Network


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

Hotspot


Dive into the research topics where Adeline Vanderver is active.

Publication


Featured researches published by Adeline Vanderver.


Nature Genetics | 2009

Mutations involved in Aicardi-Goutieres syndrome implicate SAMHD1 as regulator of the innate immune response

Gillian I. Rice; Jacquelyn Bond; Aruna Asipu; Rebecca L. Brunette; Iain W. Manfield; Ian M. Carr; Jonathan C. Fuller; Richard M. Jackson; Teresa Lamb; Tracy A. Briggs; Manir Ali; Hannah Gornall; Alec Aeby; Simon P Attard-Montalto; Enrico Bertini; C. Bodemer; Knut Brockmann; Louise Brueton; Peter Corry; Isabelle Desguerre; Elisa Fazzi; Angels Garcia Cazorla; Blanca Gener; B.C.J. Hamel; Arvid Heiberg; Matthew Hunter; Marjo S. van der Knaap; Ram Kumar; Lieven Lagae; Pierre Landrieu

Aicardi-Goutières syndrome is a mendelian mimic of congenital infection and also shows overlap with systemic lupus erythematosus at both a clinical and biochemical level. The recent identification of mutations in TREX1 and genes encoding the RNASEH2 complex and studies of the function of TREX1 in DNA metabolism have defined a previously unknown mechanism for the initiation of autoimmunity by interferon-stimulatory nucleic acid. Here we describe mutations in SAMHD1 as the cause of AGS at the AGS5 locus and present data to show that SAMHD1 may act as a negative regulator of the cell-intrinsic antiviral response.


Nature Genetics | 2012

Mutations in ADAR1 cause Aicardi-Goutières syndrome associated with a type I interferon signature

Gillian I. Rice; Paul R. Kasher; Gabriella M.A. Forte; Niamh M. Mannion; Sam M. Greenwood; Marcin Szynkiewicz; Jonathan E. Dickerson; Sanjeev Bhaskar; Massimiliano Zampini; Tracy A. Briggs; Emma M. Jenkinson; Carlos A. Bacino; Roberta Battini; Enrico Bertini; Paul A. Brogan; Louise Brueton; Marialuisa Carpanelli; Corinne De Laet; Pascale de Lonlay; Mireia del Toro; Isabelle Desguerre; Elisa Fazzi; Angels García-Cazorla; Arvid Heiberg; Masakazu Kawaguchi; Ram Kumar; Jean-Pierre Lin; Charles Marques Lourenço; Alison Male; Wilson Marques

Adenosine deaminases acting on RNA (ADARs) catalyze the hydrolytic deamination of adenosine to inosine in double-stranded RNA (dsRNA) and thereby potentially alter the information content and structure of cellular RNAs. Notably, although the overwhelming majority of such editing events occur in transcripts derived from Alu repeat elements, the biological function of non-coding RNA editing remains uncertain. Here, we show that mutations in ADAR1 (also known as ADAR) cause the autoimmune disorder Aicardi-Goutières syndrome (AGS). As in Adar1-null mice, the human disease state is associated with upregulation of interferon-stimulated genes, indicating a possible role for ADAR1 as a suppressor of type I interferon signaling. Considering recent insights derived from the study of other AGS-related proteins, we speculate that ADAR1 may limit the cytoplasmic accumulation of the dsRNA generated from genomic repetitive elements.


Nature Genetics | 2014

Gain-of-function mutations in IFIH1 cause a spectrum of human disease phenotypes associated with upregulated type I interferon signaling

Gillian I. Rice; Yoandris del Toro Duany; Emma M. Jenkinson; Gabriella M.A. Forte; Beverley Anderson; Giada Ariaudo; Brigitte Bader-Meunier; Roberta Battini; Michael W. Beresford; Manuela Casarano; Mondher Chouchane; Rolando Cimaz; Abigail Collins; Nuno J V Cordeiro; Russell C. Dale; Joyce Davidson; Liesbeth De Waele; Isabelle Desguerre; Laurence Faivre; Elisa Fazzi; Bertrand Isidor; Lieven Lagae; Andrew Latchman; Pierre Lebon; Chumei Li; John H. Livingston; Charles Marques Lourenço; Maria Margherita Mancardi; Alice Masurel-Paulet; Iain B. McInnes

The type I interferon system is integral to human antiviral immunity. However, inappropriate stimulation or defective negative regulation of this system can lead to inflammatory disease. We sought to determine the molecular basis of genetically uncharacterized cases of the type I interferonopathy Aicardi-Goutières syndrome and of other undefined neurological and immunological phenotypes also demonstrating an upregulated type I interferon response. We found that heterozygous mutations in the cytosolic double-stranded RNA receptor gene IFIH1 (also called MDA5) cause a spectrum of neuroimmunological features consistently associated with an enhanced interferon state. Cellular and biochemical assays indicate that these mutations confer gain of function such that mutant IFIH1 binds RNA more avidly, leading to increased baseline and ligand-induced interferon signaling. Our results demonstrate that aberrant sensing of nucleic acids can cause immune upregulation.


Human Mutation | 2008

Molecular and clinical genetics of mitochondrial diseases due to POLG mutations.

Lee-Jun C. Wong; Robert K. Naviaux; Nicola Brunetti-Pierri; Qing Zhang; Eric S. Schmitt; Cavatina K. Truong; Margherita Milone; Bruce H. Cohen; Beverly Wical; Jaya Ganesh; Alice Basinger; Barbara K. Burton; Kathryn J. Swoboda; Donald L. Gilbert; Adeline Vanderver; Russell P. Saneto; Bruno Maranda; Georgianne L. Arnold; Jose E. Abdenur; Paula J. Waters; William C. Copeland

Mutations in the POLG gene have emerged as one of the most common causes of inherited mitochondrial disease in children and adults. They are responsible for a heterogeneous group of at least 6 major phenotypes of neurodegenerative disease that include: 1) childhood Myocerebrohepatopathy Spectrum disorders (MCHS), 2) Alpers syndrome, 3) Ataxia Neuropathy Spectrum (ANS) disorders, 4) Myoclonus Epilepsy Myopathy Sensory Ataxia (MEMSA), 5) autosomal recessive Progressive External Ophthalmoplegia (arPEO), and 6) autosomal dominant Progressive External Ophthalmoplegia (adPEO). Due to the clinical heterogeneity, time‐dependent evolution of symptoms, overlapping phenotypes, and inconsistencies in muscle pathology findings, definitive diagnosis relies on the molecular finding of deleterious mutations. We sequenced the exons and flanking intron region from approximately 350 patients displaying a phenotype consistent with POLG related mitochondrial disease and found informative mutations in 61 (17%). Two mutant alleles were identified in 31 unrelated index patients with autosomal recessive POLG‐related disorders. Among them, 20 (67%) had Alpers syndrome, 4 (13%) had arPEO, and 3 (10%) had ANS. In addition, 30 patients carrying one altered POLG allele were found. A total of 25 novel alterations were identified, including 6 null mutations. We describe the predicted structural/functional and clinical importance of the previously unreported missense variants and discuss their likelihood of being pathogenic. In conclusion, sequence analysis allows the identification of mutations responsible for POLG‐related disorders and, in most of the autosomal recessive cases where two mutant alleles are found in trans, finding deleterious mutations can provide an unequivocal diagnosis of the disease. Published 2008 Wiley‐Liss, Inc.


Brain | 2010

Magnetic resonance imaging pattern recognition in hypomyelinating disorders.

Marjan E. Steenweg; Adeline Vanderver; Susan Blaser; Alberto Bizzi; Tom J. de Koning; Grazia M.S. Mancini; Wessel N. van Wieringen; Frederik Barkhof; Nicole I. Wolf; Marjo S. van der Knaap

Hypomyelination is observed in the context of a growing number of genetic disorders that share clinical characteristics. The aim of this study was to determine the possible role of magnetic resonance imaging pattern recognition in distinguishing different hypomyelinating disorders, which would facilitate the diagnostic process. Only patients with hypomyelination of known cause were included in this retrospective study. A total of 112 patients with Pelizaeus-Merzbacher disease, hypomyelination with congenital cataract, hypomyelination with hypogonadotropic hypogonadism and hypodontia, Pelizaeus-Merzbacher-like disease, infantile GM1 and GM2 gangliosidosis, Salla disease and fucosidosis were included. The brain scans were rated using a standard scoring list; the raters were blinded to the diagnoses. Grouping of the patients was based on cluster analysis. Ten clusters of patients with similar magnetic resonance imaging abnormalities were identified. The most important discriminating items were early cerebellar atrophy, homogeneity of the white matter signal on T(2)-weighted images, abnormal signal intensity of the basal ganglia, signal abnormalities in the pons and additional T(2) lesions in the deep white matter. Eight clusters each represented mainly a single disorder (i.e. Pelizaeus-Merzbacher disease, hypomyelination with congenital cataract, hypomyelination with hypogonadotropic hypogonadism and hypodontia, infantile GM1 and GM2 gangliosidosis, Pelizaeus-Merzbacher-like disease and fucosidosis); only two clusters contained multiple diseases. Pelizaeus-Merzbacher-like disease was divided between two clusters and Salla disease did not cluster at all. This study shows that it is possible to separate patients with hypomyelination disorders of known cause in clusters based on magnetic resonance imaging abnormalities alone. In most cases of Pelizaeus-Merzbacher disease, hypomyelination with congenital cataract, hypomyelination with hypogonadotropic hypogonadism and hypodontia, Pelizaeus-Merzbacher-like disease, infantile GM1 and GM2 gangliosidosis and fucosidosis, the imaging pattern gives clues for the diagnosis.


Lancet Neurology | 2013

Assessment of interferon-related biomarkers in Aicardi-Goutières syndrome associated with mutations in TREX1, RNASEH2A, RNASEH2B, RNASEH2C, SAMHD1, and ADAR: A case-control study

Gillian I. Rice; Gabriella M.A. Forte; Marcin Szynkiewicz; Diana Chase; Alec Aeby; Mohamed S. Abdel-Hamid; Sam Ackroyd; Rebecca L Allcock; Kathryn M. Bailey; Umberto Balottin; Christine Barnerias; Geneviève Bernard; C. Bodemer; Maria P. Botella; Cristina Cereda; Kate Chandler; Lyvia Dabydeen; Russell C. Dale; Corinne De Laet; Christian de Goede; Mireia del Toro; Laila Effat; Noemi Nunez Enamorado; Elisa Fazzi; Blanca Gener; Madli Haldre; Jean-Pierre Lin; John H. Livingston; Charles Marques Lourenço; Wilson Marques

BACKGROUND Aicardi-Goutières syndrome (AGS) is an inflammatory disorder caused by mutations in any of six genes (TREX1, RNASEH2A, RNASEH2B, RNASEH2C, SAMHD1, and ADAR). The disease is severe and effective treatments are urgently needed. We investigated the status of interferon-related biomarkers in patients with AGS with a view to future use in diagnosis and clinical trials. METHODS In this case-control study, samples were collected prospectively from patients with mutation-proven AGS. The expression of six interferon-stimulated genes (ISGs) was measured by quantitative PCR, and the median fold change, when compared with the median of healthy controls, was used to create an interferon score for each patient. Scores higher than the mean of controls plus two SD (>2·466) were designated as positive. Additionally, we collated historical data for interferon activity, measured with a viral cytopathic assay, in CSF and serum from mutation-positive patients with AGS. We also undertook neutralisation assays of interferon activity in serum, and looked for the presence of autoantibodies against a panel of interferon proteins. FINDINGS 74 (90%) of 82 patients had a positive interferon score (median 12·90, IQR 6·14-20·41) compared with two (7%) of 29 controls (median 0·93, IQR 0·57-1·30). Of the eight patients with a negative interferon score, seven had mutations in RNASEH2B (seven [27%] of all 26 patients with mutations in this gene). Repeat sampling in 16 patients was consistent for the presence or absence of an interferon signature on 39 of 41 occasions. Interferon activity (tested in 147 patients) was negatively correlated with age (CSF, r=-0·604; serum, r=-0·289), and was higher in CSF than in serum in 104 of 136 paired samples. Neutralisation assays suggested that measurable antiviral activity was related to interferon α production. We did not record significantly increased concentrations of autoantibodies to interferon subtypes in patients with AGS, or an association between the presence of autoantibodies and interferon score or serum interferon activity. INTERPRETATION AGS is consistently associated with an interferon signature, which is apparently sustained over time and can thus be used to differentiate patients with AGS from controls. If future studies show that interferon status is a reactive biomarker, the measurement of an interferon score might prove useful in the assessment of treatment efficacy in clinical trials. FUNDING European Unions Seventh Framework Programme; European Research Council.


