Your browser doesn't support javascript.
loading
Expanded phenotype of AARS1-related white matter disease.
Helman, Guy; Mendes, Marisa I; Nicita, Francesco; Darbelli, Lama; Sherbini, Omar; Moore, Travis; Derksen, Alexa; Carrozzo, Rosalba; Torraco, Alessandra; Catteruccia, Michela; Aiello, Chiara; Goffrini, Paola; Figuccia, Sonia; Smith, Desiree E C; Hadzsiev, Kinga; Hahn, Andreas; Biskup, Saskia; Brösse, Ines; Kotzaeridou, Urania; Gauck, Darja; Grebe, Theresa A; Elmslie, Frances; Stals, Karen; Gupta, Rajat; Bertini, Enrico; Thiffault, Isabelle; Taft, Ryan J; Schiffmann, Raphael; Brandl, Ulrich; Haack, Tobias B; Salomons, Gajja S; Simons, Cas; Bernard, Geneviève; van der Knaap, Marjo S; Vanderver, Adeline; Husain, Ralf A.
Afiliación
  • Helman G; Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, VIC, Australia.
  • Mendes MI; Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia.
  • Nicita F; Metabolic Unit, Department of Clinical Chemistry, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
  • Darbelli L; Department of Neurosciences, Unit of Muscular and Neurodegenerative Disorders, Laboratory of Molecular Medicine, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.
  • Sherbini O; Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada.
  • Moore T; Child Health and Human Development Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada.
  • Derksen A; Department of Pediatrics, McGill University, Montreal, QC, Canada.
  • Amy Pizzino; Department of Human Genetics, McGill University, Montreal, QC, Canada.
  • Carrozzo R; Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
  • Torraco A; Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada.
  • Catteruccia M; Child Health and Human Development Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada.
  • Aiello C; Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada.
  • Goffrini P; Child Health and Human Development Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada.
  • Figuccia S; Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
  • Smith DEC; Department of Neurosciences, Unit of Muscular and Neurodegenerative Disorders, Laboratory of Molecular Medicine, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.
  • Hadzsiev K; Department of Neurosciences, Unit of Muscular and Neurodegenerative Disorders, Laboratory of Molecular Medicine, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.
  • Hahn A; Department of Neurosciences, Unit of Muscular and Neurodegenerative Disorders, Laboratory of Molecular Medicine, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.
  • Biskup S; Department of Neurosciences, Unit of Muscular and Neurodegenerative Disorders, Laboratory of Molecular Medicine, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.
  • Brösse I; Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy.
  • Kotzaeridou U; Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy.
  • Gauck D; Metabolic Unit, Department of Clinical Chemistry, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
  • Grebe TA; Department of Medical Genetics, University of Pécs, Pécs, Hungary.
  • Elmslie F; Department of Child Neurology, Justus-Liebig-University, Giessen, Germany.
  • Stals K; Praxis fuer Humangenetik and CeGaT GmbH, Tuebingen, Germany.
  • Gupta R; Division of Child Neurology and Inherited Metabolic Diseases, Centre for Paediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany.
  • Bertini E; Division of Child Neurology and Inherited Metabolic Diseases, Centre for Paediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany.
  • Thiffault I; Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Tuebingen, Germany.
  • Taft RJ; Division of Genetics and Metabolism, Department of Child Health, Phoenix Children's Hospital, University of Arizona College of Medicine, Phoenix, AZ, USA.
  • Schiffmann R; South West Thames Regional Genetics Service, St George's University Hospital, London, UK.
  • Brandl U; Molecular Genetics Department, Royal Devon and Exeter NHS Foundation Trust, Exeter, UK.
  • Haack TB; Department of Neurology, Birmingham Children's Hospital, Birmingham, UK.
  • Salomons GS; Department of Neurosciences, Unit of Muscular and Neurodegenerative Disorders, Laboratory of Molecular Medicine, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.
  • Simons C; Children's Mercy Kansas City, Center for Pediatric Genomic Medicine, Kansas City, MO, USA.
  • Bernard G; Department of Pathology and Laboratory Medicine, Children's Mercy Hospitals, Kansas City, MO, USA.
  • van der Knaap MS; School of Medicine, University of Missouri-Kansas City, Kansas City, MO, USA.
  • Vanderver A; Illumina, Inc, San Diego, CA, USA.
  • Husain RA; Baylor Scott & White Research Institute, Dallas, TX, USA.
Genet Med ; 23(12): 2352-2359, 2021 12.
Article en En | MEDLINE | ID: mdl-34446925
ABSTRACT

PURPOSE:

Recent reports of individuals with cytoplasmic transfer RNA (tRNA) synthetase-related disorders have identified cases with phenotypic variability from the index presentations. We sought to assess phenotypic variability in individuals with AARS1-related disease.

METHODS:

A cross-sectional survey was performed on individuals with biallelic variants in AARS1. Clinical data, neuroimaging, and genetic testing results were reviewed. Alanyl tRNA synthetase (AlaRS) activity was measured in available fibroblasts.

RESULTS:

We identified 11 affected individuals. Two phenotypic presentations emerged, one with early infantile-onset disease resembling the index cases of AARS1-related epileptic encephalopathy with deficient myelination (n = 7). The second (n = 4) was a later-onset disorder, where disease onset occurred after the first year of life and was characterized on neuroimaging by a progressive posterior predominant leukoencephalopathy evolving to include the frontal white matter. AlaRS activity was significantly reduced in five affected individuals with both early infantile-onset and late-onset phenotypes.

CONCLUSION:

We suggest that variants in AARS1 result in a broader clinical spectrum than previously appreciated. The predominant form results in early infantile-onset disease with epileptic encephalopathy and deficient myelination. However, a subgroup of affected individuals manifests with late-onset disease and similarly rapid progressive clinical decline. Longitudinal imaging and clinical follow-up will be valuable in understanding factors affecting disease progression and outcome.
Asunto(s)

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Leucoencefalopatías Tipo de estudio: Observational_studies / Prevalence_studies / Prognostic_studies / Risk_factors_studies Límite: Humans Idioma: En Revista: Genet Med Asunto de la revista: GENETICA MEDICA Año: 2021 Tipo del documento: Article País de afiliación: Australia

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Leucoencefalopatías Tipo de estudio: Observational_studies / Prevalence_studies / Prognostic_studies / Risk_factors_studies Límite: Humans Idioma: En Revista: Genet Med Asunto de la revista: GENETICA MEDICA Año: 2021 Tipo del documento: Article País de afiliación: Australia