patients. In this later group, the best scores of UPDRS-III were observed for patients with the Parkin mutation before they underwent surgery. CONCLUSIONS: This study shows that surgical treatment probably has a more significant impact on LRRK2 G2019S MC than on idiopathic patients.


Asunto(s)
Estimulación Encefálica Profunda , Mutación , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/terapia , Proteínas Serina-Treonina Quinasas/genética , Núcleo Subtalámico/fisiopatología , Femenino , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Masculino , Persona de Mediana Edad , Enfermedad de Parkinson/cirugía , Resultado del Tratamiento
18.
J Natl Cancer Inst ; 108(3)2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26683220

RESUMEN

BACKGROUND: Melanoma incidence is higher in patients affected by Parkinson's disease (PD) and vice versa, but the genetic link shared by both diseases is unknown. As PARK2 is both a tumor suppressor gene and frequently mutated in young onset PD, we evaluated the role of PARK2 in melanoma predisposition and progression. METHODS: An in-depth PARK2 gene dosage analysis and sequencing was performed on 512 French case patients and 562 healthy control patients, as well as sporadic tumors and melanoma cell lines. The frequency of genetic alterations was compared between case patients and control patients using two-sided Fisher's exact tests and odds ratio (OR) calculations. We used western blotting to determine PARKIN expression in melanocytes and melanoma cell lines and transfection followed by clonogenic assays to evaluate the effect of PARKIN expression on cellular proliferation. All statistical tests were two-sided. RESULTS: Germline PARK2 mutations (including copy number variations, splicing, and putative deleterious missense mutations) were present in 25 case patients but only four control patients (OR = 3.95, 95% confidence interval = 1.34 to 15.75). Copy number variations (CNVs) and loss of heterozygosity were present in 60% and 74%, respectively, of primary tumors. PARKIN protein was expressed in melanocytes but not in most melanoma cell lines, and its expression decreased following melanocyte transformation by oncogenic NRAS. Re-expression of PARKIN in melanoma cell lines resulted in a drastic reduction of cell proliferation and inhibition of PARKIN in melanocytes stimulated their proliferation. CONCLUSION: Our results show an important role for PARK2 as a tumor suppressor both in melanoma predisposition and progression, which could explain the epidemiological association of these diseases.


Asunto(s)
Silenciador del Gen , Mutación de Línea Germinal , Melanoma/genética , Enfermedad de Parkinson/genética , Neoplasias Cutáneas/genética , Proteínas Supresoras de Tumor , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Adulto , Anciano , Western Blotting , Estudios de Casos y Controles , Línea Celular Tumoral , Proliferación Celular , Variaciones en el Número de Copia de ADN , ADN de Neoplasias/análisis , Femenino , Mutación del Sistema de Lectura , Francia/epidemiología , Dosificación de Gen , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Melanocitos/metabolismo , Melanoma/epidemiología , Melanoma/metabolismo , Persona de Mediana Edad , Mutación Missense , Oportunidad Relativa , Enfermedad de Parkinson/epidemiología , Enfermedad de Parkinson/metabolismo , Empalme de Proteína , Riesgo , Análisis de Secuencia de ADN , Neoplasias Cutáneas/epidemiología , Neoplasias Cutáneas/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
20.
Neurology ; 85(15): 1283-92, 2015 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-26354989

RESUMEN

OBJECTIVES: We aim to clarify the pathogenic role of intermediate size repeat expansions of SCA2, SCA3, SCA6, and SCA17 as risk factors for idiopathic Parkinson disease (PD). METHODS: We invited researchers from the Genetic Epidemiology of Parkinson's Disease Consortium to participate in the study. There were 12,346 cases and 8,164 controls genotyped, for a total of 4 repeats within the SCA2, SCA3, SCA6, and SCA17 genes. Fixed- and random-effects models were used to estimate the summary risk estimates for the genes. We investigated between-study heterogeneity and heterogeneity between different ethnic populations. RESULTS: We did not observe any definite pathogenic repeat expansions for SCA2, SCA3, SCA6, and SCA17 genes in patients with idiopathic PD from Caucasian and Asian populations. Furthermore, overall analysis did not reveal any significant association between intermediate repeats and PD. The effect estimates (odds ratio) ranged from 0.93 to 1.01 in the overall cohort for the SCA2, SCA3, SCA6, and SCA17 loci. CONCLUSIONS: Our study did not support a major role for definite pathogenic repeat expansions in SCA2, SCA3, SCA6, and SCA17 genes for idiopathic PD. Thus, results of this large study do not support diagnostic screening of SCA2, SCA3, SCA6, and SCA17 gene repeats in the common idiopathic form of PD. Likewise, this largest multicentered study performed to date excludes the role of intermediate repeats of these genes as a risk factor for PD.


Asunto(s)
Frecuencia de los Genes/genética , Predisposición Genética a la Enfermedad , Enfermedad de Parkinson/genética , Péptidos/genética , Expansión de Repetición de Trinucleótido/genética , Anciano , Ataxinas/genética , Ataxinas/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Enfermedad de Parkinson/epidemiología , Fenotipo , Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA