Network


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

Hotspot


Dive into the research topics where Annelot M. Dekker is active.

Publication


Featured researches published by Annelot M. Dekker.


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.


Lancet Neurology | 2017

Safety and efficacy of ozanezumab in patients with amyotrophic lateral sclerosis : a randomised, double-blind, placebo-controlled, phase 2 trial

Vincent Meininger; Angela Genge; Leonard H. van den Berg; Wim Robberecht; Albert C. Ludolph; Adriano Chiò; Seung Hyun Kim; P. Nigel Leigh; Matthew C. Kiernan; Jeremy M. Shefner; Claude Desnuelle; Karen E. Morrison; Susanne Petri; Diane Boswell; Jane Temple; Rajat Mohindra; Matthew Davies; Jonathan Bullman; Paul Rees; Arseniy Lavrov; Susanne Abdulla; Cathy Alsop; Francesca Barbieri; Stewart Bates; James D Berry; Stephan A Botez; Gaelle Bruneteau; Andrea Calvo; Rodrigo Refoios Camejo; William Camu

BACKGROUND Neurite outgrowth inhibitor A (Nogo-A) is thought to have a role in the pathophysiology of amyotrophic lateral sclerosis (ALS). A monoclonal antibody against Nogo-A showed a positive effect in the SOD1G93A mouse model of ALS, and a humanised form of this antibody (ozanezumab) was well tolerated in a first-in-human trial. Therefore, we aimed to assess the safety and efficacy of ozanezumab in patients with ALS. METHODS This randomised, double-blind, placebo-controlled, phase 2 trial was done in 34 centres in 11 countries. Patients aged 18-80 years with a diagnosis of familial or sporadic ALS were randomly assigned (1:1), centrally according to a computer-generated allocation schedule, to receive ozanezumab (15 mg/kg) or placebo as intravenous infusions over 1 h every 2 weeks for 46 weeks, followed by assessments at week 48 and week 60. Patients and study personnel were masked to treatment assignment. The primary outcome was a joint-rank analysis of function (ALS Functional Rating Scale-Revised) and overall survival, analysed at 48 weeks in all patients who received at least one dose of study drug. This study is registered with ClinicalTrials.gov, number NCT01753076, and with GSK-ClinicalStudyRegister.com, NOG112264, and is completed. FINDINGS Between Dec 20, 2012, and Nov 1, 2013, we recruited 307 patients, of whom 303 were randomly assigned to receive placebo (n=151) or ozanezumab (n=152). The adjusted mean of the joint-rank score was -14·9 (SE 13·5) for the ozanezumab group and 15·0 (13·6) for the placebo group, with a least squares mean difference of -30·0 (95% CI -67·9 to 7·9; p=0·12). Overall, reported adverse events, serious adverse events, and adverse events leading to permanent discontinuation of study drug or withdrawal from study were similar between the treatment groups, except for dyspepsia (ten [7%] in the ozanezumab group vs four [3%] in the placebo group), depression (11 [7%] vs five [3%]), and diarrhoea (25 [16%] vs 12 [8%]). Respiratory failure was the most common serious adverse event (12 [8%] vs seven [5%]). At week 60, the number of deaths was higher in the ozanezumab group (20 [13%]) than in the placebo group (16 [11%]), mainly as a result of respiratory failure (ten [7%] vs five [3%]). Two deaths were considered related to the study drug (bladder transitional cell carcinoma in the ozanezumab group and cerebrovascular accident in the placebo group). INTERPRETATION Ozanezumab did not show efficacy compared with placebo in patients with ALS. Therefore, Nogo-A does not seem to be an effective therapeutic target in ALS. FUNDING GlaxoSmithKline.


Lancet Neurology | 2018

Prognosis for patients with amyotrophic lateral sclerosis: development and validation of a personalised prediction model

Henk-Jan Westeneng; Thomas P. A. Debray; Anne E. Visser; Ruben P.A. van Eijk; James Rooney; Andrea Calvo; Sarah Martin; Christopher J McDermott; Alexander Thompson; Susana Pinto; Xenia Kobeleva; Angela Rosenbohm; Beatrice Stubendorff; Helma Sommer; Bas Middelkoop; Annelot M. Dekker; Joke J. F. A. van Vugt; Wouter van Rheenen; Alice Vajda; Mark Heverin; Mbombe Kazoka; Hannah Hollinger; Marta Gromicho; Sonja Körner; Thomas Ringer; Annekathrin Rödiger; A. Gunkel; Christopher Shaw; Annelien L Bredenoord; Michael A. van Es

BACKGROUND Amyotrophic lateral sclerosis (ALS) is a relentlessly progressive, fatal motor neuron disease with a variable natural history. There are no accurate models that predict the disease course and outcomes, which complicates risk assessment and counselling for individual patients, stratification of patients for trials, and timing of interventions. We therefore aimed to develop and validate a model for predicting a composite survival endpoint for individual patients with ALS. METHODS We obtained data for patients from 14 specialised ALS centres (each one designated as a cohort) in Belgium, France, the Netherlands, Germany, Ireland, Italy, Portugal, Switzerland, and the UK. All patients were diagnosed in the centres after excluding other diagnoses and classified according to revised El Escorial criteria. We assessed 16 patient characteristics as potential predictors of a composite survival outcome (time between onset of symptoms and non-invasive ventilation for more than 23 h per day, tracheostomy, or death) and applied backward elimination with bootstrapping in the largest population-based dataset for predictor selection. Data were gathered on the day of diagnosis or as soon as possible thereafter. Predictors that were selected in more than 70% of the bootstrap resamples were used to develop a multivariable Royston-Parmar model for predicting the composite survival outcome in individual patients. We assessed the generalisability of the model by estimating heterogeneity of predictive accuracy across external populations (ie, populations not used to develop the model) using internal-external cross-validation, and quantified the discrimination using the concordance (c) statistic (area under the receiver operator characteristic curve) and calibration using a calibration slope. FINDINGS Data were collected between Jan 1, 1992, and Sept 22, 2016 (the largest data-set included data from 1936 patients). The median follow-up time was 97·5 months (IQR 52·9-168·5). Eight candidate predictors entered the prediction model: bulbar versus non-bulbar onset (univariable hazard ratio [HR] 1·71, 95% CI 1·63-1·79), age at onset (1·03, 1·03-1·03), definite versus probable or possible ALS (1·47, 1·39-1·55), diagnostic delay (0·52, 0·51-0·53), forced vital capacity (HR 0·99, 0·99-0·99), progression rate (6·33, 5·92-6·76), frontotemporal dementia (1·34, 1·20-1·50), and presence of a C9orf72 repeat expansion (1·45, 1·31-1·61), all p<0·0001. The c statistic for external predictive accuracy of the model was 0·78 (95% CI 0·77-0·80; 95% prediction interval [PI] 0·74-0·82) and the calibration slope was 1·01 (95% CI 0·95-1·07; 95% PI 0·83-1·18). The model was used to define five groups with distinct median predicted (SE) and observed (SE) times in months from symptom onset to the composite survival outcome: very short 17·7 (0·20), 16·5 (0·23); short 25·3 (0·06), 25·2 (0·35); intermediate 32·2 (0·09), 32·8 (0·46); long 43·7 (0·21), 44·6 (0·74); and very long 91·0 (1·84), 85·6 (1·96). INTERPRETATION We have developed an externally validated model to predict survival without tracheostomy and non-invasive ventilation for more than 23 h per day in European patients with ALS. This model could be applied to individualised patient management, counselling, and future trial design, but to maximise the benefit and prevent harm it is intended to be used by medical doctors only. FUNDING Netherlands ALS Foundation.


Frontiers in Molecular Neuroscience | 2017

Targeted Genetic Screen in Amyotrophic Lateral Sclerosis Reveals Novel Genetic Variants with Synergistic Effect on Clinical Phenotype

Johnathan Cooper-Knock; Henry Robins; Isabell Niedermoser; Matthew Wyles; Paul R. Heath; Adrian Higginbottom; Theresa Walsh; Mbombe Kazoka; Guillaume M. Hautbergue; Christopher J McDermott; Janine Kirby; Pamela J. Shaw; Ahmad Al Kheifat; Ammar Al-Chalabi; Nazli Basak; Ian P. Blair; Annelot M. Dekker; Orla Hardiman; Winston Hide; Alfredo Iacoangeli; Kevin Kenna; John Landers; Russel McLaughlin; Jonathan Mill; Bas Middelkoop; Mattieu Moisse; Jesus Mora Pardina; Karen E. Morrison; Stephen Newhouse; Sara L. Pulit

Amyotrophic lateral sclerosis (ALS) is underpinned by an oligogenic rare variant architecture. Identified genetic variants of ALS include RNA-binding proteins containing prion-like domains (PrLDs). We hypothesized that screening genes encoding additional similar proteins will yield novel genetic causes of ALS. The most common genetic variant of ALS patients is a G4C2-repeat expansion within C9ORF72. We have shown that G4C2-repeat RNA sequesters RNA-binding proteins. A logical consequence of this is that loss-of-function mutations in G4C2-binding partners might contribute to ALS pathogenesis independently of and/or synergistically with C9ORF72 expansions. Targeted sequencing of genomic DNA encoding either RNA-binding proteins or known ALS genes (n = 274 genes) was performed in ALS patients to identify rare deleterious genetic variants and explore genotype-phenotype relationships. Genomic DNA was extracted from 103 ALS patients including 42 familial ALS patients and 61 young-onset (average age of onset 41 years) sporadic ALS patients; patients were chosen to maximize the probability of identifying genetic causes of ALS. Thirteen patients carried a G4C2-repeat expansion of C9ORF72. We identified 42 patients with rare deleterious variants; 6 patients carried more than one variant. Twelve mutations were discovered in known ALS genes which served as a validation of our strategy. Rare deleterious variants in RNA-binding proteins were significantly enriched in ALS patients compared to control frequencies (p = 5.31E-18). Nineteen patients featured at least one variant in a RNA-binding protein containing a PrLD. The number of variants per patient correlated with rate of disease progression (t-test, p = 0.033). We identified eighteen patients with a single variant in a G4C2-repeat binding protein. Patients with a G4C2-binding protein variant in combination with a C9ORF72 expansion had a significantly faster disease course (t-test, p = 0.025). Our data are consistent with an oligogenic model of ALS. We provide evidence for a number of entirely novel genetic variants of ALS caused by mutations in RNA-binding proteins. Moreover we show that these mutations act synergistically with each other and with C9ORF72 expansions to modify the clinical phenotype of ALS. A key finding is that this synergy is present only between functionally interacting variants. This work has significant implications for ALS therapy development.


European Journal of Human Genetics | 2018

Project MinE: study design and pilot analyses of a large-scale whole-genome sequencing study in amyotrophic lateral sclerosis

Wouter van Rheenen; Sara L. Pulit; Annelot M. Dekker; Ahmad Al Khleifat; William J. Brands; Alfredo Iacoangeli; Kevin Kenna; Ersen Kavak; Maarten Kooyman; Russell McLaughlin; Bas Middelkoop; Matthieu Moisse; Raymond D. Schellevis; Aleksey Shatunov; William Sproviero; Gijs H.P. Tazelaar; Rick A. A. van der Spek; Perry Tc Van Doormal; Kristel R. van Eijk; Joke J. F. A. van Vugt; A. Nazli Basak; Jonathan D. Glass; Orla Hardiman; Winston Hide; John Landers; Jesús S. Mora; Karen E. Morrison; Stephen Newhouse; Wim Robberecht; Christopher Shaw

The most recent genome-wide association study in amyotrophic lateral sclerosis (ALS) demonstrates a disproportionate contribution from low-frequency variants to genetic susceptibility of disease. We have therefore begun Project MinE, an international collaboration that seeks to analyse whole-genome sequence data of at least 15,000 ALS patients and 7,500 controls. Here, we report on the design of Project MinE and pilot analyses of newly whole-genome sequenced 1,264 ALS patients and 611 controls drawn from the Netherlands. As has become characteristic of sequencing studies, we find an abundance of rare genetic variation (minor allele frequency < 0.1 %), the vast majority of which is absent in public data sets. Principal component analysis reveals local geographical clustering of these variants within The Netherlands. We use the whole-genome sequence data to explore the implications of poor geographical matching of cases and controls in a sequence-based disease study and to investigate how ancestry-matched, externally sequenced controls can induce false positive associations. Also, we have publicly released genome-wide minor allele counts in cases and controls, as well as results from genic burden tests.


bioRxiv | 2018

The Project MinE databrowser: bringing large-scale whole-genome sequencing in ALS to researchers and the public.

Rick A. A. van der Spek; Wouter van Rheenen; Sara L. Pulit; Kevin P. Kenna; Russell McLaughlin; Matthieu Moisse; Annelot M. Dekker; Gijs H.P. Tazelaar; Brendan Kenna; Kristel R. van Eijk; Joke J. F. A. van Vugt; Perry T.C. van Doormaal; Bas Middelkoop; Raymond D. Schellevis; William J. Brands; Ross P. Byrne; Johnathan Cooper-Knock; Ahmad Al Khleifat; Yolanda Campos; Atay Vural; Jonathan D. Glass; Alfredo Iacoangeli; Aleksey Shatunov; William Sproviero; Ersen Kavak; Tuncay Seker; Fulya Akçimen; Cemile Kocoglu; Ceren Tunca; Nicola Ticozzi

Amyotrophic lateral sclerosis (ALS) is a rapidly progressive fatal neurodegenerative disease affecting 1 in 350 people. The aim of Project MinE is to elucidate the pathophysiology of ALS through whole-genome sequencing at least 15,000 ALS patients and 7,500 controls at 30X coverage. Here, we present the Project MinE data browser (databrowser.projectmine.com), a unique and intuitive one-stop, open-access server that provides detailed information on genetic variation analyzed in a new and still growing set of 4,366 ALS cases and 1,832 matched controls. Through its visual components and interactive design, the browser specifically aims to be a resource to those without a biostatistics background and allow clinicians and preclinical researchers to integrate Project MinE data into their own research. The browser allows users to query a transcript and immediately access a unique combination of detailed (meta)data, annotations and association statistics that would otherwise require analytic expertise and visits to scattered resources.


Neurobiology of Aging | 2016

Large-scale screening in sporadic amyotrophic lateral sclerosis identifies genetic modifiers in C9orf72 repeat carriers

Annelot M. Dekker; Meinie Seelen; Perry T.C. van Doormaal; Wouter van Rheenen; Reinoud J.P. Bothof; Tim van Riessen; William J. Brands; Anneke J. van der Kooi; Marianne de Visser; Nicol C. Voermans; R. Jeroen Pasterkamp; Jan H. Veldink; Leonard H. van den Berg; Michael A. van Es


Neurobiology of Aging | 2018

Association of NIPA1 repeat expansions with amyotrophic lateral sclerosis in a large international cohort

Gijs H.P. Tazelaar; Annelot M. Dekker; Joke J. F. A. van Vugt; Rick A. A. van der Spek; Henk-Jan Westeneng; Lindy J.B.G. Kool; Kevin Kenna; Wouter van Rheenen; Sara L. Pulit; Russell McLaughlin; William Sproviero; Alfredo Iacoangeli; Annemarie Hübers; David A. Brenner; Karen E. Morrison; Pamela J. Shaw; Christopher Shaw; Mónica Povedano Panades; Jesus Mora Pardina; Jonathan D. Glass; Orla Hardiman; Ammar Al-Chalabi; Philip Van Damme; Wim Robberecht; John Landers; Albert C. Ludolph; Jochen H. Weishaupt; Leonard H. van den Berg; Jan H. Veldink; Michael A. van Es

Collaboration


Dive into the Annelot M. Dekker'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

Kevin Kenna

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge