1
|
Iegiani G, Pallavicini G, Pezzotta A, Brix A, Ferraro A, Gai M, Boda E, Bielas SL, Pistocchi A, Di Cunto F. CITK modulates BRCA1 recruitment at DNA double strand breaks sites through HDAC6. Cell Death Dis 2025; 16:320. [PMID: 40254670 PMCID: PMC12009987 DOI: 10.1038/s41419-025-07655-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 04/07/2025] [Accepted: 04/10/2025] [Indexed: 04/22/2025]
Abstract
Citron Kinase (CITK) is a protein encoded by the CIT gene, whose pathogenic variants underlie microcephalic phenotypes that characterize MCPH17 syndrome. In neural progenitors, CITK loss leads to microtubule instability, resulting in mitotic spindle positioning defects, cytokinesis failure, and accumulation of DNA double strand breaks (DSBs), ultimately resulting in TP53-dependent senescence and apoptosis. Although DNA damage accumulation has been associated with impaired homologous recombination (HR), the role of CITK in this process and whether microtubule dynamics are involved is still unknown. In this report we show that CITK is required for proper BRCA1 localization at sites of DNA DSBs. We found that CITK's scaffolding, rather than its catalytic activity, is necessary for maintaining BRCA1 interphase levels in progenitor cells during neurodevelopment. CITK regulates the nuclear levels of HDAC6, a modulator of both microtubule stability and DNA damage repair. Targeting HDAC6 in CITK-deficient cells increases microtubule stability and recovers BRCA1 localization defects and DNA damage levels to that detected in controls. In addition, the CIT-HDAC6 axis is functionally relevant in a MCPH17 zebrafish model, as HDAC6 targeting recovers the head size phenotype produced by interfering with the CIT orthologue gene. These data provide novel insights into the functional interplay between HR and microtubule dynamics and into the pathogenesis of CITK based MCPH17, which may be relevant for development of therapeutic strategies.
Collapse
Affiliation(s)
- Giorgia Iegiani
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience 'Rita Levi Montalcini', University of Turin, Torino, Italy
| | - Gianmarco Pallavicini
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience 'Rita Levi Montalcini', University of Turin, Torino, Italy
| | - Alex Pezzotta
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milano, Italy
| | - Alessia Brix
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milano, Italy
| | - Alessia Ferraro
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience 'Rita Levi Montalcini', University of Turin, Torino, Italy
| | - Marta Gai
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy
| | - Enrica Boda
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience 'Rita Levi Montalcini', University of Turin, Torino, Italy
| | - Stephanie L Bielas
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI, USA
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Anna Pistocchi
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milano, Italy
| | - Ferdinando Di Cunto
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy.
- Department of Neuroscience 'Rita Levi Montalcini', University of Turin, Torino, Italy.
| |
Collapse
|
2
|
Harkins D, Ali S, Tockovska T, Ciganovic S, Casasbuenas DL, Watanabe S, Ouzikov S, Yuzwa SA. Clonal lineage tracing and transcriptomics of cortical progenitor populations reveal maintenance of differentiation potential. Stem Cell Reports 2025; 20:102418. [PMID: 39952245 PMCID: PMC11960535 DOI: 10.1016/j.stemcr.2025.102418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 01/09/2025] [Accepted: 01/13/2025] [Indexed: 02/17/2025] Open
Abstract
Postnatal neocortical development is a complex period wherein radial glial progenitors (RGPs) complete excitatory neurogenesis and transition to the production of glia. Here, we take advantage of a multi-layered lineage tracing tool pbacBarcode, to examine the contributions of individual cortical RGPs to the postnatal cortex. We reveal that some individual cortical RGPs are multipotent and give rise to olfactory bulb interneurons, astrocytes, and oligodendrocytes in a ∼2:1:1 ratio. We provide evidence that differentiation potential into terminal cell types is maintained as late as post-natal day (P)4, suggesting that a population decline model, as opposed to cell fate restriction, underlies postnatal neocortical development. Moreover, a pool of proliferative intermediary cells, which may represent a multipotent postnatal intermediate progenitor cell population, may contribute to the production of the three major cell types. Lastly, we examine RGP postnatal contribution to oligodendrocytes and show that oligodendrocyte progenitor founder cell production by cortical RGPs is largely complete by P3.
Collapse
Affiliation(s)
- Danyon Harkins
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto. 1 King's College Circle, Toronto, ON M5S 1A8, Canada
| | - Shawar Ali
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto. 1 King's College Circle, Toronto, ON M5S 1A8, Canada
| | - Teodora Tockovska
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto. 1 King's College Circle, Toronto, ON M5S 1A8, Canada; Bioinformatics and HPC Core, Princess Margaret Cancer Research Tower, 101 College Street, Toronto, ON M5G 1L7, Canada
| | - Sara Ciganovic
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto. 1 King's College Circle, Toronto, ON M5S 1A8, Canada
| | - Daniela Lozano Casasbuenas
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto. 1 King's College Circle, Toronto, ON M5S 1A8, Canada
| | - Samuel Watanabe
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto. 1 King's College Circle, Toronto, ON M5S 1A8, Canada
| | - Stephanie Ouzikov
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto. 1 King's College Circle, Toronto, ON M5S 1A8, Canada
| | - Scott A Yuzwa
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto. 1 King's College Circle, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
3
|
Feng X, Du X, Yang X, Chen C, Liang Z, Xu X, Wang Y, Zheng JC, Xia X, Liu J. miR-185-5p regulates the proliferation and differentiation of neural stem/progenitor cells. Front Cell Dev Biol 2024; 12:1510746. [PMID: 39703696 PMCID: PMC11656079 DOI: 10.3389/fcell.2024.1510746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 11/20/2024] [Indexed: 12/21/2024] Open
Abstract
Background MicroRNAs (miRNAs) have emerged as an essential regulator of the cell fate commitment of neural stem/progenitor cells (NPCs), although the impacts of certain miRNAs on NPCs remain vague. The aim of this study is to investigate the regulatory effects of miR-185-5p on the cell fate commitment of NPCs. Methods We investigated the impact of miR-185-5p on the proliferation and differentiation capacities of primary NPCs by manipulating the expression of miR-185-5p using specific mimics and inhibitors. The effects of miR-185-5p on NPCs was confirmed in vivo through stereotactic injection of miR-185-5p antagonists to the brains of mice at postnatal day 1 (P1). Results The expression levels of miR-185-5p kept increasing in the differentiation process of NPCs in vivo and in vitro. Perturbation of miR-185-5p's function showed that miR-185-5p inhibited NPCs' proliferation and promoted embryonic NPCs to differentiate more favorably to the glial lineage. We then validated the anti-proliferation and pro-glial roles of miR-185-5p using NPCs isolated from P1 mouse brains. In vivo study further showed enlarged NPCs pools and inhibited gliogenesis in the brains of P1 mice after animals received antagomir-185-5p. Conclusion Our study suggests miR-185-5p as an important regulator for the proliferation and glial fate commitment of NPCs.
Collapse
Affiliation(s)
- Xuanran Feng
- Department of Anesthesiology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xue Du
- Department of Anesthesiology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xiaoyu Yang
- Department of Anesthesiology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Changqi Chen
- Department of Anesthesiology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhanping Liang
- Department of Anesthesiology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xiaonan Xu
- Department of Anesthesiology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yi Wang
- Translational Research Center, Shanghai Yangzhi Rehabilitation Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jialin C. Zheng
- Department of Anesthesiology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Center for Translational Neurodegeneration and Regenerative Therapy, Tongji Hospital affiliated to Tongji University School of Medicine, Shanghai, China
- Shanghai Frontiers Science Center of Nanocatalytic Medicine, Tongji University, Shanghai, China
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People’s Hospital affiliated to Tongji University School of Medicine, Shanghai, China
- Collaborative Innovation Center for Brain Science, Tongji University, Shanghai, China
| | - Xiaohuan Xia
- Department of Anesthesiology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Center for Translational Neurodegeneration and Regenerative Therapy, Tongji Hospital affiliated to Tongji University School of Medicine, Shanghai, China
- Shanghai Frontiers Science Center of Nanocatalytic Medicine, Tongji University, Shanghai, China
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People’s Hospital affiliated to Tongji University School of Medicine, Shanghai, China
| | - Jianhui Liu
- Department of Anesthesiology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
4
|
Pallavicini G, Moccia A, Iegiani G, Parolisi R, Peirent ER, Berto GE, Lorenzati M, Tshuva RY, Ferraro A, Balzac F, Turco E, Salvi SU, Myklebust HF, Wang S, Eisenberg J, Chitale M, Girgla NS, Boda E, Reiner O, Buffo A, Di Cunto F, Bielas SL. Modeling primary microcephaly with human brain organoids reveals fundamental roles of CIT kinase activity. J Clin Invest 2024; 134:e175435. [PMID: 39316437 PMCID: PMC11527453 DOI: 10.1172/jci175435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 09/06/2024] [Indexed: 09/26/2024] Open
Abstract
Brain size and cellular heterogeneity are tightly regulated by species-specific proliferation and differentiation of multipotent neural progenitor cells (NPCs). Errors in this process are among the mechanisms of primary hereditary microcephaly (MCPH), a group of disorders characterized by reduced brain size and intellectual disability. Biallelic citron rho-interacting serine/threonine kinase (CIT) missense variants that disrupt kinase function (CITKI/KI) and frameshift loss-of-function variants (CITFS/FS) are the genetic basis for MCPH17; however, the function of CIT catalytic activity in brain development and NPC cytokinesis is unknown. Therefore, we created the CitKI/KI mouse model and found that it did not phenocopy human microcephaly, unlike biallelic CitFS/FS animals. Nevertheless, both Cit models exhibited binucleation, DNA damage, and apoptosis. To investigate human-specific mechanisms of CIT microcephaly, we generated CITKI/KI and CITFS/FS human forebrain organoids. We found that CITKI/KI and CITFS/FS organoids lost cytoarchitectural complexity, transitioning from pseudostratified to simple neuroepithelium. This change was associated with defects that disrupted the polarity of NPC cytokinesis, in addition to elevating apoptosis. Together, our results indicate that both CIT catalytic and scaffolding functions in NPC cytokinesis are critical for human corticogenesis. Species differences in corticogenesis and the dynamic 3D features of NPC mitosis underscore the utility of human forebrain organoid models for understanding human microcephaly.
Collapse
Affiliation(s)
- Gianmarco Pallavicini
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience “Rita Levi Montalcini,” University of Turin, Turin, Italy
| | | | - Giorgia Iegiani
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience “Rita Levi Montalcini,” University of Turin, Turin, Italy
| | - Roberta Parolisi
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience “Rita Levi Montalcini,” University of Turin, Turin, Italy
| | - Emily R. Peirent
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Gaia Elena Berto
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience “Rita Levi Montalcini,” University of Turin, Turin, Italy
| | - Martina Lorenzati
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience “Rita Levi Montalcini,” University of Turin, Turin, Italy
| | - Rami Y. Tshuva
- Departments of Molecular Genetics and Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Alessia Ferraro
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience “Rita Levi Montalcini,” University of Turin, Turin, Italy
| | - Fiorella Balzac
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Emilia Turco
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | | | | | | | - Julia Eisenberg
- Department of Human Genetics and
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | | | | | - Enrica Boda
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience “Rita Levi Montalcini,” University of Turin, Turin, Italy
| | - Orly Reiner
- Departments of Molecular Genetics and Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Annalisa Buffo
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience “Rita Levi Montalcini,” University of Turin, Turin, Italy
| | - Ferdinando Di Cunto
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience “Rita Levi Montalcini,” University of Turin, Turin, Italy
| | - Stephanie L. Bielas
- Department of Human Genetics and
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
5
|
Viola V, Chinnappa K, Francis F. Radial glia progenitor polarity in health and disease. Front Cell Dev Biol 2024; 12:1478283. [PMID: 39416687 PMCID: PMC11479994 DOI: 10.3389/fcell.2024.1478283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 09/20/2024] [Indexed: 10/19/2024] Open
Abstract
Radial glia (RG) are the main progenitor cell type in the developing cortex. These cells are highly polarized, with a long basal process spanning the entire thickness of the cortex and acting as a support for neuronal migration. The RG cell terminates by an endfoot that contacts the pial (basal) surface. A shorter apical process also terminates with an endfoot that faces the ventricle, with a primary cilium protruding in the cerebrospinal fluid. These cell domains have particular subcellular compositions that are critical for the correct functioning of RG. When altered, this can affect proper development of the cortex, ultimately leading to cortical malformations, associated with different pathological outcomes. In this review, we focus on the current knowledge concerning the cell biology of these bipolar stem cells and discuss the role of their polarity in health and disease.
Collapse
Affiliation(s)
- Valeria Viola
- Institut du Fer à Moulin, Paris, France
- Institut National de Santé et de Recherche Médicale (INSERM, UMR-S 1270), Paris, France
- Faculty of Science and Engineering, Sorbonne University, Paris, France
| | - Kaviya Chinnappa
- Institut du Fer à Moulin, Paris, France
- Institut National de Santé et de Recherche Médicale (INSERM, UMR-S 1270), Paris, France
- Faculty of Science and Engineering, Sorbonne University, Paris, France
| | - Fiona Francis
- Institut du Fer à Moulin, Paris, France
- Institut National de Santé et de Recherche Médicale (INSERM, UMR-S 1270), Paris, France
- Faculty of Science and Engineering, Sorbonne University, Paris, France
| |
Collapse
|
6
|
De Vincenti AP, Bonafina A, Ledda F, Paratcha G. Lrig1 regulates cell fate specification of glutamatergic neurons via FGF-driven Jak2/Stat3 signaling in cortical progenitors. Development 2024; 151:dev202879. [PMID: 39250533 DOI: 10.1242/dev.202879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 08/07/2024] [Indexed: 09/11/2024]
Abstract
The cell-intrinsic mechanisms underlying the decision of a stem/progenitor cell to either proliferate or differentiate remain incompletely understood. Here, we identify the transmembrane protein Lrig1 as a physiological homeostatic regulator of FGF2-driven proliferation and self-renewal of neural progenitors at early-to-mid embryonic stages of cortical development. We show that Lrig1 is expressed in cortical progenitors (CPs), and its ablation caused expansion and increased proliferation of radial/apical progenitors and of neurogenic transit-amplifying Tbr2+ intermediate progenitors. Notably, our findings identify a previously unreported EGF-independent mechanism through which Lrig1 negatively regulates neural progenitor proliferation by modulating the FGF2-induced IL6/Jak2/Stat3 pathway, a molecular cascade that plays a pivotal role in the generation and maintenance of CPs. Consistently, Lrig1 knockout mice showed a significant increase in the density of pyramidal glutamatergic neurons placed in superficial layers 2 and 3 of the postnatal neocortex. Together, these results support a model in which Lrig1 regulates cortical neurogenesis by influencing the cycling activity of a set of progenitors that are temporally specified to produce upper layer glutamatergic neurons.
Collapse
Affiliation(s)
- Ana Paula De Vincenti
- Laboratorio de Neurociencia Molecular y Celular, Instituto de Biología Celular y Neurociencias (IBCN)-CONICET-UBA, Facultad de Medicina. Universidad de Buenos Aires (UBA), Buenos Aires CP1121, Argentina
| | - Antonela Bonafina
- Laboratorio de Neurociencia Molecular y Celular, Instituto de Biología Celular y Neurociencias (IBCN)-CONICET-UBA, Facultad de Medicina. Universidad de Buenos Aires (UBA), Buenos Aires CP1121, Argentina
- Fundación Instituto Leloir, Instituto de Investigaciones Bioquímicas de Buenos Aires, Buenos Aires C1405 BWE, Argentina
| | - Fernanda Ledda
- Fundación Instituto Leloir, Instituto de Investigaciones Bioquímicas de Buenos Aires, Buenos Aires C1405 BWE, Argentina
| | - Gustavo Paratcha
- Laboratorio de Neurociencia Molecular y Celular, Instituto de Biología Celular y Neurociencias (IBCN)-CONICET-UBA, Facultad de Medicina. Universidad de Buenos Aires (UBA), Buenos Aires CP1121, Argentina
| |
Collapse
|
7
|
Ma D, Lin KY, Suresh D, Lin J, Gujar MR, Aung HY, Tan YS, Gao Y, Vincent AS, Chen T, Wang H. Arl2 GTPase associates with the centrosomal protein Cdk5rap2 to regulate cortical development via microtubule organization. PLoS Biol 2024; 22:e3002751. [PMID: 39137170 PMCID: PMC11321591 DOI: 10.1371/journal.pbio.3002751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 07/11/2024] [Indexed: 08/15/2024] Open
Abstract
ADP ribosylation factor-like GTPase 2 (Arl2) is crucial for controlling mitochondrial fusion and microtubule assembly in various organisms. Arl2 regulates the asymmetric division of neural stem cells in Drosophila via microtubule growth. However, the function of mammalian Arl2 during cortical development was unknown. Here, we demonstrate that mouse Arl2 plays a new role in corticogenesis via regulating microtubule growth, but not mitochondria functions. Arl2 knockdown (KD) leads to impaired proliferation of neural progenitor cells (NPCs) and neuronal migration. Arl2 KD in mouse NPCs significantly diminishes centrosomal microtubule growth and delocalization of centrosomal proteins Cdk5rap2 and γ-tubulin. Moreover, Arl2 physically associates with Cdk5rap2 by in silico prediction using AlphaFold multimer, which was validated by co-immunoprecipitation and proximity ligation assay. Remarkably, Cdk5rap2 overexpression significantly rescues the neurogenesis defects caused by Arl2 KD. Therefore, Arl2 plays an important role in mouse cortical development through microtubule growth via the centrosomal protein Cdk5rap2.
Collapse
Affiliation(s)
- Dongliang Ma
- Program in Neuroscience and Behavioural Disorders, Duke-NUS Medical School, Singapore
| | - Kun-Yang Lin
- Program in Neuroscience and Behavioural Disorders, Duke-NUS Medical School, Singapore
| | - Divya Suresh
- Program in Neuroscience and Behavioural Disorders, Duke-NUS Medical School, Singapore
| | - Jiaen Lin
- Program in Neuroscience and Behavioural Disorders, Duke-NUS Medical School, Singapore
| | - Mahekta R. Gujar
- Program in Neuroscience and Behavioural Disorders, Duke-NUS Medical School, Singapore
| | - Htet Yamin Aung
- Program in Neuroscience and Behavioural Disorders, Duke-NUS Medical School, Singapore
| | - Ye Sing Tan
- Program in Neuroscience and Behavioural Disorders, Duke-NUS Medical School, Singapore
| | - Yang Gao
- Program in Neuroscience and Behavioural Disorders, Duke-NUS Medical School, Singapore
| | - Anselm S. Vincent
- Program in Neuroscience and Behavioural Disorders, Duke-NUS Medical School, Singapore
| | - Teng Chen
- College of Forensic Medicine, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, PR China
- The Key Laboratory of Health Ministry for Forensic Science, Xi’an Jiaotong University, Shaanxi, PR China
| | - Hongyan Wang
- Program in Neuroscience and Behavioural Disorders, Duke-NUS Medical School, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Integrative Sciences and Engineering Programme, National University of Singapore, Singapore
| |
Collapse
|
8
|
Garber A, Weingarten LS, Abreu NJ, Elloumi HZ, Haack T, Hildebrant C, Martínez-Gil N, Mathews J, Müller AJ, Valenzuela Palafoll I, Steigerwald C, Chung WK. Rare predicted deleterious FEZF2 variants are associated with a neurodevelopmental phenotype. Am J Med Genet A 2024; 194:e63578. [PMID: 38425142 PMCID: PMC11161304 DOI: 10.1002/ajmg.a.63578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 02/07/2024] [Accepted: 02/17/2024] [Indexed: 03/02/2024]
Abstract
FEZF2 encodes a transcription factor critical to neurodevelopment that regulates other neurodevelopment genes. Rare variants in FEZF2 have previously been suggested to play a role in autism, and cases of 3p14 microdeletions that include FEZF2 share a neurodevelopmental phenotype including mild dysmorphic features and intellectual disability. We identified seven heterozygous predicted deleterious variants in FEZF2 (three frameshifts, one recurrent missense in two independent cases, one nonsense, and one complete gene deletion) in unrelated individuals with neurodevelopmental disorders including developmental delay/intellectual disability, autism, and/or attention-deficit/hyperactivity. Variants were confirmed to be de novo in five of seven cases and paternally inherited from an affected father in one. Predicted deleterious variants in FEZF2 may affect the expression of genes that are involved in fate choice pathways in developing neurons, and thus contribute to the neurodevelopmental phenotype. Future studies are needed to clarify the mechanism by which FEZF2 leads to this neurodevelopmental disorder.
Collapse
Affiliation(s)
- Alison Garber
- Department of Pediatrics, Columbia University, New York, New York, USA
| | - Lisa S Weingarten
- Department of Pediatrics, Columbia University, New York, New York, USA
| | - Nicolas J Abreu
- Department of Neurology, NYU Grossman School of Medicine, New York, New York, USA
| | | | - Tobias Haack
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Clara Hildebrant
- Department of Pediatrics, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Núria Martínez-Gil
- Department of Clinical and Molecular Genetics, Vall d'Hebron University Hospital, Barcelona, Spain
- Medical Genetics Group, Vall d'Hebron Research Institute, Barcelona, Spain
| | - Jennifer Mathews
- Department of Pediatrics, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Amelie Johanna Müller
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Irene Valenzuela Palafoll
- Department of Clinical and Molecular Genetics, Vall d'Hebron University Hospital, Barcelona, Spain
- Medical Genetics Group, Vall d'Hebron Research Institute, Barcelona, Spain
| | - Connolly Steigerwald
- Department of Neurology, NYU Grossman School of Medicine, New York, New York, USA
| | - Wendy K Chung
- Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
9
|
Liu Z, Shen X, Lin K, Wang F, Gao J, Yao Y, Sun J. Balloon cells in malformations of cortical development: friends or foes? ACTA EPILEPTOLOGICA 2024; 6:20. [PMID: 40217486 PMCID: PMC11960319 DOI: 10.1186/s42494-024-00164-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 04/06/2024] [Indexed: 04/14/2025] Open
Abstract
Balloon cells (BCs) are specific pathological marker of cortical malformations during brain development, often associated with epilepsy and development delay. Although a large number of studies have investigated the role of BCs in these diseases, the specific function of BCs as either epileptogenic or antiepileptic remains controversial. Therefore, we reviewed literatures on BCs, delved into the molecular mechanisms and signaling pathways, and updated their profile in several aspects. Firstly, BCs are heterogeneous and some of them show progenitor/stem cell characteristics. Secondly, BCs are relatively silent in electrophysiology but not completely isolated from their surroundings. Notably, abnormal mTOR signaling and aberrant immunogenic process have been observed within BCs-containing malformations of cortical development (MCDs). The question whether BCs function as the evildoer or the defender in BCs-containing MCDs is further discussed. Importantly, this review provides perspectives on future investigations of the potential role of BCs in epilepsy.
Collapse
Affiliation(s)
- Zili Liu
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, 518055, China
- The Brain Cognition and Brain Disease Institute, SIAT, CAS, Shenzhen, 518055, China
- HH-SIAT Joint Center for Epilepsy Research, Fujian Medical University Affiliated Xiamen Humanity Hospital, Xiamen, 361003, China
| | - Xuefeng Shen
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, 518055, China
- The Brain Cognition and Brain Disease Institute, SIAT, CAS, Shenzhen, 518055, China
- HH-SIAT Joint Center for Epilepsy Research, Fujian Medical University Affiliated Xiamen Humanity Hospital, Xiamen, 361003, China
| | - Kaomin Lin
- Epilepsy Center, Fujian Medical University Affiliated Xiamen Humanity Hospital, Xiamen, 361003, China
- HH-SIAT Joint Center for Epilepsy Research, Fujian Medical University Affiliated Xiamen Humanity Hospital, Xiamen, 361003, China
| | - Fengpeng Wang
- Epilepsy Center, Fujian Medical University Affiliated Xiamen Humanity Hospital, Xiamen, 361003, China
- HH-SIAT Joint Center for Epilepsy Research, Fujian Medical University Affiliated Xiamen Humanity Hospital, Xiamen, 361003, China
| | - Jin Gao
- Department of Pathology, Fujian Medical University Affiliated Xiamen Humanity Hospital, Xiamen, 361003, China
- HH-SIAT Joint Center for Epilepsy Research, Fujian Medical University Affiliated Xiamen Humanity Hospital, Xiamen, 361003, China
| | - Yi Yao
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, 518055, China.
- Epilepsy Center, Fujian Medical University Affiliated Xiamen Humanity Hospital, Xiamen, 361003, China.
- HH-SIAT Joint Center for Epilepsy Research, Fujian Medical University Affiliated Xiamen Humanity Hospital, Xiamen, 361003, China.
| | - Jianyuan Sun
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, 518055, China.
- The Brain Cognition and Brain Disease Institute, SIAT, CAS, Shenzhen, 518055, China.
- HH-SIAT Joint Center for Epilepsy Research, Fujian Medical University Affiliated Xiamen Humanity Hospital, Xiamen, 361003, China.
| |
Collapse
|
10
|
Phalnikar K, Srividya M, Mythri SV, Vasavi NS, Ganguly A, Kumar A, S P, Kalia K, Mishra SS, Dhanya SK, Paul P, Holla B, Ganesh S, Reddy PC, Sud R, Viswanath B, Muralidharan B. Altered neuroepithelial morphogenesis and migration defects in iPSC-derived cerebral organoids and 2D neural stem cells in familial bipolar disorder. OXFORD OPEN NEUROSCIENCE 2024; 3:kvae007. [PMID: 38638145 PMCID: PMC11024480 DOI: 10.1093/oons/kvae007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 01/26/2024] [Accepted: 02/26/2024] [Indexed: 04/20/2024]
Abstract
Bipolar disorder (BD) is a severe mental illness that can result from neurodevelopmental aberrations, particularly in familial BD, which may include causative genetic variants. In the present study, we derived cortical organoids from BD patients and healthy (control) individuals from a clinically dense family in the Indian population. Our data reveal that the patient organoids show neurodevelopmental anomalies, including organisational, proliferation and migration defects. The BD organoids show a reduction in both the number of neuroepithelial buds/cortical rosettes and the ventricular zone size. Additionally, patient organoids show a lower number of SOX2-positive and EdU-positive cycling progenitors, suggesting a progenitor proliferation defect. Further, the patient neurons show abnormal positioning in the ventricular/intermediate zone of the neuroepithelial bud. Transcriptomic analysis of control and patient organoids supports our cellular topology data and reveals dysregulation of genes crucial for progenitor proliferation and neuronal migration. Lastly, time-lapse imaging of neural stem cells in 2D in vitro cultures reveals abnormal cellular migration in BD samples. Overall, our study pinpoints a cellular and molecular deficit in BD patient-derived organoids and neural stem cell cultures.
Collapse
Affiliation(s)
- Kruttika Phalnikar
- Institute for Stem Cell Science and Regenerative Medicine (inStem), GKVK - Post, Bellary Road, Bengaluru, Karnataka, India-560065
| | - M Srividya
- National Institute of Mental Health and Neurosciences (NIMHANS), Hosur Road Bengaluru, Karnataka, India-560029
| | - S V Mythri
- National Institute of Mental Health and Neurosciences (NIMHANS), Hosur Road Bengaluru, Karnataka, India-560029
| | - N S Vasavi
- National Institute of Mental Health and Neurosciences (NIMHANS), Hosur Road Bengaluru, Karnataka, India-560029
| | - Archisha Ganguly
- Institute for Stem Cell Science and Regenerative Medicine (inStem), GKVK - Post, Bellary Road, Bengaluru, Karnataka, India-560065
| | - Aparajita Kumar
- Institute for Stem Cell Science and Regenerative Medicine (inStem), GKVK - Post, Bellary Road, Bengaluru, Karnataka, India-560065
| | - Padmaja S
- Institute for Stem Cell Science and Regenerative Medicine (inStem), GKVK - Post, Bellary Road, Bengaluru, Karnataka, India-560065
| | - Kishan Kalia
- Institute for Stem Cell Science and Regenerative Medicine (inStem), GKVK - Post, Bellary Road, Bengaluru, Karnataka, India-560065
| | - Srishti S Mishra
- Institute for Stem Cell Science and Regenerative Medicine (inStem), GKVK - Post, Bellary Road, Bengaluru, Karnataka, India-560065
| | - Sreeja Kumari Dhanya
- Institute for Stem Cell Science and Regenerative Medicine (inStem), GKVK - Post, Bellary Road, Bengaluru, Karnataka, India-560065
| | - Pradip Paul
- National Institute of Mental Health and Neurosciences (NIMHANS), Hosur Road Bengaluru, Karnataka, India-560029
| | - Bharath Holla
- National Institute of Mental Health and Neurosciences (NIMHANS), Hosur Road Bengaluru, Karnataka, India-560029
| | - Suhas Ganesh
- National Institute of Mental Health and Neurosciences (NIMHANS), Hosur Road Bengaluru, Karnataka, India-560029
| | - Puli Chandramouli Reddy
- Centre of Excellence in Epigenetics, Department of Life Sciences, Shiv Nadar Institution of Eminence, Delhi-NCR, India-201314
| | - Reeteka Sud
- National Institute of Mental Health and Neurosciences (NIMHANS), Hosur Road Bengaluru, Karnataka, India-560029
| | - Biju Viswanath
- National Institute of Mental Health and Neurosciences (NIMHANS), Hosur Road Bengaluru, Karnataka, India-560029
| | - Bhavana Muralidharan
- Institute for Stem Cell Science and Regenerative Medicine (inStem), GKVK - Post, Bellary Road, Bengaluru, Karnataka, India-560065
| |
Collapse
|
11
|
Sullivan D, Vaglio BJ, Cararo-Lopes MM, Wong RDP, Graudejus O, Firestein BL. Stretch-Induced Injury Affects Cortical Neuronal Networks in a Time- and Severity-Dependent Manner. Ann Biomed Eng 2024; 52:1021-1038. [PMID: 38294641 DOI: 10.1007/s10439-023-03438-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 12/22/2023] [Indexed: 02/01/2024]
Abstract
Traumatic brain injury (TBI) is the leading cause of accident-related death and disability in the world and can lead to long-term neuropsychiatric symptoms, such as a decline in cognitive function and neurodegeneration. TBI includes primary and secondary injury, with head trauma and deformation of the brain caused by the physical force of the impact as primary injury, and cellular and molecular cascades that lead to cell death as secondary injury. Currently, there is no treatment for TBI-induced cell damage and neural circuit dysfunction in the brain, and thus, it is important to understand the underlying cellular mechanisms that lead to cell damage. In the current study, we use stretchable microelectrode arrays (sMEAs) to model the primary injury of TBI to study the electrophysiological effects of physically injuring cortical cells. We recorded electrophysiological activity before injury and then stretched the flexible membrane of the sMEAs to injure the cells to varying degrees. At 1, 24, and 72 h post-stretch, we recorded activity to analyze differences in spike rate, Fano factor, burstlet rate, burstlet width, synchrony of firing, local network efficiency, and Q statistic. Our results demonstrate that mechanical injury changes the firing properties of cortical neuron networks in culture in a time- and severity-dependent manner. Our results suggest that changes to electrophysiological properties after stretch are dependent on the strength of synchronization between neurons prior to injury.