Cell | 2015

Ubiquitous L1 Mosaicism in Hippocampal Neurons

Kyle R. Upton; Daniel J. Gerhardt; J. Samuel Jesuadian; Sandra R. Richardson; Francisco J. Sánchez-Luque; Gabriela O. Bodea; Adam D. Ewing; Carmen Salvador-Palomeque; Marjo S. van der Knaap; Paul M. Brennan; Adeline Vanderver; Geoffrey J. Faulkner

Summary Somatic LINE-1 (L1) retrotransposition during neurogenesis is a potential source of genotypic variation among neurons. As a neurogenic niche, the hippocampus supports pronounced L1 activity. However, the basal parameters and biological impact of L1-driven mosaicism remain unclear. Here, we performed single-cell retrotransposon capture sequencing (RC-seq) on individual human hippocampal neurons and glia, as well as cortical neurons. An estimated 13.7 somatic L1 insertions occurred per hippocampal neuron and carried the sequence hallmarks of target-primed reverse transcription. Notably, hippocampal neuron L1 insertions were specifically enriched in transcribed neuronal stem cell enhancers and hippocampus genes, increasing their probability of functional relevance. In addition, bias against intronic L1 insertions sense oriented relative to their host gene was observed, perhaps indicating moderate selection against this configuration in vivo. These experiments demonstrate pervasive L1 mosaicism at genomic loci expressed in hippocampal neurons.


American Journal of Human Genetics | 2013

A De Novo Mutation in the β-Tubulin Gene TUBB4A Results in the Leukoencephalopathy Hypomyelination with Atrophy of the Basal Ganglia and Cerebellum

Cas Simons; Nicole I. Wolf; Nathan McNeil; Ljubica Caldovic; Joseph M. Devaney; Asako Takanohashi; Joanna Crawford; Kelin Ru; Sean M. Grimmond; David Miller; Davide Tonduti; Johanna L. Schmidt; Robert S. Chudnow; Rudy Van Coster; Lieven Lagae; Jill Kisler; Juergen Sperner; Marjo S. van der Knaap; Raphael Schiffmann; Ryan J. Taft; Adeline Vanderver

Hypomyelination with atrophy of the basal ganglia and cerebellum (H-ABC) is a rare hereditary leukoencephalopathy that was originally identified by MRI pattern analysis, and it has thus far defied all attempts at identifying the causal mutation. Only 22 cases are published in the literature to date. We performed exome sequencing on five family trios, two family quartets, and three single probands, which revealed that all eleven H-ABC-diagnosed individuals carry the same de novo single-nucleotide TUBB4A mutation resulting in nonsynonymous change p.Asp249Asn. Detailed investigation of one of the family quartets with the singular finding of an H-ABC-affected sibling pair revealed maternal mosaicism for the mutation, suggesting that rare de novo mutations that are initially phenotypically neutral in a mosaic individual can be disease causing in the subsequent generation. Modeling of TUBB4A shows that the mutation creates a nonsynonymous change at a highly conserved asparagine that sits at the intradimer interface of α-tubulin and β-tubulin, and this change might affect tubulin dimerization, microtubule polymerization, or microtubule stability. Consistent with H-ABCs clinical presentation, TUBB4A is highly expressed in neurons, and a recent report has shown that an N-terminal alteration is associated with a heritable dystonia. Together, these data demonstrate that a single de novo mutation in TUBB4A results in H-ABC.


Neurology | 2011

GFAP mutations, age at onset, and clinical subtypes in Alexander disease

Morgan Prust; Jichuan Wang; H. Morizono; Albee Messing; Michael Brenner; E. Gordon; T. Hartka; A. Sokohl; R. Schiffmann; Heather Gordish-Dressman; Roger L. Albin; H. Amartino; K. Brockman; A. Dinopoulos; M.T. Dotti; D. Fain; R. Fernandez; J. Ferreira; J. Fleming; D. Gill; M. Griebel; H. Heilstedt; D. Lewis; M. Nakagawa; R. Pedersen; A. Reddy; Y. Sawaishi; M. Schneider; E. Sherr; Y. Takiyama

Objective: To characterize Alexander disease (AxD) phenotypes and determine correlations with age at onset (AAO) and genetic mutation. AxD is an astrogliopathy usually characterized on MRI by leukodystrophy and caused by glial fibrillary acidic protein (GFAP) mutations. Methods: We present 30 new cases of AxD and reviewed 185 previously reported cases. We conducted Wilcoxon rank sum tests to identify variables scaling with AAO, survival analysis to identify predictors of mortality, and χ2 tests to assess the effects of common GFAP mutations. Finally, we performed latent class analysis (LCA) to statistically define AxD subtypes. Results: LCA identified 2 classes of AxD. Type I is characterized by early onset, seizures, macrocephaly, motor delay, encephalopathy, failure to thrive, paroxysmal deterioration, and typical MRI features. Type II is characterized by later onset, autonomic dysfunction, ocular movement abnormalities, bulbar symptoms, and atypical MRI features. Survival analysis predicted a nearly 2-fold increase in mortality among patients with type I AxD relative to those with type II. R79 and R239 GFAP mutations were most common (16.6% and 20.3% of all cases, respectively). These common mutations predicted distinct clinical outcomes, with R239 predicting the most aggressive course. Conclusions: AAO and the GFAP mutation site are important clinical predictors in AxD, with clear correlations to defined patterns of phenotypic expression. We propose revised AxD subtypes, type I and type II, based on analysis of statistically defined patient groups.


American Journal of Human Genetics | 2013

Mutations in DARS Cause Hypomyelination with Brain Stem and Spinal Cord Involvement and Leg Spasticity

Ryan J. Taft; Adeline Vanderver; Richard J. Leventer; Stephen Damiani; Cas Simons; Sean M. Grimmond; David Miller; Johanna L. Schmidt; Paul J. Lockhart; Kate Pope; Kelin Ru; Joanna Crawford; Tena Rosser; Irenaeus F.M. de Coo; Monica Juneja; Ishwar C. Verma; Prab Prabhakar; Susan Blaser; Julian Raiman; Petra J. W. Pouwels; Marianna R. Bevova; Truus E. M. Abbink; Marjo S. van der Knaap; Nicole I. Wolf

Inherited white-matter disorders are a broad class of diseases for which treatment and classification are both challenging. Indeed, nearly half of the children presenting with a leukoencephalopathy remain without a specific diagnosis. Here, we report on the application of high-throughput genome and exome sequencing to a cohort of ten individuals with a leukoencephalopathy of unknown etiology and clinically characterized by hypomyelination with brain stem and spinal cord involvement and leg spasticity (HBSL), as well as the identification of compound-heterozygous and homozygous mutations in cytoplasmic aspartyl-tRNA synthetase (DARS). These mutations cause nonsynonymous changes to seven highly conserved amino acids, five of which are unchanged between yeast and man, in the DARS C-terminal lobe adjacent to, or within, the active-site pocket. Intriguingly, HBSL bears a striking resemblance to leukoencephalopathy with brain stem and spinal cord involvement and elevated lactate (LBSL), which is caused by mutations in the mitochondria-specific DARS2, suggesting that these two diseases might share a common underlying molecular pathology. These findings add to the growing body of evidence that mutations in tRNA synthetases can cause a broad range of neurologic disorders.

Collaboration


Dive into the Adeline Vanderver's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Guy Helman

Children's National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Geneviève Bernard

Montreal Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Raphael Schiffmann

Baylor University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Nicole I. Wolf

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Cas Simons

University of Queensland

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Johanna L. Schmidt

Children's National Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Amy Pizzino

Children's National Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge