Your browser doesn't support javascript.
loading
Phenotypic and genetic spectrum of ATP6V1A encephalopathy: a disorder of lysosomal homeostasis.
Guerrini, Renzo; Mei, Davide; Kerti-Szigeti, Katalin; Pepe, Sara; Koenig, Mary Kay; Von Allmen, Gretchen; Cho, Megan T; McDonald, Kimberly; Baker, Janice; Bhambhani, Vikas; Powis, Zöe; Rodan, Lance; Nabbout, Rima; Barcia, Giulia; Rosenfeld, Jill A; Bacino, Carlos A; Mignot, Cyril; Power, Lillian H; Harris, Catharine J; Marjanovic, Dragan; Møller, Rikke S; Hammer, Trine B; Keski Filppula, Riikka; Vieira, Päivi; Hildebrandt, Clara; Sacharow, Stephanie; Maragliano, Luca; Benfenati, Fabio; Lachlan, Katherine; Benneche, Andreas; Petit, Florence; de Sainte Agathe, Jean Madeleine; Hallinan, Barbara; Si, Yue; Wentzensen, Ingrid M; Zou, Fanggeng; Narayanan, Vinodh; Matsumoto, Naomichi; Boncristiano, Alessandra; la Marca, Giancarlo; Kato, Mitsuhiro; Anderson, Kristin; Barba, Carmen; Sturiale, Luisa; Garozzo, Domenico; Bei, Roberto; Masuelli, Laura; Conti, Valerio; Novarino, Gaia; Fassio, Anna.
Afiliação
  • Guerrini R; Neuroscience Department, Children's Hospital Meyer, University of Florence, Florence, Italy.
  • Mei D; Neuroscience Department, Children's Hospital Meyer, University of Florence, Florence, Italy.
  • Kerti-Szigeti K; Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria.
  • Pepe S; Department of Experimental Medicine, University of Genoa, Italy.
  • Koenig MK; Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy.
  • Von Allmen G; Department of Pediatrics, Division of Child and Adolescent Neurology, The University of Texas McGovern Medical School, Houston, TX, USA.
  • Cho MT; Department of Pediatrics, Division of Child and Adolescent Neurology, The University of Texas McGovern Medical School, Houston, TX, USA.
  • McDonald K; GeneDx, Gaithersburg, MD 20877, USA.
  • Baker J; Pediatric Neurology, University of Mississippi Medical Center, Jackson, MS, USA.
  • Bhambhani V; Genetics and Genomics, Children's Minnesota, Minneapolis, MN, USA.
  • Powis Z; Genetics and Genomics, Children's Minnesota, Minneapolis, MN, USA.
  • Rodan L; Ambry Genetics, Aliso Viejo, CA, USA.
  • Nabbout R; Division of Genetics and Genomics and Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
  • Barcia G; Reference Centre for Rare Epilepsies, Department of Genetics, Necker Enfants Malades Hospital, APHP, member of ERN EpiCARE, Université de Paris, Paris, France.
  • Rosenfeld JA; Reference Centre for Rare Epilepsies, Department of Genetics, Necker Enfants Malades Hospital, APHP, member of ERN EpiCARE, Université de Paris, Paris, France.
  • Bacino CA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA.
  • Mignot C; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA.
  • Power LH; APHP, Sorbonne Université, Départément de Génétique, Centre de Référence Déficiences Intellectuelles de Causes Rares, Paris, France.
  • Harris CJ; Institut du Cerveau (ICM), UMR S 1127, Inserm U1127, CNRS UMR 7225, Sorbonne Université, 75013 Paris, France.
  • Marjanovic D; Pediatric Neurology, Stead Family Department of Pediatrics, University of Iowa Stead Family Children's Hospital, Iowa City, IA, USA.
  • Møller RS; Department of Pediatric Genetics, University of Missouri Medical Center, Columbia, MO 65212, USA.
  • Hammer TB; Danish Epilepsy Centre Filadelfia, Adult Neurology, Dianalund, Denmark.
  • Keski Filppula R; Department of Regional Health Services, University of Southern Denmark, Odense, Denmark.
  • Vieira P; Department of Epilepsy Genetics and Personalized Treatment, Danish Epilepsy Center Filadelfia, Dianalund, Denmark.
  • Sacharow S; Department of Clinical Genetics, Oulu University Hospital, Medical Research Center Oulu and PEDEGO Research Unit, University of Oulu, Oulu, Finland.
  • Maragliano L; Division of Genetics and Genomics, Metabolism Program, Boston Children's Hospital, Boston, MA, USA.
  • Benfenati F; Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
  • Benneche A; Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy.
  • Petit F; Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy.
  • de Sainte Agathe JM; Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy.
  • Hallinan B; IRCCS Ospedale Policlinico San Martino, Genova, Italy.
