Your browser doesn't support javascript.
loading
Functional interpretation of ATAD3A variants in neuro-mitochondrial phenotypes.
Yap, Zheng Yie; Park, Yo Han; Wortmann, Saskia B; Gunning, Adam C; Ezer, Shlomit; Lee, Sukyeong; Duraine, Lita; Wilichowski, Ekkehard; Wilson, Kate; Mayr, Johannes A; Wagner, Matias; Li, Hong; Kini, Usha; Black, Emily Davis; Monaghan, Kristin G; Lupski, James R; Ellard, Sian; Westphal, Dominik S; Harel, Tamar; Yoon, Wan Hee.
Afiliación
  • Yap ZY; Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA.
  • Park YH; Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA.
  • Wortmann SB; Institute of Human Genetics, Technical University Munich, Munich, Germany.
  • Gunning AC; University Children's Hospital, Paracelsus Medical University (PMU), Salzburg, Austria.
  • Ezer S; Radboud Centre for Mitochondrial Medicine (RCMM), Amalia Children's Hospital, Nijmegen, The Netherlands.
  • Lee S; Exeter Genomics Laboratory, Royal Devon and Exeter NHS Foundation Trust, Exeter, EX2 5DW, UK.
  • Duraine L; Institute of Biomedical and Clinical Science, College of Medicine and Health, University of Exeter, Exeter, EX2 5DW, UK.
  • Wilichowski E; Department of Genetics, Hadassah Medical Center, POB 12000, 9112001, Jerusalem, Israel.
  • Wilson K; Faculty of Medicine, Hebrew University of Jerusalem, POB 12000, 9112001, Jerusalem, Israel.
  • Mayr JA; Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA.
  • Wagner M; Jan and Dan Duncan Neurological Research Institute, Baylor College of Medicine, Houston, TX, USA.
  • Li H; Department of Pediatrics and Pediatric Neurology, University Medical Center Göttingen, Georg-August-Universität Göttingen, Göttingen, Germany.
  • Kini U; Oxford Centre for Genomic Medicine, Oxford University Hospitals NHS Foundation Trust, Oxford, UK.
  • Black ED; University Children's Hospital, Paracelsus Medical University (PMU), Salzburg, Austria.
  • Monaghan KG; Institute of Human Genetics, Technical University Munich, Munich, Germany.
  • Lupski JR; Institute of Neurogenomics, Helmholtz Zentrum München, Neuherberg, Germany.
  • Ellard S; Department of Human Genetics, School of Medicine, Emory University, Atlanta, GA, USA.
  • Westphal DS; Department of Pediatrics, School of Medicine, Children's Healthcare of Atlanta, Emory University, Atlanta, GA, USA.
  • Harel T; Oxford Centre for Genomic Medicine, Oxford University Hospitals NHS Foundation Trust, Oxford, UK.
  • Yoon WH; Department of Human Genetics, School of Medicine, Emory University, Atlanta, GA, USA.
Genome Med ; 13(1): 55, 2021 04 12.
Article en En | MEDLINE | ID: mdl-33845882
ABSTRACT

BACKGROUND:

ATPase family AAA-domain containing protein 3A (ATAD3A) is a nuclear-encoded mitochondrial membrane-anchored protein involved in diverse processes including mitochondrial dynamics, mitochondrial DNA organization, and cholesterol metabolism. Biallelic deletions (null), recessive missense variants (hypomorph), and heterozygous missense variants or duplications (antimorph) in ATAD3A lead to neurological syndromes in humans.

METHODS:

To expand the mutational spectrum of ATAD3A variants and to provide functional interpretation of missense alleles in trans to deletion alleles, we performed exome sequencing for identification of single nucleotide variants (SNVs) and copy number variants (CNVs) in ATAD3A in individuals with neurological and mitochondrial phenotypes. A Drosophila Atad3a Gal4 knockin-null allele was generated using CRISPR-Cas9 genome editing technology to aid the interpretation of variants.

RESULTS:

We report 13 individuals from 8 unrelated families with biallelic ATAD3A variants. The variants included four missense variants inherited in trans to loss-of-function alleles (p.(Leu77Val), p.(Phe50Leu), p.(Arg170Trp), p.(Gly236Val)), a homozygous missense variant p.(Arg327Pro), and a heterozygous non-frameshift indel p.(Lys568del). Affected individuals exhibited findings previously associated with ATAD3A pathogenic variation, including developmental delay, hypotonia, congenital cataracts, hypertrophic cardiomyopathy, and cerebellar atrophy. Drosophila studies indicated that Phe50Leu, Gly236Val, Arg327Pro, and Lys568del are severe loss-of-function alleles leading to early developmental lethality. Further, we showed that Phe50Leu, Gly236Val, and Arg327Pro cause neurogenesis defects. On the contrary, Leu77Val and Arg170Trp are partial loss-of-function alleles that cause progressive locomotion defects and whose expression leads to an increase in autophagy and mitophagy in adult muscles.

CONCLUSION:

Our findings expand the allelic spectrum of ATAD3A variants and exemplify the use of a functional assay in Drosophila to aid variant interpretation.
Asunto(s)
Palabras clave

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Variación Genética / Proteínas Mitocondriales / ATPasas Asociadas con Actividades Celulares Diversas / Proteínas de la Membrana / Mitocondrias / Neuronas Tipo de estudio: Prognostic_studies Límite: Adolescent / Adult / Animals / Female / Humans / Infant / Male / Newborn Idioma: En Revista: Genome Med Año: 2021 Tipo del documento: Article País de afiliación: Estados Unidos

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Variación Genética / Proteínas Mitocondriales / ATPasas Asociadas con Actividades Celulares Diversas / Proteínas de la Membrana / Mitocondrias / Neuronas Tipo de estudio: Prognostic_studies Límite: Adolescent / Adult / Animals / Female / Humans / Infant / Male / Newborn Idioma: En Revista: Genome Med Año: 2021 Tipo del documento: Article País de afiliación: Estados Unidos