Uncovering mechanisms of CHD2-associated epilepsy using human cortical organoids
利用人类皮质类器官揭示 CHD2 相关癫痫的机制
基本信息
- 批准号:10791545
- 负责人:
- 金额:$ 41.25万
- 依托单位:
- 依托单位国家:美国
- 项目类别:
- 财政年份:2023
- 资助国家:美国
- 起止时间:2023-09-15 至 2025-08-31
- 项目状态:未结题
- 来源:
- 关键词:AddressAffectAnimal ModelBiological ModelsBrainCHD1 geneCHD4 geneCHD7 geneCatalogsCell ProliferationCellsChromatinDNA BindingDataDefectDevelopmentDevelopmental Delay DisordersDiseaseEmbryoEnzymesEpilepsyExhibitsFamilyForebrain DevelopmentFunctional disorderGene Expression RegulationGenerationsGenesGoalsHeterozygoteHistonesHumanImmunohistochemistryImpairmentIn VitroIntellectual functioning disabilityInterneuronsInterventionKnowledgeMedialModelingMolecularMusMutationNOR MouseNeocortexNeurodevelopmental DisorderNeurogliaNeuronsNucleosomesOrganoidsPathogenicityPatient RecruitmentsPatientsPhenotypePlayProliferatingProliferation MarkerProsencephalonProtein FamilyRadialRoleTestingTherapeuticTherapeutic InterventionTimeVariantautism spectrum disordercell typechildhood epilepsychromatin remodelingdisease phenotypedisease-causing mutationepileptic encephalopathieshelicasehuman embryonic stem cellinduced pluripotent stem cellinnovationinsightknock-downmouse modelmultiple omicsmutantneocorticalnerve stem cellnervous system disorderneuralneurodevelopmentneuronal excitabilitynew technologynovelprogenitorsingle cell analysissingle-cell RNA sequencingstem cell differentiationstem cellstranscription factor
项目摘要
PROJECT SUMMARY/ABSTRACT
Mutations in CHD2 gene are associated with developmental and epileptic encephalopathy (DEE), a severe form
of childhood epilepsy. CHD2 belongs to the chromodomain helicase DNA binding (CHD) family of ATP
dependent chromatin remodelers known to play a critical role in neurodevelopment via chromatin organization
and gene regulation; and is the only one among 9 other CHD (CHD1-CHD9) family proteins that causes a brain
restricted phenotype suggesting its non-redundant role in neurodevelopmental disorders. Studies in mice have
shown that knockdown of CHD2 in embryonic cortex decreases the amplification of radial glial cells (RGCs) and
promotes the generation of intermediate progenitor cells (IPs). Moreover, Chd2+/- mice, the only known
heterozygous mouse line to date to mimic CHD2 haploinsufficiency in human, exhibits reduced number of
GABAergic interneurons, disrupts cell proliferation in the developing forebrain and displays a shift in neuronal
excitability. Additionally, a study using 2D culture of medial ganglionic eminence (MGE) like progenitors and
cortical interneurons derived from human embryonic stem cells (hESCs) has shown that CHD2 deficiency
impairs interneuron development and alters its functions. Though these studies have been critical towards our
fundamental understanding of the role of CHD2 in neurodevelopment, neither mice nor 2D neuronal cultures
represent the full breadth of human brain development and leave a critical gap in knowledge about the human
specific cellular and molecular mechanisms associated with CHD2 mutation. To fill this gap, we propose to use
patient derived induced pluripotent stem cells (iPSCs) differentiated into human cortical organoids (hCO). hCO
closely mimic the development of human forebrain and is an innovative model system to study patient specific
cellular and molecular mechanisms underlying a disease-causing mutation. Based on our preliminary data using
single cell RNA sequencing (scRNA-seq) in hCO, we hypothesize that CHD2 mutation leads to defects in
cortical progenitor proliferation and differentiation during neurodevelopment contributing to altered
cortical circuitry and epilepsy. To test our hypothesis, we will 1) determine the cellular impact of CHD2
mutation in cortical development and epilepsy using immunohistochemistry (Aim 1) and 2) delineate the
molecular mechanisms of CHD2 mutation in neurodevelopment and epilepsy by performing multiome analysis
(scRNA-seq + scATAC-seq) (Aim 2). The proposed study will not only increase our understanding of the
childhood epilepsy and related neurodevelopmental disorders but also provide greater insight into novel in vitro
cortical organoid models to study a number of pathogenic variants. To our knowledge, this is the first time that
patient derived iPSCs differentiated into cortical organoids have been proposed to study CHD2 mutation in
humans, and in our model system, we will be able to identify previously unknown cellular and molecular
mechanisms underlying CHD2 associated epilepsy paving the way for development of better therapeutic
intervention strategies.
项目概要/摘要
CHD2 基因突变与发育性癫痫性脑病 (DEE) 相关,这是一种严重的形式
儿童癫痫症。 CHD2 属于 ATP 染色质结构域解旋酶 DNA 结合 (CHD) 家族
已知依赖染色质重塑剂通过染色质组织在神经发育中发挥关键作用
和基因调控;并且是其他 9 种 CHD (CHD1-CHD9) 家族蛋白中唯一一种会导致大脑
限制表型表明其在神经发育障碍中的非冗余作用。对小鼠的研究有
研究表明,胚胎皮质中 CHD2 的敲低会降低放射状胶质细胞 (RGC) 的扩增,
促进中间祖细胞(IP)的产生。此外,Chd2+/- 小鼠是唯一已知的
迄今为止,模拟人类CHD2单倍体不足的杂合小鼠系表现出减少的数量
GABA能中间神经元,破坏发育中前脑的细胞增殖,并显示神经元的变化
兴奋性。此外,一项使用内侧神经节隆起 (MGE) 的 2D 培养(如祖细胞和
来自人类胚胎干细胞 (hESC) 的皮质中间神经元表明,CHD2 缺陷
损害中间神经元发育并改变其功能。尽管这些研究对我们来说至关重要
对 CHD2 在神经发育中的作用的基本了解,无论是小鼠还是 2D 神经元培养物
代表了人类大脑发育的全部范围,并在关于人类的知识上留下了一个关键的空白
与 CHD2 突变相关的特定细胞和分子机制。为了填补这一空白,我们建议使用
患者来源的诱导多能干细胞 (iPSC) 分化为人类皮质类器官 (hCO)。二氧化碳
密切模仿人类前脑的发育,是研究患者特异性的创新模型系统
致病突变的细胞和分子机制。根据我们的初步数据使用
hCO 中的单细胞 RNA 测序 (scRNA-seq),我们假设 CHD2 突变导致缺陷
神经发育过程中皮质祖细胞的增殖和分化导致改变
皮质电路和癫痫。为了检验我们的假设,我们将 1) 确定 CHD2 的细胞影响
使用免疫组织化学(目标 1)和 2)描述皮质发育和癫痫的突变
通过多组分析研究CHD2突变在神经发育和癫痫中的分子机制
(scRNA-seq + scATAC-seq)(目标 2)。拟议的研究不仅会增加我们对
儿童癫痫和相关的神经发育障碍,而且还提供了对新型体外实验的更深入的了解
皮质类器官模型来研究许多致病变异。据我们所知,这是第一次
已提出将患者来源的 iPSC 分化为皮质类器官来研究 CHD2 突变
人类,在我们的模型系统中,我们将能够识别以前未知的细胞和分子
CHD2相关癫痫的潜在机制为开发更好的治疗方法铺平道路
干预策略。
项目成果
期刊论文数量(0)
专著数量(0)
科研奖励数量(0)
会议论文数量(0)
专利数量(0)
数据更新时间:{{ journalArticles.updateTime }}
{{
item.title }}
{{ item.translation_title }}
- DOI:
{{ item.doi }} - 发表时间:
{{ item.publish_year }} - 期刊:
- 影响因子:{{ item.factor }}
- 作者:
{{ item.authors }} - 通讯作者:
{{ item.author }}
数据更新时间:{{ journalArticles.updateTime }}
{{ item.title }}
- 作者:
{{ item.author }}
数据更新时间:{{ monograph.updateTime }}
{{ item.title }}
- 作者:
{{ item.author }}
数据更新时间:{{ sciAawards.updateTime }}
{{ item.title }}
- 作者:
{{ item.author }}
数据更新时间:{{ conferencePapers.updateTime }}
{{ item.title }}
- 作者:
{{ item.author }}
数据更新时间:{{ patent.updateTime }}
Vanesa Nieto Estevez其他文献
Vanesa Nieto Estevez的其他文献
{{
item.title }}
{{ item.translation_title }}
- DOI:
{{ item.doi }} - 发表时间:
{{ item.publish_year }} - 期刊:
- 影响因子:{{ item.factor }}
- 作者:
{{ item.authors }} - 通讯作者:
{{ item.author }}
相似国自然基金
肾—骨应答调控骨骼VDR/RXR对糖尿病肾病动物模型FGF23分泌的影响及中药的干预作用
- 批准号:82074395
- 批准年份:2020
- 资助金额:55 万元
- 项目类别:面上项目
基于细胞自噬调控的苦参碱对多囊肾小鼠动物模型肾囊肿形成的影响和机制研究
- 批准号:
- 批准年份:2019
- 资助金额:33 万元
- 项目类别:地区科学基金项目
靶向诱导merlin/p53协同性亚细胞穿梭对听神经瘤在体生长的影响
- 批准号:81800898
- 批准年份:2018
- 资助金额:21.0 万元
- 项目类别:青年科学基金项目
伪狂犬病病毒激活三叉神经节细胞对其NF-кB和PI3K/Akt信号转导通路影响的分子机制研究
- 批准号:31860716
- 批准年份:2018
- 资助金额:39.0 万元
- 项目类别:地区科学基金项目
基于中枢胰岛素抵抗探讨自噬失调对肾虚阿尔茨海默的影响及机制研究
- 批准号:81803854
- 批准年份:2018
- 资助金额:21.0 万元
- 项目类别:青年科学基金项目
相似海外基金
Endothelial Cell Reprogramming in Familial Intracranial Aneurysm
家族性颅内动脉瘤的内皮细胞重编程
- 批准号:
10595404 - 财政年份:2023
- 资助金额:
$ 41.25万 - 项目类别:
Dravet Syndrome Anti-Epileptic Control by Targeting GIRK Channels
通过针对 GIRK 通道进行 Dravet 综合征抗癫痫控制
- 批准号:
10638439 - 财政年份:2023
- 资助金额:
$ 41.25万 - 项目类别:
Novel application of pharmaceutical AMD3100 to reduce risk in opioid use disorder: investigations of a causal relationship between CXCR4 expression and addiction vulnerability
药物 AMD3100 降低阿片类药物使用障碍风险的新应用:CXCR4 表达与成瘾脆弱性之间因果关系的研究
- 批准号:
10678062 - 财政年份:2023
- 资助金额:
$ 41.25万 - 项目类别:
Mechanisms of Metal Ion Homeostasis of Oral Streptococci
口腔链球菌金属离子稳态机制
- 批准号:
10680956 - 财政年份:2023
- 资助金额:
$ 41.25万 - 项目类别:
Design and testing of a novel circumesophageal cuff for chronic bilateral subdiaphragmatic vagal nerve stimulation (sVNS)
用于慢性双侧膈下迷走神经刺激(sVNS)的新型环食管套囊的设计和测试
- 批准号:
10702126 - 财政年份:2023
- 资助金额:
$ 41.25万 - 项目类别: