Your browser doesn't support javascript.
loading
Gain and loss of TASK3 channel function and its regulation by novel variation cause KCNK9 imprinting syndrome.
Cousin, Margot A; Veale, Emma L; Dsouza, Nikita R; Tripathi, Swarnendu; Holden, Robyn G; Arelin, Maria; Beek, Geoffrey; Bekheirnia, Mir Reza; Beygo, Jasmin; Bhambhani, Vikas; Bialer, Martin; Bigoni, Stefania; Boelman, Cyrus; Carmichael, Jenny; Courtin, Thomas; Cogne, Benjamin; Dabaj, Ivana; Doummar, Diane; Fazilleau, Laura; Ferlini, Alessandra; Gavrilova, Ralitza H; Graham, John M; Haack, Tobias B; Juusola, Jane; Kant, Sarina G; Kayani, Saima; Keren, Boris; Ketteler, Petra; Klöckner, Chiara; Koopmann, Tamara T; Kruisselbrink, Teresa M; Kuechler, Alma; Lambert, Laëtitia; Latypova, Xénia; Lebel, Robert Roger; Leduc, Magalie S; Leonardi, Emanuela; Lewis, Andrea M; Liew, Wendy; Machol, Keren; Mardini, Samir; McWalter, Kirsty; Mignot, Cyril; McLaughlin, Julie; Murgia, Alessandra; Narayanan, Vinodh; Nava, Caroline; Neuser, Sonja; Nizon, Mathilde; Ognibene, Davide.
Afiliação
  • Cousin MA; Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA.
  • Veale EL; Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA.
  • Dsouza NR; Medway School of Pharmacy, University of Kent and University of Greenwich, Central Avenue, Anson Building, Central Avenue, Chatham Maritime, ME4 4, Kent, TB, ME4 4 TB, UK.
  • Tripathi S; Bioinformatics Research and Development Laboratory, Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, USA.
  • Holden RG; Bioinformatics Research and Development Laboratory, Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, USA.
  • Arelin M; Medway School of Pharmacy, University of Kent and University of Greenwich, Central Avenue, Anson Building, Central Avenue, Chatham Maritime, ME4 4, Kent, TB, ME4 4 TB, UK.
  • Beek G; Department for Women and Child Health, Hospital for Children and Adolescents, University Hospitals, University of Leipzig, Leipzig, Germany.
  • Bekheirnia MR; Children's Hospital of Minnesota, Minneapolis, MN, USA.
  • Beygo J; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
  • Bhambhani V; Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.
  • Bialer M; Children's Hospital of Minnesota, Minneapolis, MN, USA.
  • Bigoni S; Division of Medical Genetics, Northwell Health, Manhasset, NY, USA.
  • Boelman C; Medical Genetics Unit, Department of Medical Sciences, Ferrara University, Ferrara, Italy.
  • Carmichael J; Division of Neurology, BC Children's Hospital, Vancouver, British Columbia, Canada.
  • Courtin T; Oxford Centre for Genomic Medicine, ACE Building, Nuffield Orthopaedic centre, Oxford University Hospitals NHS Foundation Trust, Windmill road, Headington, Oxford, OX3 7HE, UK.
  • Cogne B; Département of Genetics, APHP, Hôpital Pitié-Salpêtrière, Sorbonne Université, Paris, France.
  • Dabaj I; CHU Nantes, Service de génétique médicale, Nantes, France.
  • Doummar D; CHU de Rouen, Service de Néonatologie, Réanimation pédiatrique, Neuropédiatrie et éducation fonctionnelle de l'enfant, INSERM U 1245, ED497, 76000, Rouen, France.
  • Fazilleau L; APHP, Hôpital Raymond Poincaré, Hôpitaux Universitaires Paris Ile-de-France Ouest, Pôle pédiatrique, Service de Pédiatrie, Centre de Reference Nord-Est-Ile de France, 92380, Garches, France.
  • Ferlini A; APHP, Department of Neuropediatrics, National Reference Center for Neurogenetic Disorders, Hôpital Armand-Trousseau, GHUEP, Paris, France.
  • Gavrilova RH; Service de Néonatologie, CHU de Caen, Caen, France.
  • Graham JM; Medical Genetics Unit, Department of Medical Sciences, Ferrara University, Ferrara, Italy.
  • Haack TB; Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA.
  • Juusola J; Department of Clinical Genomics, Mayo Clinic, Rochester, MN, USA.
  • Kant SG; Department of Pediatrics, Harbor-UCLA Medical Center, Cedars-Sinai Medical Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
  • Kayani S; Centre for Rare Diseases, University of Tübingen, Tübingen, Germany.
  • Keren B; Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany.
  • Ketteler P; GeneDx, 207 Perry Parkway, Gaithersburg, MD, USA.
  • Klöckner C; Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands.
  • Koopmann TT; Department of Clinical Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.
  • Kruisselbrink TM; Departments of Pediatrics and Neurology, University of Texas Southwestern Medical Center and Children's Health, Dallas, TX, USA.
  • Kuechler A; APHP, Département de Génétique et Centre de Référence Déficiences Intellectuelles de Causes Rares, Hôpital de la Pitié-Salpêtrière, Assistance Publique - Hôpitaux de Paris, 75651, Paris, France.
  • Lambert L; Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.
  • Latypova X; Pediatrics III, Pediatric Oncology and Hematology, University Hospital Essen, Essen, Germany.
  • Lebel RR; Institute of Human Genetics, University of Leipzig Medical Center, 04103, Leipzig, Germany.
  • Leduc MS; Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands.
  • Leonardi E; Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA.
  • Lewis AM; Department of Clinical Genomics, Mayo Clinic, Rochester, MN, USA.
  • Liew W; Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.
  • Machol K; Service de Genetique Clinique, CHRU de Nancy, F-54000, Vandoeuvre-les-Nancy, France.
  • Mardini S; Unite INSERM N-GERE UMR_S 1256, Université de Lorraine, Faculté de Médecine, 9 avenue de la Forêt de Haye, CS 50184, Vandoeuvre-les-Nancy, France.
  • McWalter K; CHU Nantes, Service de génétique médicale, Nantes, France.
  • Mignot C; Section of Medical Genetics, SUNY Upstate University Hospital, Syracuse, NY, USA.
  • McLaughlin J; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
  • Murgia A; Molecular Genetics of Neurodevelopmental Disorders, Department of Woman and Child Health, University of Padova, Padua, Italy.
  • Narayanan V; Pediatric Research Institute, Città della Speranza, Padova, Italy.
  • Nava C; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
  • Neuser S; Department of Paediatric Medicine, KK Women's and Children's Hospital, Mount Elizabeth Hospital, Singapore, Singapore.
  • Nizon M; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
  • Ognibene D; Texas Children's Hospital, Houston, TX, USA.
Genome Med ; 14(1): 62, 2022 06 13.
Article em En | MEDLINE | ID: mdl-35698242
ABSTRACT

BACKGROUND:

Genomics enables individualized diagnosis and treatment, but large challenges remain to functionally interpret rare variants. To date, only one causative variant has been described for KCNK9 imprinting syndrome (KIS). The genotypic and phenotypic spectrum of KIS has yet to be described and the precise mechanism of disease fully understood.

METHODS:

This study discovers mechanisms underlying KCNK9 imprinting syndrome (KIS) by describing 15 novel KCNK9 alterations from 47 KIS-affected individuals. We use clinical genetics and computer-assisted facial phenotyping to describe the phenotypic spectrum of KIS. We then interrogate the functional effects of the variants in the encoded TASK3 channel using sequence-based analysis, 3D molecular mechanic and dynamic protein modeling, and in vitro electrophysiological and functional methodologies.

RESULTS:

We describe the broader genetic and phenotypic variability for KIS in a cohort of individuals identifying an additional mutational hotspot at p.Arg131 and demonstrating the common features of this neurodevelopmental disorder to include motor and speech delay, intellectual disability, early feeding difficulties, muscular hypotonia, behavioral abnormalities, and dysmorphic features. The computational protein modeling and in vitro electrophysiological studies discover variability of the impact of KCNK9 variants on TASK3 channel function identifying variants causing gain and others causing loss of conductance. The most consistent functional impact of KCNK9 genetic variants, however, was altered channel regulation.

CONCLUSIONS:

This study extends our understanding of KIS mechanisms demonstrating its complex etiology including gain and loss of channel function and consistent loss of channel regulation. These data are rapidly applicable to diagnostic strategies, as KIS is not identifiable from clinical features alone and thus should be molecularly diagnosed. Furthermore, our data suggests unique therapeutic strategies may be needed to address the specific functional consequences of KCNK9 variation on channel function and regulation.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Canais de Potássio de Domínios Poros em Tandem / Deficiência Intelectual Limite: Humans Idioma: En Ano de publicação: 2022 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Canais de Potássio de Domínios Poros em Tandem / Deficiência Intelectual Limite: Humans Idioma: En Ano de publicação: 2022 Tipo de documento: Article