Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Carles Vilariño-Güell is active.

Publication


Featured researches published by Carles Vilariño-Güell.


American Journal of Human Genetics | 2011

VPS35 Mutations in Parkinson Disease

Carles Vilariño-Güell; Christian Wider; Owen A. Ross; Justus C. Dachsel; Jennifer M. Kachergus; Sarah Lincoln; Alexandra I. Soto-Ortolaza; Stephanie A. Cobb; Greggory J. Wilhoite; Justin A. Bacon; Behrouz Bahareh Behrouz; Heather L. Melrose; Emna Hentati; Andreas Puschmann; Daniel M. Evans; Elizabeth Conibear; Wyeth W. Wasserman; Jan O. Aasly; Pierre Burkhard; Ruth Djaldetti; Joseph Ghika; F. Hentati; Anna Krygowska-Wajs; Timothy Lynch; Eldad Melamed; Alex Rajput; Ali H. Rajput; Alessandra Solida; Ruey-Meei Wu; Ryan J. Uitti

The identification of genetic causes for Mendelian disorders has been based on the collection of multi-incident families, linkage analysis, and sequencing of genes in candidate intervals. This study describes the application of next-generation sequencing technologies to a Swiss kindred presenting with autosomal-dominant, late-onset Parkinson disease (PD). The family has tremor-predominant dopa-responsive parkinsonism with a mean onset of 50.6 ± 7.3 years. Exome analysis suggests that an aspartic-acid-to-asparagine mutation within vacuolar protein sorting 35 (VPS35 c.1858G>A; p.Asp620Asn) is the genetic determinant of disease. VPS35 is a central component of the retromer cargo-recognition complex, is critical for endosome-trans-golgi trafficking and membrane-protein recycling, and is evolutionarily highly conserved. VPS35 c.1858G>A was found in all affected members of the Swiss kindred and in three more families and one patient with sporadic PD, but it was not observed in 3,309 controls. Further sequencing of familial affected probands revealed only one other missense variant, VPS35 c.946C>T; (p.Pro316Ser), in a pedigree with one unaffected and two affected carriers, and thus the pathogenicity of this mutation remains uncertain. Retromer-mediated sorting and transport is best characterized for acid hydrolase receptors. However, the complex has many types of cargo and is involved in a diverse array of biologic pathways from developmental Wnt signaling to lysosome biogenesis. Our study implicates disruption of VPS35 and retromer-mediated trans-membrane protein sorting, rescue, and recycling in the neurodegenerative process leading to PD.


Movement Disorders | 2013

Alpha-synuclein p.H50Q, a novel pathogenic mutation for Parkinson's disease.

Silke Appel-Cresswell; Carles Vilariño-Güell; Holly E. Sherman; Irene Yu; Brinda Shah; David Weir; Christina Thompson; Chelsea Szu-Tu; Joanne Trinh; Jan O. Aasly; Alex Rajput; Ali H. Rajput; A. J. Stoessl; Matthew J. Farrer

Alpha‐synuclein plays a central role in the pathophysiology of Parkinsons disease. Three missense mutations in SNCA, the gene encoding alpha‐synuclein, as well as genomic multiplications have been identified as causes for autosomal‐dominantly inherited Parkinsonism.


Nature Genetics | 2009

DCTN1 mutations in Perry syndrome

Matthew J. Farrer; Mary M. Hulihan; Jennifer M. Kachergus; Justus C. Dachsel; A. Jon Stoessl; Linda L. Grantier; Susan Calne; Donald B. Calne; Bernard Lechevalier; Françoise Chapon; Yoshio Tsuboi; Tatsuo Yamada; Ludwig Gutmann; Bulent Elibol; Kailash P. Bhatia; Christian Wider; Carles Vilariño-Güell; Owen A. Ross; L. Brown; Monica Castanedes-Casey; Dennis W. Dickson; Zbigniew K. Wszolek

Perry syndrome consists of early-onset parkinsonism, depression, severe weight loss and hypoventilation, with brain pathology characterized by TDP-43 immunostaining. We carried out genome-wide linkage analysis and identified five disease-segregating mutations affecting the CAP-Gly domain of dynactin (encoded by DCTN1) in eight families with Perry syndrome; these mutations diminish microtubule binding and lead to intracytoplasmic inclusions. Our findings show that DCTN1 mutations, previously associated with motor neuron disease, can underlie the selective vulnerability of other neuronal populations in distinct neurodegenerative disorders.


American Journal of Human Genetics | 2011

Translation Initiator EIF4G1 Mutations in Familial Parkinson Disease

Marie Christine Chartier-Harlin; Justus C. Dachsel; Carles Vilariño-Güell; Sarah Lincoln; Frédéric Leprêtre; Mary M. Hulihan; Jennifer M. Kachergus; Austen J. Milnerwood; Lucia Tapia; Mee Sook Song; Emilie Le Rhun; Eugénie Mutez; Lydie Larvor; Aurélie Duflot; Christel Vanbesien-Mailliot; Alexandre Kreisler; Owen A. Ross; Kenya Nishioka; Alexandra I. Soto-Ortolaza; Stephanie A. Cobb; Heather L. Melrose; Bahareh Behrouz; Brett H. Keeling; Justin A. Bacon; Emna Hentati; Williams L; Akiko Yanagiya; Nahum Sonenberg; Paul J. Lockhart; Abba C. Zubair

Genome-wide analysis of a multi-incident family with autosomal-dominant parkinsonism has implicated a locus on chromosomal region 3q26-q28. Linkage and disease segregation is explained by a missense mutation c.3614G>A (p.Arg1205His) in eukaryotic translation initiation factor 4-gamma (EIF4G1). Subsequent sequence and genotype analysis identified EIF4G1 c.1505C>T (p.Ala502Val), c.2056G>T (p.Gly686Cys), c.3490A>C (p.Ser1164Arg), c.3589C>T (p.Arg1197Trp) and c.3614G>A (p.Arg1205His) substitutions in affected subjects with familial parkinsonism and idiopathic Lewy body disease but not in control subjects. Despite different countries of origin, persons with EIF4G1 c.1505C>T (p.Ala502Val) or c.3614G>A (p.Arg1205His) mutations appear to share haplotypes consistent with ancestral founders. eIF4G1 p.Ala502Val and p.Arg1205His disrupt eIF4E or eIF3e binding, although the wild-type protein does not, and render mutant cells more vulnerable to reactive oxidative species. EIF4G1 mutations implicate mRNA translation initiation in familial parkinsonism and highlight a convergent pathway for monogenic, toxin and perhaps virally-induced Parkinson disease.


Lancet Neurology | 2011

Association of LRRK2 exonic variants with susceptibility to Parkinson's disease: A case-control study

Owen A. Ross; Alexandra I. Soto-Ortolaza; Michael G. Heckman; Jan O. Aasly; Nadine Abahuni; Grazia Annesi; Justin A. Bacon; Soraya Bardien; Maria Bozi; Alexis Brice; Laura Brighina; Christine Van Broeckhoven; Jonathan Carr; Marie Christine Chartier-Harlin; Efthimios Dardiotis; Dennis W. Dickson; Nancy N. Diehl; Alexis Elbaz; Carlo Ferrarese; Alessandro Ferraris; Brian K. Fiske; J. Mark Gibson; Rachel A. Gibson; Georgios M. Hadjigeorgiou; Nobutaka Hattori; John P. A. Ioannidis; Barbara Jasinska-Myga; Beom S. Jeon; Yun Joong Kim; Christine Klein