  • Si Y; Wessex Clinical Genetics Service, University Hospital Southampton NHS Foundation Trust, Southampton, UK.
  • Wentzensen IM; Human Development and Health, Faculty of Medicine University of Southampton, Southampton, UK.
  • Zou F; Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway.
  • Narayanan V; CHU Lille, Clinique de Génétique, F-59000 Lille, France.
  • Matsumoto N; Laboratoire de Biologie Médicale Multi Sites SeqOIA, Laboratoire de Médecine Génomique, APHP. Sorbonne Université, Paris, France.
  • Boncristiano A; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
  • la Marca G; Division of Child Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
  • Kato M; GeneDx, Gaithersburg, MD 20877, USA.
  • Anderson K; GeneDx, Gaithersburg, MD 20877, USA.
  • Barba C; GeneDx, Gaithersburg, MD 20877, USA.
  • Sturiale L; Neurogenomics Division, Center for Rare Childhood Disorders, Translational Genomics Research Institute (TGen), Phoenix, AZ 85012, USA.
  • Garozzo D; Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan.
  • Bei R; Neuroscience Department, Children's Hospital Meyer, University of Florence, Florence, Italy.
  • Masuelli L; Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy.
  • Conti V; Department of Pediatrics, Showa University School of Medicine and Epilepsy Medical Center, Showa University Hospital, Tokyo, Japan.
  • Novarino G; Founder and Research Liaison, 'ATP6V1A Families' Facebook group.
  • Fassio A; Neuroscience Department, Children's Hospital Meyer, University of Florence, Florence, Italy.
Brain ; 145(8): 2687-2703, 2022 08 27.
Article em En | MEDLINE | ID: mdl-35675510
ABSTRACT
Vacuolar-type H+-ATPase (V-ATPase) is a multimeric complex present in a variety of cellular membranes that acts as an ATP-dependent proton pump and plays a key role in pH homeostasis and intracellular signalling pathways. In humans, 22 autosomal genes encode for a redundant set of subunits allowing the composition of diverse V-ATPase complexes with specific properties and expression. Sixteen subunits have been linked to human disease. Here we describe 26 patients harbouring 20 distinct pathogenic de novo missense ATP6V1A variants, mainly clustering within the ATP synthase α/ß family-nucleotide-binding domain. At a mean age of 7 years (extremes 6 weeks, youngest deceased patient to 22 years, oldest patient) clinical pictures included early lethal encephalopathies with rapidly progressive massive brain atrophy, severe developmental epileptic encephalopathies and static intellectual disability with epilepsy. The first clinical manifestation was early hypotonia, in 70%; 81% developed epilepsy, manifested as developmental epileptic encephalopathies in 58% of the cohort and with infantile spasms in 62%; 63% of developmental epileptic encephalopathies failed to achieve any developmental, communicative or motor skills. Less severe outcomes were observed in 23% of patients who, at a mean age of 10 years and 6 months, exhibited moderate intellectual disability, with independent walking and variable epilepsy. None of the patients developed communicative language. Microcephaly (38%) and amelogenesis imperfecta/enamel dysplasia (42%) were additional clinical features. Brain MRI demonstrated hypomyelination and generalized atrophy in 68%. Atrophy was progressive in all eight individuals undergoing repeated MRIs. Fibroblasts of two patients with developmental epileptic encephalopathies showed decreased LAMP1 expression, Lysotracker staining and increased organelle pH, consistent with lysosomal impairment and loss of V-ATPase function. Fibroblasts of two patients with milder disease, exhibited a different phenotype with increased Lysotracker staining, decreased organelle pH and no significant modification in LAMP1 expression. Quantification of substrates for lysosomal enzymes in cellular extracts from four patients revealed discrete accumulation. Transmission electron microscopy of fibroblasts of four patients with variable severity and of induced pluripotent stem cell-derived neurons from two patients with developmental epileptic encephalopathies showed electron-dense inclusions, lipid droplets, osmiophilic material and lamellated membrane structures resembling phospholipids. Quantitative assessment in induced pluripotent stem cell-derived neurons identified significantly smaller lysosomes. ATP6V1A-related encephalopathy represents a new paradigm among lysosomal disorders. It results from a dysfunctional endo-lysosomal membrane protein causing altered pH homeostasis. Its pathophysiology implies intracellular accumulation of substrates whose composition remains unclear, and a combination of developmental brain abnormalities and neurodegenerative changes established during prenatal and early postanal development, whose severity is variably determined by specific pathogenic variants.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Espasmos Infantis / Encefalopatias / ATPases Vacuolares Próton-Translocadoras / Epilepsia / Deficiência Intelectual Idioma: En Ano de publicação: 2022 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Espasmos Infantis / Encefalopatias / ATPases Vacuolares Próton-Translocadoras / Epilepsia / Deficiência Intelectual Idioma: En Ano de publicação: 2022 Tipo de documento: Article