Developmental deficits of MGE-derived interneurons in the Cntnap2 knockout mouse model of autism spectrum disorder

Interneurons are fundamental cells for maintaining the excitation-inhibition balance in the brain in health and disease. While interneurons have been shown to play a key role in the pathophysiology of autism spectrum disorder (ASD) in adult mice, little is known about how their maturation is altered...

Full description

Bibliographic Details
Main Authors: Noorya Yasmin Ahmed, Rhys Knowles, Lixinyu Liu, Yiming Yan, Xiaohan Li, Ulrike Schumann, Yumeng Wang, Yovina Sontani, Nathan Reynolds, Riccardo Natoli, Jiayu Wen, Isabel Del Pino, Da Mi, Nathalie Dehorter
Format: Article
Language:English
Published: Frontiers Media S.A. 2023-02-01
Series:Frontiers in Cell and Developmental Biology
Subjects:
Online Access:https://www.frontiersin.org/articles/10.3389/fcell.2023.1112062/full
_version_ 1797937339460222976
author Noorya Yasmin Ahmed
Rhys Knowles
Lixinyu Liu
Yiming Yan
Xiaohan Li
Ulrike Schumann
Yumeng Wang
Yovina Sontani
Nathan Reynolds
Riccardo Natoli
Jiayu Wen
Isabel Del Pino
Da Mi
Nathalie Dehorter
author_facet Noorya Yasmin Ahmed
Rhys Knowles
Lixinyu Liu
Yiming Yan
Xiaohan Li
Ulrike Schumann
Yumeng Wang
Yovina Sontani
Nathan Reynolds
Riccardo Natoli
Jiayu Wen
Isabel Del Pino
Da Mi
Nathalie Dehorter
author_sort Noorya Yasmin Ahmed
collection DOAJ
description Interneurons are fundamental cells for maintaining the excitation-inhibition balance in the brain in health and disease. While interneurons have been shown to play a key role in the pathophysiology of autism spectrum disorder (ASD) in adult mice, little is known about how their maturation is altered in the developing striatum in ASD. Here, we aimed to track striatal developing interneurons and elucidate the molecular and physiological alterations in the Cntnap2 knockout mouse model. Using Stereo-seq and single-cell RNA sequencing data, we first characterized the pattern of expression of Cntnap2 in the adult brain and at embryonic stages in the medial ganglionic eminence (MGE), a transitory structure producing most cortical and striatal interneurons. We found that Cntnap2 is enriched in the striatum, compared to the cortex, particularly in the developing striatal cholinergic interneurons. We then revealed enhanced MGE-derived cell proliferation, followed by increased cell loss during the canonical window of developmental cell death in the Cntnap2 knockout mice. We uncovered specific cellular and molecular alterations in the developing Lhx6-expressing cholinergic interneurons of the striatum, which impacts interneuron firing properties during the first postnatal week. Overall, our work unveils some of the mechanisms underlying the shift in the developmental trajectory of striatal interneurons which greatly contribute to the ASD pathogenesis.
first_indexed 2024-04-10T18:43:04Z
format Article
id doaj.art-a02577ef8e674f5ab20f5224e83bb859
institution Directory Open Access Journal
issn 2296-634X
language English
last_indexed 2024-04-10T18:43:04Z
publishDate 2023-02-01
publisher Frontiers Media S.A.
record_format Article
series Frontiers in Cell and Developmental Biology
spelling doaj.art-a02577ef8e674f5ab20f5224e83bb8592023-02-01T11:26:35ZengFrontiers Media S.A.Frontiers in Cell and Developmental Biology2296-634X2023-02-011110.3389/fcell.2023.11120621112062Developmental deficits of MGE-derived interneurons in the Cntnap2 knockout mouse model of autism spectrum disorderNoorya Yasmin Ahmed0Rhys Knowles1Lixinyu Liu2Yiming Yan3Xiaohan Li4Ulrike Schumann5Yumeng Wang6Yovina Sontani7Nathan Reynolds8Riccardo Natoli9Jiayu Wen10Isabel Del Pino11Da Mi12Nathalie Dehorter13The Australian National University, The John Curtin School of Medical Research, Canberra, ACT, AustraliaThe Australian National University, The John Curtin School of Medical Research, Canberra, ACT, AustraliaThe Australian National University, The John Curtin School of Medical Research, Canberra, ACT, AustraliaTsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing, ChinaTsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing, ChinaThe Australian National University, The John Curtin School of Medical Research, Canberra, ACT, AustraliaThe Australian National University, The John Curtin School of Medical Research, Canberra, ACT, AustraliaThe Australian National University, The John Curtin School of Medical Research, Canberra, ACT, AustraliaThe Australian National University, The John Curtin School of Medical Research, Canberra, ACT, AustraliaThe Australian National University, The John Curtin School of Medical Research, Canberra, ACT, AustraliaThe Australian National University, The John Curtin School of Medical Research, Canberra, ACT, AustraliaInstitute of