Your browser doesn't support javascript.
loading
Vascular-derived SPARC and SerpinE1 regulate interneuron tangential migration and accelerate functional maturation of human stem cell-derived interneurons.
Genestine, Matthieu; Ambriz, Daisy; Crabtree, Gregg W; Dummer, Patrick; Molotkova, Anna; Quintero, Michael; Mela, Angeliki; Biswas, Saptarshi; Feng, Huijuan; Zhang, Chaolin; Canoll, Peter; Hargus, Gunnar; Agalliu, Dritan; Gogos, Joseph A; Au, Edmund.
Affiliation
  • Genestine M; Department of Pathology and Cell Biology, Columbia University, New York, United States.
  • Ambriz D; Department of Pathology and Cell Biology, Columbia University, New York, United States.
  • Crabtree GW; Department of Neurology, Columbia University Irving Medical Center, New York, United States.
  • Dummer P; Department of Pathology and Cell Biology, Columbia University, New York, United States.
  • Molotkova A; Department of Pathology and Cell Biology, Columbia University, New York, United States.
  • Quintero M; Department of Pathology and Cell Biology, Columbia University, New York, United States.
  • Mela A; Department of Pathology and Cell Biology, Columbia University, New York, United States.
  • Biswas S; Department of Neurology, Columbia University Irving Medical Center, New York, United States.
  • Feng H; Department of Department of Systems Biology, Columbia University Irving Medical Center, New York, United States.
  • Zhang C; Department of Department of Systems Biology, Columbia University Irving Medical Center, New York, United States.
  • Canoll P; Department of Pathology and Cell Biology, Columbia University, New York, United States.
  • Hargus G; Department of Pathology and Cell Biology, Columbia University, New York, United States.
  • Agalliu D; Department of Pathology and Cell Biology, Columbia University, New York, United States.
  • Gogos JA; Department of Neurology, Columbia University Irving Medical Center, New York, United States.
  • Au E; Department of Cellular Physiology and Biophysics, Columbia University, New York, United States.
Elife ; 102021 04 27.
Article in En | MEDLINE | ID: mdl-33904394
ABSTRACT
Cortical interneurons establish inhibitory microcircuits throughout the neocortex and their dysfunction has been implicated in epilepsy and neuropsychiatric diseases. Developmentally, interneurons migrate from a distal progenitor domain in order to populate the neocortex - a process that occurs at a slower rate in humans than in mice. In this study, we sought to identify factors that regulate the rate of interneuron maturation across the two species. Using embryonic mouse development as a model system, we found that the process of initiating interneuron migration is regulated by blood vessels of the medial ganglionic eminence (MGE), an interneuron progenitor domain. We identified two endothelial cell-derived paracrine factors, SPARC and SerpinE1, that enhance interneuron migration in mouse MGE explants and organotypic cultures. Moreover, pre-treatment of human stem cell-derived interneurons (hSC-interneurons) with SPARC and SerpinE1 prior to transplantation into neonatal mouse cortex enhanced their migration and morphological elaboration in the host cortex. Further, SPARC and SerpinE1-treated hSC-interneurons also exhibited more mature electrophysiological characteristics compared to controls. Overall, our studies suggest a critical role for CNS vasculature in regulating interneuron developmental maturation in both mice and humans.
Subject(s)
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Osteonectin / Cell Movement / Cerebral Cortex / Plasminogen Activator Inhibitor 1 / Neurogenesis / Induced Pluripotent Stem Cells / Neural Stem Cells / Interneurons / Median Eminence Type of study: Prognostic_studies Limits: Animals / Humans Language: En Journal: Elife Year: 2021 Document type: Article Affiliation country: United States

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Osteonectin / Cell Movement / Cerebral Cortex / Plasminogen Activator Inhibitor 1 / Neurogenesis / Induced Pluripotent Stem Cells / Neural Stem Cells / Interneurons / Median Eminence Type of study: Prognostic_studies Limits: Animals / Humans Language: En Journal: Elife Year: 2021 Document type: Article Affiliation country: United States