Your browser doesn't support javascript.
loading
Excitatory Dysfunction Drives Network and Calcium Handling Deficits in 16p11.2 Duplication Schizophrenia Induced Pluripotent Stem Cell-Derived Neurons.
Parnell, Euan; Culotta, Lorenza; Forrest, Marc P; Jalloul, Hiba A; Eckman, Blair L; Loizzo, Daniel D; Horan, Katherine K E; Dos Santos, Marc; Piguel, Nicolas H; Tai, Derek J C; Zhang, Hanwen; Gertler, Tracy S; Simkin, Dina; Sanders, Alan R; Talkowski, Michael E; Gejman, Pablo V; Kiskinis, Evangelos; Duan, Jubao; Penzes, Peter.
Afiliação
  • Parnell E; Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Northwestern University Center for Autism and Neurodevelopment, Chicago, Illinois.
  • Culotta L; Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Northwestern University Center for Autism and Neurodevelopment, Chicago, Illinois.
  • Forrest MP; Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Northwestern University Center for Autism and Neurodevelopment, Chicago, Illinois.
  • Jalloul HA; Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Northwestern University Center for Autism and Neurodevelopment, Chicago, Illinois.
  • Eckman BL; Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Northwestern University Center for Autism and Neurodevelopment, Chicago, Illinois.
  • Loizzo DD; Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Northwestern University Center for Autism and Neurodevelopment, Chicago, Illinois.
  • Horan KKE; Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Northwestern University Center for Autism and Neurodevelopment, Chicago, Illinois.
  • Dos Santos M; Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Northwestern University Center for Autism and Neurodevelopment, Chicago, Illinois.
  • Piguel NH; Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Northwestern University Center for Autism and Neurodevelopment, Chicago, Illinois.
  • Tai DJC; Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts; Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.
  • Zhang H; Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, Illinois; Department of Psychiatry and Behavioral Neurosciences, The University of Chicago, Chicago, Illinois.
  • Gertler TS; Division of Neurology, Department of Pediatrics, Ann and Robert H Lurie Childrens Hospital of Chicago, Chicago, Illinois; Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
  • Simkin D; Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
  • Sanders AR; Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, Illinois; Department of Psychiatry and Behavioral Neurosciences, The University of Chicago, Chicago, Illinois.
  • Talkowski ME; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts; Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts.
  • Gejman PV; Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, Illinois; Department of Psychiatry and Behavioral Neurosciences, The University of Chicago, Chicago, Illinois.
  • Kiskinis E; Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
  • Duan J; Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, Illinois; Department of Psychiatry and Behavioral Neurosciences, The University of Chicago, Chicago, Illinois.
  • Penzes P; Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Northwestern University Center for Autism and Neurodevelopment, Chicago, Illinois; Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, Illin
Biol Psychiatry ; 94(2): 153-163, 2023 07 15.
Article em En | MEDLINE | ID: mdl-36581494
ABSTRACT

BACKGROUND:

Schizophrenia (SCZ) is a debilitating psychiatric disorder with a large genetic contribution; however, its neurodevelopmental substrates remain largely unknown. Modeling pathogenic processes in SCZ using human induced pluripotent stem cell-derived neurons (iNs) has emerged as a promising strategy. Copy number variants confer high genetic risk for SCZ, with duplication of the 16p11.2 locus increasing the risk 14.5-fold.

METHODS:

To dissect the contribution of induced excitatory neurons (iENs) versus GABAergic (gamma-aminobutyric acidergic) neurons (iGNs) to SCZ pathophysiology, we induced iNs from CRISPR (clustered regularly interspaced short palindromic repeats)-Cas9 isogenic and SCZ patient-derived induced pluripotent stem cells and analyzed SCZ-related phenotypes in iEN monocultures and iEN/iGN cocultures.

RESULTS:

In iEN/iGN cocultures, neuronal firing and synchrony were reduced at later, but not earlier, stages of in vitro development. These were fully recapitulated in iEN monocultures, indicating a primary role for iENs. Moreover, isogenic iENs showed reduced dendrite length and deficits in calcium handling. iENs from 16p11.2 duplication-carrying patients with SCZ displayed overlapping deficits in network synchrony, dendrite outgrowth, and calcium handling. Transcriptomic analysis of both iEN cohorts revealed molecular markers of disease related to the glutamatergic synapse, neuroarchitecture, and calcium regulation.

CONCLUSIONS:

Our results indicate the presence of 16p11.2 duplication-dependent alterations in SCZ patient-derived iENs. Transcriptomics and cellular phenotyping reveal overlap between isogenic and patient-derived iENs, suggesting a central role of glutamatergic, morphological, and calcium dysregulation in 16p11.2 duplication-mediated pathogenesis. Moreover, excitatory dysfunction during early neurodevelopment is implicated as the basis of SCZ pathogenesis in 16p11.2 duplication carriers. Our results support network synchrony and calcium handling as outcomes directly linked to this genetic risk variant.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Esquizofrenia / Células-Tronco Pluripotentes Induzidas Limite: Humans Idioma: En Ano de publicação: 2023 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Esquizofrenia / Células-Tronco Pluripotentes Induzidas Limite: Humans Idioma: En Ano de publicação: 2023 Tipo de documento: Article