Neurosciences, Spanish National Research Council (CSIC), Sant Joan d’Alacant, SpainTsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing, ChinaThe Australian National University, The John Curtin School of Medical Research, Canberra, ACT, AustraliaInterneurons are fundamental cells for maintaining the excitation-inhibition balance in the brain in health and disease. While interneurons have been shown to play a key role in the pathophysiology of autism spectrum disorder (ASD) in adult mice, little is known about how their maturation is altered in the developing striatum in ASD. Here, we aimed to track striatal developing interneurons and elucidate the molecular and physiological alterations in the Cntnap2 knockout mouse model. Using Stereo-seq and single-cell RNA sequencing data, we first characterized the pattern of expression of Cntnap2 in the adult brain and at embryonic stages in the medial ganglionic eminence (MGE), a transitory structure producing most cortical and striatal interneurons. We found that Cntnap2 is enriched in the striatum, compared to the cortex, particularly in the developing striatal cholinergic interneurons. We then revealed enhanced MGE-derived cell proliferation, followed by increased cell loss during the canonical window of developmental cell death in the Cntnap2 knockout mice. We uncovered specific cellular and molecular alterations in the developing Lhx6-expressing cholinergic interneurons of the striatum, which impacts interneuron firing properties during the first postnatal week. Overall, our work unveils some of the mechanisms underlying the shift in the developmental trajectory of striatal interneurons which greatly contribute to the ASD pathogenesis.https://www.frontiersin.org/articles/10.3389/fcell.2023.1112062/fullinterneuronmaturationstriatumautismCNTNAP2
spellingShingle Noorya Yasmin Ahmed
Rhys Knowles
Lixinyu Liu
Yiming Yan
Xiaohan Li
Ulrike Schumann
Yumeng Wang
Yovina Sontani
Nathan Reynolds
Riccardo Natoli
Jiayu Wen
Isabel Del Pino
Da Mi
Nathalie Dehorter
Developmental deficits of MGE-derived interneurons in the Cntnap2 knockout mouse model of autism spectrum disorder
Frontiers in Cell and Developmental Biology
interneuron
maturation
striatum
autism
CNTNAP2
title Developmental deficits of MGE-derived interneurons in the Cntnap2 knockout mouse model of autism spectrum disorder
title_full Developmental deficits of MGE-derived interneurons in the Cntnap2 knockout mouse model of autism spectrum disorder
title_fullStr Developmental deficits of MGE-derived interneurons in the Cntnap2 knockout mouse model of autism spectrum disorder
title_full_unstemmed Developmental deficits of MGE-derived interneurons in the Cntnap2 knockout mouse model of autism spectrum disorder
title_short Developmental deficits of MGE-derived interneurons in the Cntnap2 knockout mouse model of autism spectrum disorder
title_sort developmental deficits of mge derived interneurons in the cntnap2 knockout mouse model of autism spectrum disorder
topic interneuron
maturation
striatum
autism
CNTNAP2
url https://www.frontiersin.org/articles/10.3389/fcell.2023.1112062/full
work_keys_str_mv AT nooryayasminahmed developmentaldeficitsofmgederivedinterneuronsinthecntnap2knockoutmousemodelofautismspectrumdisorder
AT rhysknowles developmentaldeficitsofmgederivedinterneuronsinthecntnap2knockoutmousemodelofautismspectrumdisorder
AT lixinyuliu developmentaldeficitsofmgederivedinterneuronsinthecntnap2knockoutmousemodelofautismspectrumdisorder
AT yimingyan developmentaldeficitsofmgederivedinterneuronsinthecntnap2knockoutmousemodelofautismspectrumdisorder
AT xiaohanli developmentaldeficitsofmgederivedinterneuronsinthecntnap2knockoutmousemodelofautismspectrumdisorder
AT ulrikeschumann developmentaldeficitsofmgederivedinterneuronsinthecntnap2knockoutmousemodelofautismspectrumdisorder
AT yumengwang developmentaldeficitsofmgederivedinterneuronsinthecntnap2knockoutmousemodelofautismspectrumdisorder
AT yovinasontani developmentaldeficitsofmgederivedinterneuronsinthecntnap2knockoutmousemodelofautismspectrumdisorder
AT nathanreynolds developmentaldeficitsofmgederivedinterneuronsinthecntnap2knockoutmousemodelofautismspectrumdisorder
AT riccardonatoli developmentaldeficitsofmgederivedinterneuronsinthecntnap2knockoutmousemodelofautismspectrumdisorder
AT jiayuwen developmentaldeficitsofmgederivedinterneuronsinthecntnap2knockoutmousemodelofautismspectrumdisorder
AT isabeldelpino developmentaldeficitsofmgederivedinterneuronsinthecntnap2knockoutmousemodelofautismspectrumdisorder
AT dami developmentaldeficitsofmgederivedinterneuronsinthecntnap2knockoutmousemodelofautismspectrumdisorder
AT nathaliedehorter developmentaldeficitsofmgederivedinterneuronsinthecntnap2knockoutmousemodelofautismspectrumdisorder