BACKGROUND Background The leucine-rich repeat kinase 2 gene (LRRK2) harbours highly penetrant mutations that are linked to familial parkinsonism. However, the extent of its polymorphic variability in relation to risk of Parkinsons disease (PD) has not been assessed systematically. We therefore assessed the frequency of LRRK2 exonic variants in individuals with and without PD, to investigate the role of the variants in PD susceptibility. METHODS LRRK2 was genotyped in patients with PD and controls from three series (white, Asian, and Arab-Berber) from sites participating in the Genetic Epidemiology of Parkinsons Disease Consortium. Genotyping was done for exonic variants of LRRK2 that were identified through searches of literature and the personal communications of consortium members. Associations with PD were assessed by use of logistic regression models. For variants that had a minor allele frequency of 0·5% or greater, single variant associations were assessed, whereas for rarer variants information was collapsed across variants. FINDINGS 121 exonic LRRK2 variants were assessed in 15 540 individuals: 6995 white patients with PD and 5595 controls, 1376 Asian patients and 962 controls, and 240 Arab-Berber patients and 372 controls. After exclusion of carriers of known pathogenic mutations, new independent risk associations were identified for polymorphic variants in white individuals (M1646T, odds ratio 1·43, 95% CI 1·15-1·78; p=0·0012) and Asian individuals (A419V, 2·27, 1·35-3·83; p=0·0011). A protective haplotype (N551K-R1398H-K1423K) was noted at a frequency greater than 5% in the white and Asian series, with a similar finding in the Arab-Berber series (combined odds ratio 0·82, 0·72-0·94; p=0·0043). Of the two previously reported Asian risk variants, G2385R was associated with disease (1·73, 1·20-2·49; p=0·0026), but no association was noted for R1628P (0·62, 0·36-1·07; p=0·087). In the Arab-Berber series, Y2189C showed potential evidence of risk association with PD (4·48, 1·33-15·09; p=0·012). INTERPRETATION The results for LRRK2 show that several rare and common genetic variants in the same gene can have independent effects on disease risk. LRRK2, and the pathway in which it functions, is important in the cause and pathogenesis of PD in a greater proportion of patients with this disease than previously believed. These results will help discriminate those patients who will benefit most from therapies targeted at LRRK2 pathogenic activity. FUNDING Michael J Fox Foundation and National Institutes of Health.


Human Molecular Genetics | 2014

DNAJC13 mutations in Parkinson disease

Carles Vilariño-Güell; Alex Rajput; Austen J. Milnerwood; Brinda Shah; Chelsea Szu-Tu; Joanne Trinh; Irene Yu; Lise N. Munsie; Lucia Tapia; Emil K. Gustavsson; Patrick Chou; Igor Tatarnikov; Daniel M. Evans; Frederick T. Pishotta; Mattia Volta; Dayne Beccano-Kelly; Christina Thompson; Michelle K. Lin; Holly E. Sherman; Heather Han; Bruce L. Guenther; Wyeth W. Wasserman; Virginie Bernard; Colin Ross; Silke Appel-Cresswell; A. Jon Stoessl; Christopher A. Robinson; Dennis W. Dickson; Owen A. Ross; Zbigniew K. Wszolek

A Saskatchewan multi-incident family was clinically characterized with Parkinson disease (PD) and Lewy body pathology. PD segregates as an autosomal-dominant trait, which could not be ascribed to any known mutation. DNA from three affected members was subjected to exome sequencing. Genome alignment, variant annotation and comparative analyses were used to identify shared coding mutations. Sanger sequencing was performed within the extended family and ethnically matched controls. Subsequent genotyping was performed in a multi-ethnic case-control series consisting of 2928 patients and 2676 control subjects from Canada, Norway, Taiwan, Tunisia, and the USA. A novel mutation in receptor-mediated endocytosis 8/RME-8 (DNAJC13 p.Asn855Ser) was found to segregate with disease. Screening of cases and controls identified four additional patients with the mutation, of which two had familial parkinsonism. All carriers shared an ancestral DNAJC13 p.Asn855Ser haplotype and claimed Dutch-German-Russian Mennonite heritage. DNAJC13 regulates the dynamics of clathrin coats on early endosomes. Cellular analysis shows that the mutation confers a toxic gain-of-function and impairs endosomal transport. DNAJC13 immunoreactivity was also noted within Lewy body inclusions. In late-onset disease which is most reminiscent of idiopathic PD subtle deficits in endosomal receptor-sorting/recycling are highlighted by the discovery of pathogenic mutations VPS35, LRRK2 and now DNAJC13. With this latest discovery, and from a neuronal perspective, a temporal and functional ecology is emerging that connects synaptic exo- and endocytosis, vesicular trafficking, endosomal recycling and the endo-lysosomal degradative pathway. Molecular deficits in these processes are genetically linked to the phenotypic spectrum of parkinsonism associated with Lewy body pathology.


PLOS Genetics | 2011

Genome-Wide Association Study Identifies Novel Restless Legs Syndrome Susceptibility Loci on 2p14 and 16q12.1

Juliane Winkelmann; Darina Czamara; Barbara Schormair; Franziska Knauf; Eva C. Schulte; Claudia Trenkwalder; Yves Dauvilliers; Olli Polo; Birgit Högl; Klaus Berger; Andrea Fuhs; Nadine Gross; Karin Stiasny-Kolster; Wolfgang H. Oertel; Cornelius G. Bachmann; Walter Paulus; Lan Xiong; Jacques Montplaisir; Guy A. Rouleau; Ingo Fietze; Jana Vávrová; David Kemlink; Karel Sonka; Sona Nevsimalova; Siong Chi Lin; Zbigniew K. Wszolek; Carles Vilariño-Güell; Matthew J. Farrer; Viola Gschliesser; Birgit Frauscher

Restless legs syndrome (RLS) is a sensorimotor disorder with an age-dependent prevalence of up to 10% in the general population above 65 years of age. Affected individuals suffer from uncomfortable sensations and an urge to move in the lower limbs that occurs mainly in resting situations during the evening or at night. Moving the legs or walking leads to an improvement of symptoms. Concomitantly, patients report sleep disturbances with consequences such as reduced daytime functioning. We conducted a genome-wide association study (GWA) for RLS in 922 cases and 1,526 controls (using 301,406 SNPs) followed by a replication of 76 candidate SNPs in 3,935 cases and 5,754 controls, all of European ancestry. Herein, we identified six RLS susceptibility loci of genome-wide significance, two of them novel: an intergenic region on chromosome 2p14 (rs6747972, P = 9.03 × 10−11, OR = 1.23) and a locus on 16q12.1 (rs3104767, P = 9.4 × 10−19, OR = 1.35) in a linkage disequilibrium block of 140 kb containing the 5′-end of TOX3 and the adjacent non-coding RNA BC034767.


Neurology | 2012

Large-scale replication and heterogeneity in Parkinson disease genetic loci

Manu Sharma; John P. A. Ioannidis; Jan O. Aasly; Grazia Annesi; Alexis Brice; Christine Van Broeckhoven; Lars Bertram; Maria Bozi; David Crosiers; Carl E Clarke; Maurizio F. Facheris; Matthew J. Farrer; Gaëtan Garraux; Suzana Gispert; Georg Auburger; Carles Vilariño-Güell; Georgios M. Hadjigeorgiou; Andrew A. Hicks; Nobutaka Hattori; Beom S. Jeon; Suzanne Lesage; Christina M. Lill; Juei Jueng Lin; Timothy Lynch; Peter Lichtner; Anthony E. Lang; Vincent Mok; Barbara Jasinska-Myga; George D. Mellick; Karen E. Morrison

Objective: Eleven genetic loci have reached genome-wide significance in a recent meta-analysis of genome-wide association studies in Parkinson disease (PD) based on populations of Caucasian descent. The extent to which these genetic effects are consistent across different populations is unknown. Methods: Investigators from the Genetic Epidemiology of Parkinsons Disease Consortium were invited to participate in the study. A total of 11 SNPs were genotyped in 8,750 cases and 8,955 controls. Fixed as well as random effects models were used to provide the summary risk estimates for these variants. We evaluated between-study heterogeneity and heterogeneity between populations of different ancestry. Results: In the overall analysis, single nucleotide polymorphisms (SNPs) in 9 loci showed significant associations with protective per-allele odds ratios of 0.78–0.87 (LAMP3, BST1, and MAPT) and susceptibility per-allele odds ratios of 1.14–1.43 (STK39, GAK, SNCA, LRRK2, SYT11, and HIP1R). For 5 of the 9 replicated SNPs there was nominally significant between-site heterogeneity in the effect sizes (I2 estimates ranged from 39% to 48%). Subgroup analysis by ethnicity showed significantly stronger effects for the BST1 (rs11724635) in Asian vs Caucasian populations and similar effects for SNCA, LRRK2, LAMP3, HIP1R, and STK39 in Asian and Caucasian populations, while MAPT rs2942168 and SYT11 rs34372695 were monomorphic in the Asian population, highlighting the role of population-specific heterogeneity in PD. Conclusion: Our study allows insight to understand the distribution of newly identified genetic factors contributing to PD and shows that large-scale evaluation in diverse populations is important to understand the role of population-specific heterogeneity. Neurology® 2012;79:659–667


Neurogenetics | 2010

LINGO1 and LINGO2 variants are associated with essential tremor and Parkinson disease.

Carles Vilariño-Güell; Christian Wider; Owen A. Ross; Barbara Jasinska-Myga; Jennifer M. Kachergus; Stephanie A. Cobb; Alexandra I. Soto-Ortolaza; Bahareh Behrouz; Michael G. Heckman; Nancy N. Diehl; Claudia M. Testa; Zbigniew K. Wszolek; Ryan J. Uitti; Joseph Jankovic; Elan D. Louis; Lorraine N. Clark; Alex Rajput; Matthew J. Farrer

Genetic variation in the leucine-rich repeat and Ig domain containing 1 gene (LINGO1) was recently associated with an increased risk of developing essential tremor (ET) and Parkinson disease (PD). Herein, we performed a comprehensive study of LINGO1 and its paralog LINGO2 in ET and PD by sequencing both genes in patients (ET, n = 95; PD, n = 96) and by examining haplotype-tagging single-nucleotide polymorphisms (tSNPs) in a multicenter North American series of patients (ET, n = 1,247; PD, n = 633) and controls (n = 642). The sequencing study identified six novel coding variants in LINGO1 (p.S4C, p.V107M, p.A277T, p.R423R, p.G537A, p.D610D) and three in LINGO2 (p.D135D, p.P217P, p.V565V), however segregation analysis did not support pathogenicity. The association study employed 16 tSNPs at the LINGO1 locus and 21 at the LINGO2 locus. One variant in LINGO1 (rs9652490) displayed evidence of an association with ET (odds ratio (OR) = 0.63; P = 0.026) and PD (OR = 0.54; P = 0.016). Additionally, four other tSNPs in LINGO1 and one in LINGO2 were associated with ET and one tSNP in LINGO2 associated with PD (P < 0.05). Further analysis identified one tSNP in LINGO1 and two in LINGO2 which influenced age at onset of ET and two tSNPs in LINGO1 which altered age at onset of PD (P < 0.05). Our results support a role for LINGO1 and LINGO2 in determining risk for and perhaps age at onset of ET and PD. Further studies are warranted to confirm these findings and to determine the pathogenic mechanisms involved.


Mutation Research | 2003

Germline mutation induction at mouse repeat DNA loci by chemical mutagens.

Carles Vilariño-Güell; Andrew G. Smith; Yuri E. Dubrova

Mutation rates at two expanded simple tandem repeat (ESTR) loci were studied in the germline of male mice exposed to two monofunctional alkylating agents, ethylnitrosourea (ENU) and isopropyl methanesulfonate (iPMS), and a topoisomerase II inhibitor, etoposide. Pre-meiotic exposure to the alkylating agents resulted in a highly significant increase in ESTR mutation rate, but did not alter post-meiotically exposed cells. Pre-meiotic mutation induction by ENU and iPMS was linear within the interval of doses from 12.5 to 25mg/kg and reached a plateau at higher concentrations. Paternal exposure to etoposide resulted in ESTR mutation induction at meiotic stages but did not affect post- or pre-meiotic cells. The pattern of ESTR mutation induction after pre-meiotic and meiotic exposure to chemical mutagens was similar to that previously obtained by various traditional approaches for monitoring germline mutation in mice. The results of this study show that ESTR loci provide a new efficient experimental system for monitoring the genetic effects of chemical mutagens, capable of detecting increases in mutation rates at low doses of exposure.

Collaboration


Dive into the Carles Vilariño-Güell's collaboration.

Top Co-Authors

Avatar

Matthew J. Farrer

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jan O. Aasly

Norwegian University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Alex Rajput

University of Saskatchewan

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Timothy Lynch

Mater Misericordiae University Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge