Network


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

Hotspot


Dive into the research topics where William Sproviero is active.

Publication


Featured researches published by William Sproviero.


Nature Genetics | 2016

NEK1 variants confer susceptibility to amyotrophic lateral sclerosis

Kevin Kenna; Perry T.C. van Doormaal; Annelot M. Dekker; Nicola Ticozzi; Brendan J. Kenna; Frank P. Diekstra; Wouter van Rheenen; Kristel R. van Eijk; Ashley Jones; Pamela Keagle; Aleksey Shatunov; William Sproviero; Bradley Smith; Michael A. van Es; Simon Topp; Aoife Kenna; John Miller; Claudia Fallini; Cinzia Tiloca; Russell McLaughlin; Caroline Vance; Claire Troakes; Claudia Colombrita; Gabriele Mora; Andrea Calvo; Federico Verde; Safa Al-Sarraj; Andrew King; Daniela Calini; Jacqueline de Belleroche

To identify genetic factors contributing to amyotrophic lateral sclerosis (ALS), we conducted whole-exome analyses of 1,022 index familial ALS (FALS) cases and 7,315 controls. In a new screening strategy, we performed gene-burden analyses trained with established ALS genes and identified a significant association between loss-of-function (LOF) NEK1 variants and FALS risk. Independently, autozygosity mapping for an isolated community in the Netherlands identified a NEK1 p.Arg261His variant as a candidate risk factor. Replication analyses of sporadic ALS (SALS) cases and independent control cohorts confirmed significant disease association for both p.Arg261His (10,589 samples analyzed) and NEK1 LOF variants (3,362 samples analyzed). In total, we observed NEK1 risk variants in nearly 3% of ALS cases. NEK1 has been linked to several cellular functions, including cilia formation, DNA-damage response, microtubule stability, neuronal morphology and axonal polarity. Our results provide new and important insights into ALS etiopathogenesis and genetic etiology.


Genome Research | 2017

Detection of long repeat expansions from PCR-free whole-genome sequence data

Egor Dolzhenko; Joke J. F. A. van Vugt; Richard Shaw; Mitchell A. Bekritsky; Marka van Blitterswijk; Giuseppe Narzisi; Subramanian S. Ajay; Vani Rajan; Bryan Lajoie; Nathan H. Johnson; Zoya Kingsbury; Sean Humphray; Raymond D. Schellevis; William J. Brands; Matt Baker; Rosa Rademakers; Maarten Kooyman; Gijs H.P. Tazelaar; Michael A. van Es; Russell McLaughlin; William Sproviero; Aleksey Shatunov; Ashley Jones; Ahmad Al Khleifat; Alan Pittman; Sarah Morgan; Orla Hardiman; Ammar Al-Chalabi; Christopher Shaw; Bradley Smith

Identifying large expansions of short tandem repeats (STRs), such as those that cause amyotrophic lateral sclerosis (ALS) and fragile X syndrome, is challenging for short-read whole-genome sequencing (WGS) data. A solution to this problem is an important step toward integrating WGS into precision medicine. We developed a software tool called ExpansionHunter that, using PCR-free WGS short-read data, can genotype repeats at the locus of interest, even if the expanded repeat is larger than the read length. We applied our algorithm to WGS data from 3001 ALS patients who have been tested for the presence of the C9orf72 repeat expansion with repeat-primed PCR (RP-PCR). Compared against this truth data, ExpansionHunter correctly classified all (212/212, 95% CI [0.98, 1.00]) of the expanded samples as either expansions (208) or potential expansions (4). Additionally, 99.9% (2786/2789, 95% CI [0.997, 1.00]) of the wild-type samples were correctly classified as wild type by this method with the remaining three samples identified as possible expansions. We further applied our algorithm to a set of 152 samples in which every sample had one of eight different pathogenic repeat expansions, including those associated with fragile X syndrome, Friedreichs ataxia, and Huntingtons disease, and correctly flagged all but one of the known repeat expansions. Thus, ExpansionHunter can be used to accurately detect known pathogenic repeat expansions and provides researchers with a tool that can be used to identify new pathogenic repeat expansions.


JAMA Neurology | 2016

Association of a locus in the CAMTA1 gene with survival in patients with sporadic amyotrophic lateral sclerosis

Isabella Fogh; Kuang Lin; Cinzia Tiloca; James Rooney; Cinzia Gellera; Frank P. Diekstra; Antonia Ratti; Aleksey Shatunov; Michael A. van Es; Petroula Proitsi; Ashley Jones; William Sproviero; Adriano Chiò; Russell McLaughlin; Gianni Sorarù; Lucia Corrado; Daniel Stahl; Roberto Del Bo; Cristina Cereda; Barbara Castellotti; Jonathan D. Glass; S Newhouse; Richard Dobson; Bradley Smith; Simon Topp; Wouter van Rheenen; Vincent Meininger; Judith Melki; Karen E. Morrison; Pamela J. Shaw

IMPORTANCE Amyotrophic lateral sclerosis (ALS) is a devastating adult-onset neurodegenerative disorder with a poor prognosis and a median survival of 3 years. However, a significant proportion of patients survive more than 10 years from symptom onset. OBJECTIVE To identify gene variants influencing survival in ALS. DESIGN, SETTING, AND PARTICIPANTS This genome-wide association study (GWAS) analyzed survival in data sets from several European countries and the United States that were collected by the Italian Consortium for the Genetics of ALS and the International Consortium on Amyotrophic Lateral Sclerosis Genetics. The study population included 4256 patients with ALS (3125 [73.4%] deceased) with genotype data extended to 7 174 392 variants by imputation analysis. Samples of DNA were collected from January 1, 1993, to December 31, 2009, and analyzed from March 1, 2014, to February 28, 2015. MAIN OUTCOMES AND MEASURES Cox proportional hazards regression under an additive model with adjustment for age at onset, sex, and the first 4 principal components of ancestry, followed by meta-analysis, were used to analyze data. Survival distributions for the most associated genetic variants were assessed by Kaplan-Meier analysis. RESULTS Among the 4256 patients included in the analysis (2589 male [60.8%] and 1667 female [39.2%]; mean [SD] age at onset, 59 [12] years), the following 2 novel loci were significantly associated with ALS survival: at 10q23 (rs139550538; P = 1.87 × 10-9) and in the CAMTA1 gene at 1p36 (rs2412208, P = 3.53 × 10-8). At locus 10q23, the adjusted hazard ratio for patients with the rs139550538 AA or AT genotype was 1.61 (95% CI, 1.38-1.89; P = 1.87 × 10-9), corresponding to an 8-month reduction in survival compared with TT carriers. For rs2412208 CAMTA1, the adjusted hazard ratio for patients with the GG or GT genotype was 1.17 (95% CI, 1.11-1.24; P = 3.53 × 10-8), corresponding to a 4-month reduction in survival compared with TT carriers. CONCLUSIONS AND RELEVANCE This GWAS robustly identified 2 loci at genome-wide levels of significance that influence survival in patients with ALS. Because ALS is a rare disease and prevention is not feasible, treatment that modifies survival is the most realistic strategy. Therefore, identification of modifier genes that might influence ALS survival could improve the understanding of the biology of the disease and suggest biological targets for pharmaceutical intervention. In addition, genetic risk scores for survival could be used as an adjunct to clinical trials to account for the genetic contribution to survival.


Neurobiology of Aging | 2015

Investigation of next-generation sequencing technologies as a diagnostic tool for amyotrophic lateral sclerosis

Sarah Morgan; Maryam Shoai; Pietro Fratta; Katie Sidle; Richard W. Orrell; Mary G. Sweeney; Aleksey Shatunov; William Sproviero; Ashley Jones; Ammar Al-Chalabi; Andrea Malaspina; Henry Houlden; John Hardy; Alan Pittman

The future of genetic diagnostics will see a move toward massively parallel next-generation sequencing of a patients DNA. Amyotrophic lateral sclerosis (ALS) is one of the diseases that would benefit from this prospect. Exploring this idea, we designed a screening panel to sequence 25 ALS-linked genes and examined samples from 95 patients with both familial and sporadic ALS. Forty-three rare polymorphisms were detected in this cohort. A third of these have already been reported with respect to ALS, leaving 28 novel variants all open for further investigation. This study highlights the potential benefits of next-generation sequencing as a reliable, cost and time efficient, diagnostic, and research tool for ALS.


Neurobiology of Aging | 2013

Residual association at C9orf72 suggests an alternative amyotrophic lateral sclerosis-causing hexanucleotide repeat

Ashley Jones; Ione O.C. Woollacott; Aleksey Shatunov; Johnathan Cooper-Knock; Vladimir L. Buchman; William Sproviero; Bradley Smith; Kirsten M. Scott; Rubika Balendra; Olubunmi Abel; Peter McGuffin; Catherine M. Ellis; Pamela J. Shaw; Karen E. Morrison; Anne Farmer; Cathryn M. Lewis; P. Nigel Leigh; Christopher Shaw; John Powell; Ammar Al-Chalabi

Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease of motor neurons. Single-nucleotide polymorphism rs3849942 is associated with ALS, tagging a hexanucleotide repeat mutation in the C9orf72 gene. It is possible that there is more than 1 disease-causing genetic variation at this locus, in which case association might remain after removal of cases carrying the mutation. DNA from patients with ALS was therefore tested for the mutation. Genome-wide association testing was performed first using all samples, and then restricting the analysis to samples not carrying the mutation. rs3849942 and rs903603 were strongly associated with ALS when all samples were included (rs3849942, p = [3 × 2] × 10−6, rank 7/442,057; rs903603, p = [7 × 6] × 10−8, rank 2/442,057). Removal of the mutation-carrying cases resulted in loss of association for rs3849942 (p = [2 × 6] × 10−3, rank 1225/442,068), but had little effect on rs903603 (p = [1 × 9] × 10−5, rank 8/442,068). Those with a risk allele of rs903603 had an excess of apparent homozygosity for wild type repeat alleles, consistent with polymerase chain reaction failure of 1 allele because of massive repeat expansion. These results indicate residual association at the C9orf72 locus suggesting a second disease-causing repeat mutation.


Neurobiology of Aging | 2017

ATXN2 trinucleotide repeat length correlates with risk of ALS

William Sproviero; Aleksey Shatunov; Daniel Stahl; Maryam Shoai; Wouter van Rheenen; Ashley Jones; Safa Al-Sarraj; Peter Andersen; Nancy M. Bonini; Francesca Luisa Conforti; Philip Van Damme; Hussein Daoud; Maria del Mar Amador; Isabella Fogh; Monica Forzan; Ben Gaastra; Cinzia Gellera; Aaron D. Gitler; John Hardy; Pietro Fratta; Vincenzo La Bella; Isabelle Le Ber; Tim Van Langenhove; Serena Lattante; Yi-Chung Lee; Andrea Malaspina; Vincent Meininger; Stéphanie Millecamps; Richard W. Orrell; Rosa Rademakers

We investigated a CAG trinucleotide repeat expansion in the ATXN2 gene in amyotrophic lateral sclerosis (ALS). Two new case-control studies, a British dataset of 1474 ALS cases and 567 controls, and a Dutch dataset of 1328 ALS cases and 691 controls were analyzed. In addition, to increase power, we systematically searched PubMed for case-control studies published after 1 August 2010 that investigated the association between ATXN2 intermediate repeats and ALS. We conducted a meta-analysis of the new and existing studies for the relative risks of ATXN2 intermediate repeat alleles of between 24 and 34 CAG trinucleotide repeats and ALS. There was an overall increased risk of ALS for those carrying intermediate sized trinucleotide repeat alleles (odds ratio 3.06 [95% confidence interval 2.37–3.94]; p = 6 × 10−18), with an exponential relationship between repeat length and ALS risk for alleles of 29–32 repeats (R2 = 0.91, p = 0.0002). No relationship was seen for repeat length and age of onset or survival. In contrast to trinucleotide repeat diseases, intermediate ATXN2 trinucleotide repeat expansion in ALS does not predict age of onset but does predict disease risk.


Brain | 2017

A comprehensive analysis of rare genetic variation in amyotrophic lateral sclerosis in the UK.

Sarah Morgan; Aleksey Shatunov; William Sproviero; Ashley Jones; Maryam Shoai; Deborah Hughes; Ahmad Al Khleifat; Andrea Malaspina; Karen E. Morrison; Pamela J. Shaw; Christopher Shaw; Katie Sidle; Richard W. Orrell; Pietro Fratta; John Hardy; Alan Pittman; Ammar Al-Chalabi

The genetic landscape of amyotrophic lateral sclerosis (ALS) is poorly understood. By examining known ALS-associated genes in a large cohort, Morgan, Shatunov et al. report an increase in mutations within the untranslated prime regions of the genes and a greater than expected number of patients with multiple potentially pathogenic variants.


Neurology | 2017

Meta-analysis of pharmacogenetic interactions in amyotrophic lateral sclerosis clinical trials

Ruben P.A. van Eijk; Ashley Jones; William Sproviero; Aleksey Shatunov; Pamela J. Shaw; P. Nigel Leigh; Carolyn Young; Christopher Shaw; Gabriele Mora; Jessica Mandrioli; Giuseppe Borghero; Paolo Volanti; Frank P. Diekstra; Wouter van Rheenen; Esther Verstraete; Marinus J.C. Eijkemans; Jan H. Veldink; Adriano Chiò; Ammar Al-Chalabi; Leonard H. van den Berg; Michael A. van Es; For UKMND-LiCALS

Objective: To assess whether genetic subgroups in recent amyotrophic lateral sclerosis (ALS) trials responded to treatment with lithium carbonate, but that the treatment effect was lost in a large cohort of nonresponders. Methods: Individual participant data were obtained from 3 randomized trials investigating the efficacy of lithium carbonate. We matched clinical data with data regarding the UNC13A and C9orf72 genotype. Our primary outcome was survival at 12 months. On an exploratory basis, we assessed whether the effect of lithium depended on the genotype. Results: Clinical data were available for 518 of the 606 participants. Overall, treatment with lithium carbonate did not improve 12-month survival (hazard ratio [HR] 1.0, 95% confidence interval [CI] 0.7–1.4; p = 0.96). Both the UNC13A and C9orf72 genotype were independent predictors of survival (HR 2.4, 95% CI 1.3–4.3; p = 0.006 and HR 2.5, 95% CI 1.1–5.2; p = 0.032, respectively). The effect of lithium was different for UNC13A carriers (p = 0.027), but not for C9orf72 carriers (p = 0.22). The 12-month survival probability for UNC13A carriers treated with lithium carbonate improved from 40.1% (95% CI 23.2–69.1) to 69.7% (95% CI 50.4–96.3). Conclusions: This study incorporated genetic data into past ALS trials to determine treatment effects in a genetic post hoc analysis. Our results suggest that we should reorient our strategies toward finding treatments for ALS, start focusing on genotype-targeted treatments, and standardize genotyping in order to optimize randomization and analysis for future clinical trials.


Amyotrophic Lateral Sclerosis | 2016

Rare genetic variation in UNC13A may modify survival in amyotrophic lateral sclerosis

Benjamin Gaastra; Aleksey Shatunov; Sara L. Pulit; Ashley Jones; William Sproviero; Alexandra Gillett; Zhongbo Chen; Janine Kirby; Isabella Fogh; John Powell; P. Nigel Leigh; Karen E. Morrison; Pamela J. Shaw; Christopher Shaw; Leonard H. van den Berg; Jan H. Veldink; Cathryn M. Lewis; Ammar Al-Chalabi

Abstract Our objective was to identify whether rare genetic variation in amyotrophic lateral sclerosis (ALS) candidate survival genes modifies ALS survival. Candidate genes were selected based on evidence for modifying ALS survival. Each tail of the extreme 1.5% of survival was selected from the UK MND DNA Bank and all samples available underwent whole genome sequencing. A replication set from the Netherlands was used for validation. Sequences of candidate survival genes were extracted and variants passing quality control with a minor allele frequency ≤0.05 were selected for association testing. Analysis was by burden testing using SKAT. Candidate survival genes UNC13A, KIFAP3, and EPHA4 were tested for association in a UK sample comprising 25 short survivors and 25 long survivors. Results showed that only SNVs in UNC13A were associated with survival (p = 6.57 × 10−3). SNV rs10419420:G > A was found exclusively in long survivors (3/25) and rs4808092:G > A exclusively in short survivors (4/25). These findings were not replicated in a Dutch sample. In conclusion, population specific rare variants of UNC13A may modulate survival in ALS.


Journal of Medical Genetics | 2018

Identification of new risk factors for rolandic epilepsy: CNV at Xp22.31 and alterations at cholinergic synapses

Laura Addis; William Sproviero; Sanjeev V Thomas; Roberto Horacio Caraballo; Stephen Newhouse; Kumudini Gomez; Elaine Hughes; Maria Kinali; David McCormick; Siobhan Hannan; Silvia Cossu; Jacqueline Taylor; Cigdem I. Akman; Steven M. Wolf; David E. Mandelbaum; Rajesh Gupta; Rick A. A. van der Spek; Dario Pruna; Deb K. Pal

Background Rolandic epilepsy (RE) is the most common genetic childhood epilepsy, consisting of focal, nocturnal seizures and frequent neurodevelopmental impairments in speech, language, literacy and attention. A complex genetic aetiology is presumed in most, with monogenic mutations in GRIN2A accounting for >5% of cases. Objective To identify rare, causal CNV in patients with RE. Methods We used high-density SNP arrays to analyse the presence of rare CNVs in 186 patients with RE from the UK, the USA, Sardinia, Argentina and Kerala, India. Results We identified 84 patients with one or more rare CNVs, and, within this group, 14 (7.5%) with recurrent risk factor CNVs and 15 (8.0%) with likely pathogenic CNVs. Nine patients carried recurrent hotspot CNVs including at 16p13.11 and 1p36, with the most striking finding that four individuals (three from Sardinia) carried a duplication, and one a deletion, at Xp22.31. Five patients with RE carried a rare CNV that disrupted genes associated with other epilepsies (KCTD7, ARHGEF15, CACNA2D1, GRIN2A and ARHGEF4), and 17 cases carried CNVs that disrupted genes associated with other neurological conditions or that are involved in neuronal signalling/development. Network analysis of disrupted genes with high brain expression identified significant enrichment in pathways of the cholinergic synapse, guanine-exchange factor activation and the mammalian target of rapamycin. Conclusion Our results provide a CNV profile of an ethnically diverse cohort of patients with RE, uncovering new areas of research focus, and emphasise the importance of studying non-western European populations in oligogenic disorders to uncover a full picture of risk variation.

Collaboration


Dive into the William Sproviero's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge