Your browser doesn't support javascript.
loading
Deleterious variants in CRLS1 lead to cardiolipin deficiency and cause an autosomal recessive multi-system mitochondrial disease.
Lee, Richard G; Balasubramaniam, Shanti; Stentenbach, Maike; Kralj, Tom; McCubbin, Tim; Padman, Benjamin; Smith, Janine; Riley, Lisa G; Priyadarshi, Archana; Peng, Liuyu; Nuske, Madison R; Webster, Richard; Peacock, Ken; Roberts, Philip; Stark, Zornitza; Lemire, Gabrielle; Ito, Yoko A; Boycott, Kym M; Geraghty, Michael T; van Klinken, Jan Bert; Ferdinandusse, Sacha; Zhou, Ying; Walsh, Rebecca; Marcellin, Esteban; Thorburn, David R; Rosciolli, Tony; Fletcher, Janice; Rackham, Oliver; Vaz, Frédéric M; Reid, Gavin E; Filipovska, Aleksandra.
Affiliation
  • Lee RG; Telethon Kids Institute, Northern Entrance, Perth Children's Hospital, Nedlands, WA 6009, Australia.
  • Balasubramaniam S; Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, WA 6009, Australia.
  • Stentenbach M; ARC Centre of Excellence in Synthetic Biology, Centre for Medical Research, The University of Western Australia, QEII Medical Centre, Nedlands, WA 6009, Australia.
  • Kralj T; Genetic Metabolic Disorders Service, The Children's Hospital at Westmead, Sydney, NSW 2145, Australia.
  • McCubbin T; Discipline of Genomic Medicine, Sydney Medical School, University of Sydney, Sydney, NSW 2006, Australia.
  • Padman B; Telethon Kids Institute, Northern Entrance, Perth Children's Hospital, Nedlands, WA 6009, Australia.
  • Smith J; Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, WA 6009, Australia.
  • Riley LG; ARC Centre of Excellence in Synthetic Biology, Centre for Medical Research, The University of Western Australia, QEII Medical Centre, Nedlands, WA 6009, Australia.
  • Priyadarshi A; School of Chemistry, The University of Melbourne, Parkville, VIC 3010, Australia.
  • Peng L; Australian Institute for Bioengineering and Nanotechnology, and Queensland Node of Metabolomics Australia,The University of Queensland, St Lucia, QLD 4072, Australia.
  • Nuske MR; Centre for Microscopy, Characterisation and Analysis, The University of WA, Perth, WA 6009, Australia.
  • Webster R; Discipline of Genomic Medicine, Sydney Medical School, University of Sydney, Sydney, NSW 2006, Australia.
  • Peacock K; Department of Clinical Genetics, The Children's Hospital at Westmead, Sydney, NSW 2145, Australia.
  • Roberts P; Rare Diseases Functional Genomics, Kids Research, The Children's Hospital at Westmead and Children's Medical Research Institute, Sydney, NSW 2145, Australia.
  • Stark Z; Discipline of Child and Adolescent Health, University of Sydney, Sydney, NSW 2145, Australia.
  • Lemire G; Discipline of Child and Adolescent Health, University of Sydney, Sydney, NSW 2145, Australia.
  • Ito YA; Neonatal Intensive Care Unit, Westmead Hospital, Sydney, NSW 2145, Australia.
  • Boycott KM; School of Chemistry, The University of Melbourne, Parkville, VIC 3010, Australia.
  • Geraghty MT; Department of Paediatrics, University of Melbourne, VIC 3052, Australia.
  • van Klinken JB; Kids Neuroscience Centre, The Children's Hospital at Westmead, Sydney, NSW 2145, Australia.
  • Ferdinandusse S; General Paediatric Medicine, The Children's Hospital at Westmead, Sydney, NSW 2145, Australia.
  • Zhou Y; Heart Centre for Children, The Children's Hospital at Westmead, Sydney, NSW 2145, Australia.
  • Walsh R; University of Melbourne, Parkville, VIC 3052, Australia.
  • Marcellin E; Australian Genomics, Melbourne, VIC 3052, Australia.
  • Thorburn DR; Victorian Clinical Genetics Services, Melbourne, VIC 3052, Australia.
  • Rosciolli T; Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON K1H 8L1, Canada.
  • Fletcher J; Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON K1H 8L1, Canada.
  • Vaz FM; Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON K1H 8L1, Canada.
  • Reid GE; Metabolics and Newborn Screening, Pediatrics, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa, ON K1H 8L1, Canada.
  • Filipovska A; Department of Clinical Chemistry, Laboratory Genetic Metabolic Diseases, University of Amsterdam, Amsterdam Gastroenterology Endocrinology Metabolism, 1105 AZ Amsterdam, The Netherlands.
Hum Mol Genet ; 31(21): 3597-3612, 2022 10 28.
Article in En | MEDLINE | ID: mdl-35147173
ABSTRACT
Mitochondrial diseases are a group of inherited diseases with highly varied and complex clinical presentations. Here, we report four individuals, including two siblings, affected by a progressive mitochondrial encephalopathy with biallelic variants in the cardiolipin biosynthesis gene CRLS1. Three affected individuals had a similar infantile presentation comprising progressive encephalopathy, bull's eye maculopathy, auditory neuropathy, diabetes insipidus, autonomic instability, cardiac defects and early death. The fourth affected individual presented with chronic encephalopathy with neurodevelopmental regression, congenital nystagmus with decreased vision, sensorineural hearing loss, failure to thrive and acquired microcephaly. Using patient-derived fibroblasts, we characterized cardiolipin synthase 1 (CRLS1) dysfunction that impaired mitochondrial morphology and biogenesis, providing functional evidence that the CRLS1 variants cause mitochondrial disease. Lipid profiling in fibroblasts from two patients further confirmed the functional defect demonstrating reduced cardiolipin levels, altered acyl-chain composition and significantly increased levels of phosphatidylglycerol, the substrate of CRLS1. Proteomic profiling of patient cells and mouse Crls1 knockout cell lines identified both endoplasmic reticular and mitochondrial stress responses, and key features that distinguish between varying degrees of cardiolipin insufficiency. These findings support that deleterious variants in CRLS1 cause an autosomal recessive mitochondrial disease, presenting as a severe encephalopathy with multi-systemic involvement. Furthermore, we identify key signatures in cardiolipin and proteome profiles across various degrees of cardiolipin loss, facilitating the use of omics technologies to guide future diagnosis of mitochondrial diseases.
Subject(s)

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Brain Diseases / Mitochondrial Diseases Type of study: Prognostic_studies Limits: Animals Language: En Journal: Hum Mol Genet Journal subject: BIOLOGIA MOLECULAR / GENETICA MEDICA Year: 2022 Type: Article Affiliation country: Australia

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Brain Diseases / Mitochondrial Diseases Type of study: Prognostic_studies Limits: Animals Language: En Journal: Hum Mol Genet Journal subject: BIOLOGIA MOLECULAR / GENETICA MEDICA Year: 2022 Type: Article Affiliation country: Australia