Collapse
Affiliation(s)
- Dylan Sullivan
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
- Cell and Developmental Biology Graduate Program, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Brandon J Vaglio
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
- Biomedical Engineering Graduate Program, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Marina M Cararo-Lopes
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
- Cell and Developmental Biology Graduate Program, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Ruben D Ponce Wong
- BioMedical Sustainable Elastic Electronic Devices (BMSEED), Mesa, AZ, USA
| | - Oliver Graudejus
- BioMedical Sustainable Elastic Electronic Devices (BMSEED), Mesa, AZ, USA
- School of Molecular Science, Arizona State University, Tempe, AZ, USA
| | - Bonnie L Firestein
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ, 08854-8082, USA.
| |
Collapse
|
12
|
Gavrish M, Kustova A, Celis Suescún JC, Bessa P, Mitina N, Tarabykin V. Molecular mechanisms of corpus callosum development: a four-step journey. Front Neuroanat 2024; 17:1276325. [PMID: 38298831 PMCID: PMC10827913 DOI: 10.3389/fnana.2023.1276325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 12/18/2023] [Indexed: 02/02/2024] Open
Abstract
The Corpus Callosum (CC) is a bundle of axons connecting the cerebral hemispheres. It is the most recent structure to have appeared during evolution of placental mammals. Its development is controlled by a very complex interplay of many molecules. In humans it contains almost 80% of all commissural axons in the brain. The formation of the CC can be divided into four main stages, each controlled by numerous intracellular and extracellular molecular factors. First, a newborn neuron has to specify an axon, leave proliferative compartments, the Ventricular Zone (VZ) and Subventricular Zone (SVZ), migrate through the Intermediate Zone (IZ), and then settle at the Cortical Plate (CP). During the second stage, callosal axons navigate toward the midline within a compact bundle. Next stage is the midline crossing into contralateral hemisphere. The last step is targeting a defined area and synapse formation. This review provides an insight into these four phases of callosal axons development, as well as a description of the main molecular players involved.
Collapse
Affiliation(s)
- Maria Gavrish
- Laboratory of Genetics of Brain Development, Research Institute of Neurosciences, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Angelina Kustova
- Laboratory of Genetics of Brain Development, Research Institute of Neurosciences, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Juan C. Celis Suescún
- Laboratory of Genetics of Brain Development, Research Institute of Neurosciences, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Paraskevi Bessa
- Charité Hospital, Institute of Cell Biology and Neurobiology, Berlin, Germany
| | - Natalia Mitina
- Laboratory of Genetics of Brain Development, Research Institute of Neurosciences, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Victor Tarabykin
- Charité Hospital, Institute of Cell Biology and Neurobiology, Berlin, Germany
| |
Collapse
|
13
|
Human cytomegalovirus pUL97 upregulates SOCS3 expression via transcription factor RFX7 in neural progenitor cells. PLoS Pathog 2023; 19:e1011166. [PMID: 36753521 PMCID: PMC9942973 DOI: 10.1371/journal.ppat.1011166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 02/21/2023] [Accepted: 01/30/2023] [Indexed: 02/09/2023] Open
Abstract
Congenital human cytomegalovirus (HCMV) infection causes severe damage to the fetal brain, and the underlying mechanisms remain elusive. Cytokine signaling is delicately controlled in the fetal central nervous system to ensure proper development. Here we show that suppressor of cytokine signaling 3 (SOCS3), a negative feedback regulator of the IL-6 cytokine family signaling, was upregulated during HCMV infection in primary neural progenitor cells (NPCs) with a biphasic expression pattern. From viral protein screening, pUL97 emerged as the viral factor responsible for prolonged SOCS3 upregulation. Further, by proteomic analysis of the pUL97-interacting host proteins, regulatory factor X 7 (RFX7) was identified as the transcription factor responsible for the regulation. Depletion of either pUL97 or RFX7 prevented the HCMV-induced SOCS3 upregulation in NPCs. With a promoter-luciferase activity assay, we demonstrated that the pUL97 kinase activity and RFX7 were required for SOCS3 upregulation. Moreover, the RFX7 phosphorylation level was increased by either UL97-expressing or HCMV-infection in NPCs, suggesting that pUL97 induces RFX7 phosphorylation to drive SOCS3 transcription. We further revealed that elevated SOCS3 expression impaired NPC proliferation and migration in vitro and caused NPCs migration defects in vivo. Taken together, these findings uncover a novel regulatory mechanism of sustained SOCS3 expression in HCMV-infected NPCs, which perturbs IL-6 cytokine family signaling, leads to NPCs proliferation and migration defects, and consequently affects fetal brain development.
Collapse
|
14
|
Nam HS, Capecchi MR. Lrig1 expression identifies quiescent stem cells in the ventricular-subventricular zone from postnatal development to adulthood and limits their persistent hyperproliferation. Neural Dev 2023; 18:1. [PMID: 36631891 PMCID: PMC9832784 DOI: 10.1186/s13064-022-00169-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 11/26/2022] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND We previously identified Leucine-rich repeats and immunoglobulin-like domains 1 (Lrig1) as a marker of long-term neurogenic stem cells in the lateral wall of the adult mouse brain. The morphology of the stem cells thus identified differed from the canonical B1 type stem cells, raising a question about their cellular origin. Thus, we investigated the development of these stem cells in the postnatal and juvenile brain. Furthermore, because Lrig1 is a known regulator of quiescence, we also investigated the effect(s) of its deletion on the cellular proliferation in the lateral wall. METHODS To observe the development of the Lrig1-lineage stem cells, genetic inducible fate mapping studies in combination with thymidine analog administration were conducted using a previously published Lrig1T2A-iCreERT2 mouse line. To identify the long-term consequence(s) of Lrig1 germline deletion, old Lrig1 knock-out mice were generated using two different Lrig1 null alleles in the C57BL/6J background. The lateral walls from these mice were analyzed using an optimized whole mount immunofluorescence protocol and confocal microscopy. RESULTS We observed the Lrig1-lineage labeled cells with morphologies consistent with neurogenic stem cell identity in postnatal, juvenile, and adult mouse brains. Interestingly, when induced at postnatal or juvenile ages, morphologically distinct cells were revealed, including cells with the canonical B1 type stem cell morphology. Almost all of the presumptive stem cells labeled were non-proliferative at these ages. In the old Lrig1 germline knock-out mice, increased proliferation was observed compared to wildtype littermates without concomitant increase in apoptosis. CONCLUSIONS Once set aside during embryogenesis, the Lrig1-lineage stem cells remain largely quiescent during postnatal and juvenile development until activation in adult age. The absence of premature proliferative exhaustion in the Lrig1 knock-out stem cell niche during aging is likely due to a complex cascade of effects on the adult stem cell pool. Thus, we suggest that the adult stem cell pool size may be genetically constrained via Lrig1.
Collapse
Affiliation(s)
- Hyung-song Nam
- grid.223827.e0000 0001 2193 0096Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT 84112-5331 USA
| | - Mario R. Capecchi
- grid.223827.e0000 0001 2193 0096Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT 84112-5331 USA
| |
Collapse
|
15
|
Amberg N, Pauler FM, Streicher C, Hippenmeyer S. Tissue-wide genetic and cellular landscape shapes the execution of sequential PRC2 functions in neural stem cell lineage progression. SCIENCE ADVANCES 2022; 8:eabq1263. [PMID: 36322669 PMCID: PMC9629739 DOI: 10.1126/sciadv.abq1263] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 09/15/2022] [Indexed: 06/16/2023]
Abstract
The generation of a correctly sized cerebral cortex with all-embracing neuronal and glial cell-type diversity critically depends on faithful radial glial progenitor (RGP) cell proliferation/differentiation programs. Temporal RGP lineage progression is regulated by Polycomb repressive complex 2 (PRC2), and loss of PRC2 activity results in severe neurogenesis defects and microcephaly. How PRC2-dependent gene expression instructs RGP lineage progression is unknown. Here, we use mosaic analysis with double markers (MADM)-based single-cell technology and demonstrate that PRC2 is not cell-autonomously required in neurogenic RGPs but rather acts at the global tissue-wide level. Conversely, cortical astrocyte production and maturation is cell-autonomously controlled by PRC2-dependent transcriptional regulation. We thus reveal highly distinct and sequential PRC2 functions in RGP lineage progression that are dependent on complex interplays between intrinsic and tissue-wide properties. In a broader context, our results imply a critical role for the genetic and cellular niche environment in neural stem cell behavior.
Collapse
Affiliation(s)
| | - Florian M. Pauler
- Institute of Science and Technology Austria (ISTA), Am Campus 1, 3400 Klosterneuburg, Austria
| | - Carmen Streicher
- Institute of Science and Technology Austria (ISTA), Am Campus 1, 3400 Klosterneuburg, Austria
| | | |
Collapse
|
16
|
Ning S, Zhao J, Lombard AP, D’Abronzo LS, Leslie AR, Sharifi M, Lou W, Liu C, Yang JC, Evans CP, Corey E, Chen HW, Yu A, Ghosh PM, Gao AC. Activation of neural lineage networks and ARHGEF2 in enzalutamide-resistant and neuroendocrine prostate cancer and association with patient outcomes. COMMUNICATIONS MEDICINE 2022; 2:118. [PMID: 36159187 PMCID: PMC9492734 DOI: 10.1038/s43856-022-00182-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 09/05/2022] [Indexed: 01/26/2023] Open
Abstract
Background Treatment-emergent neuroendocrine prostate cancer (NEPC) after androgen receptor (AR) targeted therapies is an aggressive variant of prostate cancer with an unfavorable prognosis. The underlying mechanisms for early neuroendocrine differentiation are poorly defined and diagnostic and prognostic biomarkers are needed. Methods We performed transcriptomic analysis on the enzalutamide-resistant prostate cancer cell line C4-2B MDVR and NEPC patient databases to identify neural lineage signature (NLS) genes. Correlation of NLS genes with clinicopathologic features was determined. Cell viability was determined in C4-2B MDVR and H660 cells after knocking down ARHGEF2 using siRNA. Organoid viability of patient-derived xenografts was measured after knocking down ARHGEF2. Results We identify a 95-gene NLS representing the molecular landscape of neural precursor cell proliferation, embryonic stem cell pluripotency, and neural stem cell differentiation, which may indicate an early or intermediate stage of neuroendocrine differentiation. These NLS genes positively correlate with conventional neuroendocrine markers such as chromogranin and synaptophysin, and negatively correlate with AR and AR target genes in advanced prostate cancer. Differentially expressed NLS genes stratify small-cell NEPC from prostate adenocarcinoma, which are closely associated with clinicopathologic features such as Gleason Score and metastasis status. Higher ARGHEF2, LHX2, and EPHB2 levels among the 95 NLS genes correlate with a shortened survival time in NEPC patients. Furthermore, downregulation of ARHGEF2 gene expression suppresses cell viability and markers of neuroendocrine differentiation in enzalutamide-resistant and neuroendocrine cells. Conclusions The 95 neural lineage gene signatures capture an early molecular shift toward neuroendocrine differentiation, which could stratify advanced prostate cancer patients to optimize clinical treatment and serve as a source of potential therapeutic targets in advanced prostate cancer.
Collapse
Affiliation(s)
- Shu Ning
- grid.27860.3b0000 0004 1936 9684Department of Urologic Surgery, University of California Davis, Sacramento, CA USA
| | - Jinge Zhao
- grid.27860.3b0000 0004 1936 9684Department of Urologic Surgery, University of California Davis, Sacramento, CA USA ,grid.13291.380000 0001 0807 1581Present Address: Department of Urology, West China Hospital, Sichuan University, Sichuan, China
| | - Alan P. Lombard
- grid.27860.3b0000 0004 1936 9684Department of Urologic Surgery, University of California Davis, Sacramento, CA USA
| | - Leandro S. D’Abronzo
- grid.27860.3b0000 0004 1936 9684Department of Urologic Surgery, University of California Davis, Sacramento, CA USA
| | - Amy R. Leslie
- grid.27860.3b0000 0004 1936 9684Department of Urologic Surgery, University of California Davis, Sacramento, CA USA
| | - Masuda Sharifi
- grid.27860.3b0000 0004 1936 9684Department of Urologic Surgery, University of California Davis, Sacramento, CA USA
| | - Wei Lou
- grid.27860.3b0000 0004 1936 9684Department of Urologic Surgery, University of California Davis, Sacramento, CA USA
| | - Chengfei Liu
- grid.27860.3b0000 0004 1936 9684Department of Urologic Surgery, University of California Davis, Sacramento, CA USA ,grid.27860.3b0000 0004 1936 9684UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA USA
| | - Joy C. Yang
- grid.27860.3b0000 0004 1936 9684Department of Urologic Surgery, University of California Davis, Sacramento, CA USA
| | - Christopher P. Evans
- grid.27860.3b0000 0004 1936 9684Department of Urologic Surgery, University of California Davis, Sacramento, CA USA ,grid.27860.3b0000 0004 1936 9684UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA USA
| | - Eva Corey
- grid.34477.330000000122986657Department of Urology, University of Washington, Seattle, WA USA
| | - Hong-Wu Chen
- grid.27860.3b0000 0004 1936 9684UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA USA ,grid.27860.3b0000 0004 1936 9684Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA USA
| | - Aiming Yu
- grid.27860.3b0000 0004 1936 9684UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA USA ,grid.27860.3b0000 0004 1936 9684Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA USA
| | - Paramita M. Ghosh
- grid.27860.3b0000 0004 1936 9684Department of Urologic Surgery, University of California Davis, Sacramento, CA USA ,grid.27860.3b0000 0004 1936 9684UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA USA ,grid.27860.3b0000 0004 1936 9684Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA USA ,grid.413933.f0000 0004 0419 2847VA Northern California Health Care System, Sacramento, CA USA
| | - Allen C. Gao
- grid.27860.3b0000 0004 1936 9684Department of Urologic Surgery, University of California Davis, Sacramento, CA USA ,grid.27860.3b0000 0004 1936 9684UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA USA ,grid.413933.f0000 0004 0419 2847VA Northern California Health Care System, Sacramento, CA USA
| |
Collapse
|
17
|
Transit Amplifying Progenitors in the Cerebellum: Similarities to and Differences from Transit Amplifying Cells in Other Brain Regions and between Species. Cells 2022; 11:cells11040726. [PMID: 35203375 PMCID: PMC8870322 DOI: 10.3390/cells11040726] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 02/07/2022] [Accepted: 02/15/2022] [Indexed: 12/15/2022] Open
Abstract
Transit amplification of neural progenitors/precursors is widely used in the development of the central nervous system and for tissue homeostasis. In most cases, stem cells, which are relatively less proliferative, first differentiate into transit amplifying cells, which are more proliferative, losing their stemness. Subsequently, transit amplifying cells undergo a limited number of mitoses and differentiation to expand the progeny of differentiated cells. This step-by-step proliferation is considered an efficient system for increasing the number of differentiated cells while maintaining the stem cells. Recently, we reported that cerebellar granule cell progenitors also undergo transit amplification in mice. In this review, we summarize our and others’ recent findings and the prospective contribution of transit amplification to neural development and evolution, as well as the molecular mechanisms regulating transit amplification.
Collapse
|
18
|
Aghanoori MR, Burns KM, Subha M, Williams L, Hua M, Nobakht F, Krawec T, Yang G. Immunohistochemical analysis of the developing mouse cortex. Methods Cell Biol 2022; 170:31-46. [DOI: 10.1016/bs.mcb.2022.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
19
|
D-cysteine is an endogenous regulator of neural progenitor cell dynamics in the mammalian brain. Proc Natl Acad Sci U S A 2021; 118:2110610118. [PMID: 34556581 DOI: 10.1073/pnas.2110610118] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/19/2021] [Indexed: 12/19/2022] Open
Abstract
d-amino acids are increasingly recognized as important signaling molecules in the mammalian central nervous system. However, the d-stereoisomer of the amino acid with the fastest spontaneous racemization ratein vitro in vitro, cysteine, has not been examined in mammals. Using chiral high-performance liquid chromatography and a stereospecific luciferase assay, we identify endogenous d-cysteine in the mammalian brain. We identify serine racemase (SR), which generates the N-methyl-d-aspartate (NMDA) glutamate receptor coagonist d-serine, as a candidate biosynthetic enzyme for d-cysteine. d-cysteine is enriched more than 20-fold in the embryonic mouse brain compared with the adult brain. d-cysteine reduces the proliferation of cultured mouse embryonic neural progenitor cells (NPCs) by ∼50%, effects not shared with d-serine or l-cysteine. The antiproliferative effect of d-cysteine is mediated by the transcription factors FoxO1 and FoxO3a. The selective influence of d-cysteine on NPC proliferation is reflected in overgrowth and aberrant lamination of the cerebral cortex in neonatal SR knockout mice. Finally, we perform an unbiased screen for d-cysteine-binding proteins in NPCs by immunoprecipitation with a d-cysteine-specific antibody followed by mass spectrometry. This approach identifies myristoylated alanine-rich C-kinase substrate (MARCKS) as a putative d-cysteine-binding protein. Together, these results establish endogenous mammalian d-cysteine and implicate it as a physiologic regulator of NPC homeostasis in the developing brain.
Collapse
|
20
|
Different Flavors of Astrocytes: Revising the Origins of Astrocyte Diversity and Epigenetic Signatures to Understand Heterogeneity after Injury. Int J Mol Sci 2021; 22:ijms22136867. [PMID: 34206710 PMCID: PMC8268487 DOI: 10.3390/ijms22136867] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/31/2021] [Accepted: 06/02/2021] [Indexed: 12/11/2022] Open
Abstract
Astrocytes are a specific type of neuroglial cells that confer metabolic and structural support to neurons. Astrocytes populate all regions of the nervous system and adopt a variety of phenotypes depending on their location and their respective functions, which are also pleiotropic in nature. For example, astrocytes adapt to pathological conditions with a specific cellular response known as reactive astrogliosis, which includes extensive phenotypic and transcriptional changes. Reactive astrocytes may lose some of their homeostatic functions and gain protective or detrimental properties with great impact on damage propagation. Different astrocyte subpopulations seemingly coexist in reactive astrogliosis, however, the source of such heterogeneity is not completely understood. Altered cellular signaling in pathological compared to healthy conditions might be one source fueling astrocyte heterogeneity. Moreover, diversity might also be encoded cell-autonomously, for example as a result of astrocyte subtype specification during development. We hypothesize and propose here that elucidating the epigenetic signature underlying the phenotype of each astrocyte subtype is of high relevance to understand another regulative layer of astrocyte heterogeneity, in general as well as after injury or as a result of other pathological conditions. High resolution methods should allow enlightening diverse cell states and subtypes of astrocyte, their adaptation to pathological conditions and ultimately allow controlling and manipulating astrocyte functions in disease states. Here, we review novel literature reporting on astrocyte diversity from a developmental perspective and we focus on epigenetic signatures that might account for cell type specification.
Collapse
|
21
|
Shohayeb B, Muzar Z, Cooper HM. Conservation of neural progenitor identity and the emergence of neocortical neuronal diversity. Semin Cell Dev Biol 2021; 118:4-13. [PMID: 34083116 DOI: 10.1016/j.semcdb.2021.05.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/17/2021] [Accepted: 05/18/2021] [Indexed: 11/27/2022]
Abstract
One paramount challenge for neuroscientists over the past century has been to identify the embryonic origins of the enormous diversity of cortical neurons found in the adult human neocortex and to unravel the developmental processes governing their emergence. In all mammals, including humans, the radial glia lining the ventricles of the embryonic telencephalon, more recently reclassified as apical radial glia (aRGs), have been identified as the neural progenitors giving rise to all excitatory neurons and inhibitory interneurons of the six-layered cortex. In this review, we explore the fundamental molecular and cellular mechanisms that regulate aRG function and the generation of neuronal diversity in the dorsal telencephalon. We survey the key structural features essential for the retention of the highly polarized aRG morphology and therefore impose aRG identity after cytokinesis. We discuss how these structures and associated molecular signaling complexes influence aRG proliferative capacity and the decision to undergo proliferative self-renewing symmetric or neurogenic asymmetric divisions. We also explore the intriguing and complex question of how the extensive neuronal diversity within the adult neocortex arises from the small aRG population located within the cortical proliferative zone. We further highlight the recent clonal lineage tracing and single-cell transcriptomic profiling studies providing compelling evidence that individual neuronal identity emerges as a consequence of exposure to temporally regulated extrinsic cues which coordinate waves of transcriptional activity that evolve over time to drive neuronal commitment and maturation.
Collapse
Affiliation(s)
- Belal Shohayeb
- The University of Queensland, Queensland Brain Institute, Brisbane, Queensland 4072, Australia.
| | - Zukhrofi Muzar
- The University of Queensland, Queensland Brain Institute, Brisbane, Queensland 4072, Australia
| | - Helen M Cooper
- The University of Queensland, Queensland Brain Institute, Brisbane, Queensland 4072, Australia.
| |
Collapse
|
22
|
Shabangu T, Chen HL, Zhuang ZH, Pierani A, Chen CFF, Chou SJ. Specific contribution of neurons from the Dbx1 lineage to the piriform cortex. Sci Rep 2021; 11:8349. [PMID: 33863910 PMCID: PMC8052341 DOI: 10.1038/s41598-021-86512-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 03/15/2021] [Indexed: 11/20/2022] Open
Abstract
The piriform cortex (PC) is a major cortical processing center for the sense of smell that receives direct inputs from the olfactory bulb. In mice, the PC consists of three neuronal layers, which are populated by cells with distinct developmental origins. One origin of PC neurons is the pool of Dbx1-expressing neural progenitors located in the ventral pallium at the pallial-subpallial boundary. Since the precise mechanisms of PC neuron development are largely unknown, we sought to define the distribution, timing of neurogenesis, morphology and projection patterns of PC neurons from the Dbx1 lineage. We found that Dbx1-lineage neurons are preferentially distributed in layer 2 and enriched in the ventral portion of the PC. Further, Dbx1 neurons are early-born neurons and contribute to most neuronal subtypes in the PC. Our data also revealed an enrichment of Dbx1-lineage neurons in the ventral anterior PC that project to the orbitofrontal cortex. These findings suggest a specific association between the developmental origin of PC neurons and their neuronal properties.
Collapse
Affiliation(s)
- Thando Shabangu
- Molecular Cell Biology, Taiwan International Graduate Program, Academia Sinica, Taipei, Taiwan
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Hung-Lun Chen
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Zi-Hui Zhuang
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Alessandra Pierani
- Imagine Institute, Team Genetics and Development of the Cerebral Cortex, Université de Paris, F-75015, Paris, France
- Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, Université de Paris, F-75014, Paris, France
| | - Chien-Fu F Chen
- Molecular Cell Biology, Taiwan International Graduate Program, Academia Sinica, Taipei, Taiwan.
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan.
| | - Shen-Ju Chou
- Molecular Cell Biology, Taiwan International Graduate Program, Academia Sinica, Taipei, Taiwan.
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan.
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
23
|
The microcephaly gene Donson is essential for progenitors of cortical glutamatergic and GABAergic neurons. PLoS Genet 2021; 17:e1009441. [PMID: 33739968 PMCID: PMC8011756 DOI: 10.1371/journal.pgen.1009441] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 03/31/2021] [Accepted: 02/23/2021] [Indexed: 11/19/2022] Open
Abstract
Biallelic mutations in DONSON, an essential gene encoding for a replication fork protection factor, were linked to skeletal abnormalities and microcephaly. To better understand DONSON function in corticogenesis, we characterized Donson expression and consequences of conditional Donson deletion in the mouse telencephalon. Donson was widely expressed in the proliferation and differentiation zones of the embryonic dorsal and ventral telencephalon, which was followed by a postnatal expression decrease. Emx1-Cre-mediated Donson deletion in progenitors of cortical glutamatergic neurons caused extensive apoptosis in the early dorsomedial neuroepithelium, thus preventing formation of the neocortex and hippocampus. At the place of the missing lateral neocortex, these mutants exhibited a dorsal extension of an early-generated paleocortex. Targeting cortical neurons at the intermediate progenitor stage using Tbr2-Cre evoked no apparent malformations, whereas Nkx2.1-Cre-mediated Donson deletion in subpallial progenitors ablated 75% of Nkx2.1-derived cortical GABAergic neurons. Thus, the early telencephalic neuroepithelium depends critically on Donson function. Our findings help explain why the neocortex is most severely affected in individuals with DONSON mutations and suggest that DONSON-dependent microcephaly might be associated with so far unrecognized defects in cortical GABAergic neurons. Targeting Donson using an appropriate recombinase is proposed as a feasible strategy to ablate proliferating and nascent cells in experimental research. The cerebral cortex constitutes the largest part of the mammalian brain and is generated prenatally by highly proliferative progenitors. Genes encoding proteins that are essential for chromosomal segregation, mitotic division, DNA repair, and DNA damage response are frequently mutated in individuals diagnosed with microcephaly, a clinical condition characterized by cerebrocortical hypotrophy. Recent findings suggest that biallelic mutations in DONSON, a replication fork stabilization factor, cause microcephaly and skeletal defects, but this has not been formally tested. Here, we find that Cre-mediated Donson deletion in progenitors of cortical glutamatergic and cortical GABAergic neurons causes extensive programmed cell death at early stages of cortical development in mice. Cell death is induced in the proliferation zones and the postmitotic differentiation zones of the targeted progenitors. Mice undergoing Donson ablation in glutamatergic progenitors do not develop the hippocampus and dorsolateral neocortex, which leads to a dorsal shift of the early-generated piriform cortex. Donson deletion in GABAergic progenitors eliminates the vast majority of GABAergic neurons and oligodendrocyte precursors arising in the targeted lineage. We thus establish that Donson is essential for diverse early telencephalic progenitors. Targeting Donson might be used to kill off highly proliferating cells in experimental and probably therapeutic settings.
Collapse
|
24
|
Khuu MA, Nallamothu T, Castro-Rivera CI, Arias-Cavieres A, Szujewski CC, Garcia Iii AJ. Stage-dependent effects of intermittent hypoxia influence the outcome of hippocampal adult neurogenesis. Sci Rep 2021; 11:6005. [PMID: 33727588 PMCID: PMC7966401 DOI: 10.1038/s41598-021-85357-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 02/25/2021] [Indexed: 12/14/2022] Open
Abstract
Over one billion adults worldwide are estimated to suffer from sleep apnea, a condition with wide-reaching effects on brain health. Sleep apnea causes cognitive decline and is a risk factor for neurodegenerative conditions such as Alzheimer’s disease. Rodents exposed to intermittent hypoxia (IH), a hallmark of sleep apnea, exhibit spatial memory deficits associated with impaired hippocampal neurophysiology and dysregulated adult neurogenesis. We demonstrate that IH creates a pro-oxidant condition that reduces the Tbr2+ neural progenitor pool early in the process, while also suppressing terminal differentiation of adult born neurons during late adult neurogenesis. We further show that IH-dependent cell-autonomous hypoxia inducible factor 1-alpha (HIF1a) signaling is activated in early neuroprogenitors and enhances the generation of adult born neurons upon termination of IH. Our findings indicate that oscillations in oxygen homeostasis, such as those found in sleep apnea, have complex stage-dependent influence over hippocampal adult neurogenesis.
Collapse
Affiliation(s)
- Maggie A Khuu
- Institute for Integrative Physiology, Section of Emergency Medicine, The University of Chicago, 5841 S Maryland Ave, Chicago, IL, 60637, USA
| | - Thara Nallamothu
- Institute for Integrative Physiology, Section of Emergency Medicine, The University of Chicago, 5841 S Maryland Ave, Chicago, IL, 60637, USA
| | - Carolina I Castro-Rivera
- Institute for Integrative Physiology, Section of Emergency Medicine, The University of Chicago, 5841 S Maryland Ave, Chicago, IL, 60637, USA.,Committee On Neurobiology, The University of Chicago, Chicago, IL, 60307, USA.,Grossman Institute for Neuroscience, Quantitative Biology and Human Behavior, The University of Chicago, Chicago, IL, 60637, USA
| | - Alejandra Arias-Cavieres
- Institute for Integrative Physiology, Section of Emergency Medicine, The University of Chicago, 5841 S Maryland Ave, Chicago, IL, 60637, USA
| | - Caroline C Szujewski
- Institute for Integrative Physiology, Section of Emergency Medicine, The University of Chicago, 5841 S Maryland Ave, Chicago, IL, 60637, USA.,Committee On Neurobiology, The University of Chicago, Chicago, IL, 60307, USA.,Grossman Institute for Neuroscience, Quantitative Biology and Human Behavior, The University of Chicago, Chicago, IL, 60637, USA
| | - Alfredo J Garcia Iii
- Institute for Integrative Physiology, Section of Emergency Medicine, The University of Chicago, 5841 S Maryland Ave, Chicago, IL, 60637, USA. .,Committee On Neurobiology, The University of Chicago, Chicago, IL, 60307, USA. .,Grossman Institute for Neuroscience, Quantitative Biology and Human Behavior, The University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
25
|
Pearson CA, Moore DM, Tucker HO, Dekker JD, Hu H, Miquelajáuregui A, Novitch BG. Foxp1 Regulates Neural Stem Cell Self-Renewal and Bias Toward Deep Layer Cortical Fates. Cell Rep 2021; 30:1964-1981.e3. [PMID: 32049024 DOI: 10.1016/j.celrep.2020.01.034] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 12/20/2019] [Accepted: 01/08/2020] [Indexed: 02/06/2023] Open
Abstract
The laminar architecture of the mammalian neocortex depends on the orderly generation of distinct neuronal subtypes by apical radial glia (aRG) during embryogenesis. Here, we identify critical roles for the autism risk gene Foxp1 in maintaining aRG identity and gating the temporal competency for deep-layer neurogenesis. Early in development, aRG express high levels of Foxp1 mRNA and protein, which promote self-renewing cell divisions and deep-layer neuron production. Foxp1 levels subsequently decline during the transition to superficial-layer neurogenesis. Sustained Foxp1 expression impedes this transition, preserving a population of cells with aRG identity throughout development and extending the early neurogenic period into postnatal life. FOXP1 expression is further associated with the initial formation and expansion of basal RG (bRG) during human corticogenesis and can promote the formation of cells exhibiting characteristics of bRG when misexpressed in the mouse cortex. Together, these findings reveal broad functions for Foxp1 in cortical neurogenesis.
Collapse
Affiliation(s)
- Caroline Alayne Pearson
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; Intellectual and Developmental Disabilities Research Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Destaye M Moore
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; Intellectual and Developmental Disabilities Research Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Haley O Tucker
- Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Joseph D Dekker
- Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Hui Hu
- Department of Microbiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35205, USA
| | - Amaya Miquelajáuregui
- Institute of Neurobiology, University of Puerto Rico Medical Sciences Campus, San Juan, PR 00911, USA
| | - Bennett G Novitch
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; Intellectual and Developmental Disabilities Research Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA.
| |
Collapse
|
26
|
Shmakova AA, Balatskiy AV, Kulebyakina MA, Schaub T, Karagyaur MN, Kulebyakin KY, Rysenkova KD, Tarabykin VS, Tkachuk VA, Semina EV. Urokinase Receptor uPAR Overexpression in Mouse Brain Stimulates the Migration of Neurons into the Cortex during Embryogenesis. Russ J Dev Biol 2021. [DOI: 10.1134/s1062360421010069] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
27
|
Loss of Coiled-Coil Protein Cep55 Impairs Neural Stem Cell Abscission and Results in p53-Dependent Apoptosis in Developing Cortex. J Neurosci 2021; 41:3344-3365. [PMID: 33622776 DOI: 10.1523/jneurosci.1955-20.2021] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 12/24/2020] [Accepted: 02/13/2021] [Indexed: 12/23/2022] Open
Abstract
To build the brain, embryonic neural stem cells (NSCs) tightly regulate their cell divisions, undergoing a polarized form of cytokinesis that is poorly understood. Cytokinetic abscission is mediated by the midbody to sever the daughter cells at the apical membrane. In cell lines, the coiled-coil protein Cep55 was reported to be required for abscission. Mutations of Cep55 in humans cause a variety of cortical malformations. However, its role in the specialized divisions of NSCs is unclear. Here, we elucidate the roles of Cep55 in abscission and brain development. KO of Cep55 in mice causes abscission defects in neural and non-neural cell types, and postnatal lethality. The brain is disproportionately affected, with severe microcephaly at birth. Quantitative analyses of abscission in fixed and live cortical NSCs show that Cep55 acts to increase the speed and success rate of abscission, by facilitating ESCRT recruitment and timely microtubule disassembly. However, most NSCs complete abscission successfully in the absence of Cep55 Those that fail show a tissue-specific response: binucleate NSCs and neurons elevate p53, but binucleate fibroblasts do not. This leads to massive apoptosis in the brain, but not other tissues. Double KO of both p53 and Cep55 blocks apoptosis but only partially rescues Cep55 -/- brain size. This may be because of the persistent NSC cell division defects and p53-independent premature cell cycle exit. This work adds to emerging evidence that abscission regulation and error tolerance vary by cell type and are especially crucial in neural stem cells as they build the brain.SIGNIFICANCE STATEMENT During brain growth, embryonic neural stem cells (NSCs) must divide many times. In the last step of cell division, the daughter cell severs its connection to the mother stem cell, a process called abscission. The protein Cep55 is thought to be essential for recruiting proteins to the mother-daughter cell connection to complete abscission. We find that Cep55 mutants have very small brains with disturbed structure, but almost normal size bodies. NSC abscission can occur, but it is slower than normal, and failures are increased. Furthermore, NSCs that do fail abscission activate a signal for programmed cell death, whereas non-neural cells do not. Blocking this signal only partly restores brain growth, showing that regulation of abscission is crucial for brain development.
Collapse
|
28
|
Kim KT, Kwak YJ, Han SC, Hwang JH. Impairment of motor coordination and interneuron migration in perinatal exposure to glufosinate-ammonium. Sci Rep 2020; 10:20647. [PMID: 33244012 PMCID: PMC7691990 DOI: 10.1038/s41598-020-76869-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 08/03/2020] [Indexed: 11/09/2022] Open
Abstract
Glufosinate-ammonium (GLA) is a broad-spectrum herbicide for agricultural weed control and crop desiccation. Due to many GLA-resistant crops being developed to effectively control weeds and increase harvest yields, herbicide usage and the residual GLA in food has increased significantly. Though perinatal exposure by the residual GLA in food might affect brain development, the developmental neurotoxicity of GLA is still unclear. Therefore, this study aimed to investigate the effects of perinatal exposure to GLA on cortical development. The analysis revealed that perinatal GLA exposure altered behavioral changes in offspring, especially motor functional behavior. Moreover, perinatal GLA exposure affected cortical development, particularly by disrupting interneuron migration. These results provide new evidence that early life exposure to GLA alters cortical development.
Collapse
Affiliation(s)
- Kyung-Tai Kim
- Jeonbuk Branch Institute, Korea Institute of Toxicology, 30 Baekhak1-gil, Jeongeup, Jeollabuk-do, 56212, Republic of Korea
| | - Ye-Jung Kwak
- Jeonbuk Branch Institute, Korea Institute of Toxicology, 30 Baekhak1-gil, Jeongeup, Jeollabuk-do, 56212, Republic of Korea
| | - Su-Cheol Han
- Jeonbuk Branch Institute, Korea Institute of Toxicology, 30 Baekhak1-gil, Jeongeup, Jeollabuk-do, 56212, Republic of Korea.
| | - Jeong Ho Hwang
- Jeonbuk Branch Institute, Korea Institute of Toxicology, 30 Baekhak1-gil, Jeongeup, Jeollabuk-do, 56212, Republic of Korea.
| |
Collapse
|
29
|
Hoellerbauer P, Kufeld M, Arora S, Wu H, Feldman HM, Paddison PJ. A simple and highly efficient method for multi-allelic CRISPR-Cas9 editing in primary cell cultures. Cancer Rep (Hoboken) 2020; 3:e1269. [PMID: 32721120 PMCID: PMC7685144 DOI: 10.1002/cnr2.1269] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 06/24/2020] [Accepted: 06/25/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND CRISPR-Cas9-based technologies have revolutionized experimental manipulation of mammalian genomes. None-the-less, limitations of the delivery and efficacy of these technologies restrict their application in primary cells. AIMS To create an optimized protocol for penetrant, reproducible, and fast targeted genome editing in cell cultures derived from primary cells, using patient-derived glioblastoma stem-like cells (GSCs) and human neural stem/progenitor cells (NSCs) for proof-of-concept experiments. METHODS AND RESULTS We employed transient nucleofection of Cas9:sgRNA ribonucleoprotein complexes composed of chemically synthesized 2'-O-methyl 3'phosphorothioate-modified sgRNAs and purified Cas9 protein. Insertion-deletion mutation (indel) frequency and size distribution were measured via computational deconvolution of Sanger sequencing trace data. We found that this optimized technique routinely allows for >90% indel formation in only 3 days, without the need to create clonal lines for simple loss-of-function experiments. Using Western blotting, we observed near-total protein loss of target genes in cell pools. Additionally, we found that this approach allows for the creation of targeted genomic deletions. Furthermore, by using RNA-seq in edited NSCs to assess gene expression changes resulting from knockout of tumor suppressors commonly altered in glioblastoma, we also demonstrated the utility of this method for quickly creating a series of gene knockouts that allow for the study of oncogenic activities. CONCLUSION Our data suggest that this relatively simple method can be used for highly efficient and fast gene knockout, as well as for targeted genomic deletions, even in hyperdiploid cells (such as GSCs). This represents an extremely useful tool for the cancer research community when wishing to inactivate not only coding genes, but also non-coding RNAs, UTRs, enhancers, and promoters. This method can be readily applied to diverse cell types by varying the nucleofection conditions.
Collapse
Affiliation(s)
- Pia Hoellerbauer
- Human Biology DivisionFred Hutchinson Cancer Research CenterSeattleWashingtonUSA
- Molecular and Cellular Biology ProgramUniversity of WashingtonSeattleWashingtonUSA
| | - Megan Kufeld
- Human Biology DivisionFred Hutchinson Cancer Research CenterSeattleWashingtonUSA
- Antibody DiscoverySeattle GeneticsBothellWashingtonUSA
| | - Sonali Arora
- Human Biology DivisionFred Hutchinson Cancer Research CenterSeattleWashingtonUSA
| | - Hua‐Jun Wu
- Department of Biostatistics and Computational BiologyDana‐Farber Cancer Institute, and Department of Biostatistics, Harvard School of Public HealthBostonMassachusettsUSA
| | - Heather M. Feldman
- Human Biology DivisionFred Hutchinson Cancer Research CenterSeattleWashingtonUSA
| | - Patrick J. Paddison
- Human Biology DivisionFred Hutchinson Cancer Research CenterSeattleWashingtonUSA
- Molecular and Cellular Biology ProgramUniversity of WashingtonSeattleWashingtonUSA
| |
Collapse
|
30
|
Tran TN, Bader GD. Tempora: Cell trajectory inference using time-series single-cell RNA sequencing data. PLoS Comput Biol 2020; 16:e1008205. [PMID: 32903255 PMCID: PMC7505465 DOI: 10.1371/journal.pcbi.1008205] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 09/21/2020] [Accepted: 07/29/2020] [Indexed: 12/21/2022] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) can map cell types, states and transitions during dynamic biological processes such as tissue development and regeneration. Many trajectory inference methods have been developed to order cells by their progression through a dynamic process. However, when time series data is available, most of these methods do not consider the available time information when ordering cells and are instead designed to work only on a single scRNA-seq data snapshot. We present Tempora, a novel cell trajectory inference method that orders cells using time information from time-series scRNA-seq data. In performance comparison tests, Tempora inferred known developmental lineages from three diverse tissue development time series data sets, beating state of the art methods in accuracy and speed. Tempora works at the level of cell clusters (types) and uses biological pathway information to help identify cell type relationships. This approach increases gene expression signal from single cells, processing speed, and interpretability of the inferred trajectory. Our results demonstrate the utility of a combination of time and pathway information to supervise trajectory inference for scRNA-seq based analysis. Single-cell RNA sequencing (scRNA-seq) enables an unparalleled ability to map the heterogeneity of dynamic multicellular processes, such as tissue development, tumor growth, wound response and repair, and inflammation. Multiple methods have been developed to order cells along a pseudotime axis that represents a trajectory through such processes using the concept that cells that are closely related in a lineage will have similar transcriptomes. However, time series experiments provide another useful information source to order cells, from earlier to later time point. By introducing a novel use of biological pathway prior information, our Tempora algorithm improves the accuracy and speed of cell trajectory inference from time-series scRNA-seq data as measured by reconstructing known developmental trajectories from three diverse data sets. By analyzing scRNA-seq data at the cluster (cell type) level instead of at the single-cell level and by using known pathway information, Tempora amplifies gene expression signals from one cell using similar cells in a cluster and similar genes within a pathway. This approach also reduces computational time and resources needed to analyze large data sets because it works with a relatively small number of clusters instead of a potentially large number of cells. Finally, it eases interpretation, via operating on a relatively small number of clusters which usually represent known cell types, as well as by identifying time-dependent pathways. Tempora is useful for finding novel insights in dynamic processes.
Collapse
Affiliation(s)
- Thinh N. Tran
- Department of Molecular Genetics, University of Toronto, Ontario, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Ontario, Canada
| | - Gary D. Bader
- Department of Molecular Genetics, University of Toronto, Ontario, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Ontario, Canada
- Department of Computer Science, University of Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
31
|
Axonal Extensions along Corticospinal Tracts from Transplanted Human Cerebral Organoids. Stem Cell Reports 2020; 15:467-481. [PMID: 32679062 PMCID: PMC7419717 DOI: 10.1016/j.stemcr.2020.06.016] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 06/16/2020] [Accepted: 06/18/2020] [Indexed: 01/23/2023] Open
Abstract
The reconstruction of lost neural circuits by cell replacement is a possible treatment for neurological deficits after cerebral cortex injury. Cerebral organoids can be a novel source for cell transplantation, but because the cellular composition of the organoids changes along the time course of the development, it remains unclear which developmental stage of the organoids is most suitable for reconstructing the corticospinal tract. Here, we transplanted human embryonic stem cell-derived cerebral organoids at 6 or 10 weeks after differentiation (6w- or 10w-organoids) into mouse cerebral cortices. 6w-organoids extended more axons along the corticospinal tract but caused graft overgrowth with a higher percentage of proliferative cells. Axonal extensions from 10w-organoids were smaller in number but were enhanced when the organoids were grafted 1 week after brain injury. Finally, 10w-organoids extended axons in cynomolgus monkey brains. These results contribute to the development of a cell-replacement therapy for brain injury and stroke.
Collapse
|
32
|
Wang Y, Dai G, Gu Z, Liu G, Tang K, Pan YH, Chen Y, Lin X, Wu N, Chen H, Feng S, Qiu S, Sun H, Li Q, Xu C, Mao Y, Zhang YE, Khaitovich P, Wang YL, Liu Q, Han JDJ, Shao Z, Wei G, Xu C, Jing N, Li H. Accelerated evolution of an Lhx2 enhancer shapes mammalian social hierarchies. Cell Res 2020; 30:408-420. [PMID: 32238901 PMCID: PMC7196073 DOI: 10.1038/s41422-020-0308-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 03/12/2020] [Indexed: 12/26/2022] Open
Abstract
Social hierarchies emerged during evolution, and social rank influences behavior and health of individuals. However, the evolutionary mechanisms of social hierarchy are still unknown in amniotes. Here we developed a new method and performed a genome-wide screening for identifying regions with accelerated evolution in the ancestral lineage of placental mammals, where mammalian social hierarchies might have initially evolved. Then functional analyses were conducted for the most accelerated region designated as placental-accelerated sequence 1 (PAS1, P = 3.15 × 10-18). Multiple pieces of evidence show that PAS1 is an enhancer of the transcription factor gene Lhx2 involved in brain development. PAS1s isolated from various amniotes showed different cis-regulatory activity in vitro, and affected the expression of Lhx2 differently in the nervous system of mouse embryos. PAS1 knock-out mice lack social stratification. PAS1 knock-in mouse models demonstrate that PAS1s determine the social dominance and subordinate of adult mice, and that social ranks could even be turned over by mutated PAS1. All homozygous mutant mice had normal huddled sleeping behavior, motor coordination and strength. Therefore, PAS1-Lhx2 modulates social hierarchies and is essential for establishing social stratification in amniotes, and positive Darwinian selection on PAS1 plays pivotal roles in the occurrence of mammalian social hierarchies.
Collapse
Affiliation(s)
- Yuting Wang
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Yueyang Road 320, 200031, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Guangyi Dai
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Yueyang Road 320, 200031, Shanghai, China
| | - Zhili Gu
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Yueyang Road 320, 200031, Shanghai, China
| | - Guopeng Liu
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Yueyang Road 320, 200031, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ke Tang
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, Guangzhou, 510405, Guangdong, China
| | - Yi-Hsuan Pan
- Key Laboratory of Brain Functional Genomics of Ministry of Education, School of Life Science, East China Normal University, 200062, Shanghai, China
| | - Yujie Chen
- Bio-Med Big Data Center, CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Yueyang Road 320, 200031, Shanghai, China
| | - Xin Lin
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Yueyang Road 320, 200031, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Nan Wu
- Key Laboratory of Brain Functional Genomics of Ministry of Education, School of Life Science, East China Normal University, 200062, Shanghai, China
| | - Haoshan Chen
- University of Chinese Academy of Sciences, Beijing, China
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Yueyang Road 320, 200031, Shanghai, China
| | - Su Feng
- University of Chinese Academy of Sciences, Beijing, China
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Yueyang Road 320, 200031, Shanghai, China
| | - Shou Qiu
- University of Chinese Academy of Sciences, Beijing, China
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Yueyang Road 320, 200031, Shanghai, China
| | - Hongduo Sun
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Yueyang Road 320, 200031, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qian Li
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Yueyang Road 320, 200031, Shanghai, China
| | - Chuan Xu
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Yueyang Road 320, 200031, Shanghai, China
| | - Yanan Mao
- University of Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Zoological Systematics and Evolution & State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Yong Edward Zhang
- University of Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Zoological Systematics and Evolution & State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, 650223, Kunming, China
| | - Philipp Khaitovich
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Yueyang Road 320, 200031, Shanghai, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, 650223, Kunming, China
| | - Yan-Ling Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Qunxiu Liu
- Shanghai Zoological Park, 200335, Shanghai, China
| | - Jing-Dong Jackie Han
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Yueyang Road 320, 200031, Shanghai, China
| | - Zhen Shao
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Yueyang Road 320, 200031, Shanghai, China
| | - Gang Wei
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Yueyang Road 320, 200031, Shanghai, China
| | - Chun Xu
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Yueyang Road 320, 200031, Shanghai, China
| | - Naihe Jing
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Yueyang Road 320, 200031, Shanghai, China
| | - Haipeng Li
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Yueyang Road 320, 200031, Shanghai, China.
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, 650223, Kunming, China.
| |
Collapse
|
33
|
McNeely KC, Dwyer ND. Cytokinesis and postabscission midbody remnants are regulated during mammalian brain development. Proc Natl Acad Sci U S A 2020; 117:9584-9593. [PMID: 32273386 PMCID: PMC7197019 DOI: 10.1073/pnas.1919658117] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Building a brain of the proper size and structure requires neural stem cells (NSCs) to divide with tight temporal and spatial control to produce different daughter cell types in proper numbers and sequence. Mammalian NSCs in the embryonic cortex must maintain their polarized epithelial structure as they undergo both early proliferative divisions and later neurogenic divisions. To do this, they undergo a polarized form of cytokinesis at the apical membrane that is not well understood. Here, we investigate whether polarized furrowing and abscission in mouse NSCs are regulated differently at earlier and later stages and in a cytokinesis mutant, Kif20b This mutant was previously shown to have microcephaly and elevated apoptosis of NSCs. We developed methods to live image furrow ingression and midbody abscission in NSCs within cortical explants. We find that polarized furrow ingression occurs at a steady rate and completes in ∼15 min at two different ages. However, ingression is slower in a subset of Kif20b mutant NSCs. Abscission is usually observed on both sides of the midbody and takes 65 to 75 min to complete. Surprisingly, abscission is accelerated in the Kif20b mutant NSCs. Postabscission midbody remnants are observed at the apical membranes of daughter cells and are much more abundant in early-stage cortices. After NSC divisions in vitro, midbody remnants are more often retained on the daughter cells of early proliferative divisions. Altogether, these results suggest that regulation of abscission timing and midbody remnants in embryonic NSCs may influence proper brain growth and structure.
Collapse
Affiliation(s)
- Katrina C McNeely
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22908
- Neuroscience Graduate Program, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Noelle D Dwyer
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22908;
| |
Collapse
|
34
|
Miyazaki I, Kikuoka R, Isooka N, Takeshima M, Sonobe K, Arai R, Funakoshi H, Quin KE, Smart J, Zensho K, Asanuma M. Effects of maternal bisphenol A diglycidyl ether exposure during gestation and lactation on behavior and brain development of the offspring. Food Chem Toxicol 2020; 138:111235. [DOI: 10.1016/j.fct.2020.111235] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 02/17/2020] [Accepted: 02/27/2020] [Indexed: 10/24/2022]
|
35
|
Johnson CA, Ghashghaei HT. Sp2 regulates late neurogenic but not early expansive divisions of neural stem cells underlying population growth in the mouse cortex. Development 2020; 147:dev186056. [PMID: 32001437 PMCID: PMC7044455 DOI: 10.1242/dev.186056] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 01/23/2020] [Indexed: 12/18/2022]
Abstract
Cellular and molecular mechanisms underlying the switch from self-amplification of cortical stem cells to neuronal and glial generation are incompletely understood, despite their importance for neural development. Here, we have investigated the role of the transcription factor specificity protein 2 (Sp2) in expansive and neurogenic divisions of the developing cerebral cortex by combining conditional genetic deletion with the mosaic analysis with double markers (MADM) system in mice. We find that loss of Sp2 in progenitors undergoing neurogenic divisions results in prolonged mitosis due to extension of early mitotic stages. This disruption is correlated with depletion of the populations of upper layer neurons in the cortex. In contrast, early cortical neural stem cells proliferate and expand normally in the absence of Sp2. These results indicate a stage-specific requirement for Sp2 in neural stem and progenitor cells, and reveal mechanistic differences between the early expansive and later neurogenic periods of cortical development.This article has an associated 'The people behind the papers' interview.
Collapse
Affiliation(s)
- Caroline A Johnson
- Department of Molecular Biomedical Sciences College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27607, USA
| | - H Troy Ghashghaei
- Department of Molecular Biomedical Sciences College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27607, USA
| |
Collapse
|
36
|
Yamada T, Takechi M, Yokoyama N, Hiraoka Y, Ishikubo H, Usami T, Furutera T, Taga Y, Hirate Y, Kanai-Azuma M, Yoda T, Ogawa-Goto K, Iseki S. Heterozygous mutation of the splicing factor Sf3b4 affects development of the axial skeleton and forebrain in mouse. Dev Dyn 2020; 249:622-635. [PMID: 31900962 DOI: 10.1002/dvdy.148] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 12/22/2019] [Accepted: 12/26/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Splicing factor 3B subunit 4 (SF3B4) is a causative gene of an acrofacial dysostosis, Nager syndrome. Although in vitro analyses of SF3B4 have proposed multiple noncanonical functions unrelated to splicing, less information is available based on in vivo studies using model animals. RESULTS We performed expression and functional analyses of Sf3b4 in mice. The mouse Sf3b4 transcripts were found from two-cell stage, and were ubiquitously present during embryogenesis with high expression levels in several tissues such as forming craniofacial bones and brain. In contrast, expression of a pseudogene-like sequence of mouse Sf3b4 (Sf3b4_ps) found by in silico survey was not detected up to embryonic day 10. We generated a Sf3b4 knockout mouse using CRISPR-Cas9 system. The homozygous mutant mouse of Sf3b4 was embryonic lethal. The heterozygous mutant of Sf3b4 mouse (Sf3b4+/- ) exhibited smaller body size compared to the wild-type from postnatal to adult period, as well as homeotic posteriorization of the vertebral morphology and flattened calvaria. The flattened calvaria appears to be attributable to mild microcephaly due to a lower cell proliferation rate in the forebrain. CONCLUSIONS Our study suggests that Sf3b4 controls anterior-posterior patterning of the axial skeleton and guarantees cell proliferation for forebrain development in mice.
Collapse
Affiliation(s)
- Takahiko Yamada
- Section of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan.,Section of Maxillofacial Surgery, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Masaki Takechi
- Section of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Norisuke Yokoyama
- Section of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Yuichi Hiraoka
- Laboratory of Genome Editing for Biomedical Research, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan.,Laboratory of Molecular Neuroscience, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Harumi Ishikubo
- Laboratory of Genome Editing for Biomedical Research, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan.,Laboratory of Molecular Neuroscience, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Takako Usami
- Laboratory of Genome Editing for Biomedical Research, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan.,Laboratory of Molecular Neuroscience, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Toshiko Furutera
- Section of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Yuki Taga
- Nippi Research Institute of Biomatrix, Ibaraki, Japan
| | - Yoshikazu Hirate
- Department of Experimental Animal Model for Human Disease, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Masami Kanai-Azuma
- Department of Experimental Animal Model for Human Disease, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Tetsuya Yoda
- Section of Maxillofacial Surgery, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | | | - Sachiko Iseki
- Section of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| |
Collapse
|
37
|
Saito T. A Nucleolar Protein, Nepro, Is Essential for the Maintenance of Early Neural Stem Cells and Preimplantation Embryos. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1218:93-101. [PMID: 32060873 DOI: 10.1007/978-3-030-34436-8_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Notch signaling is required for maintaining neural stem cells (NSCs) in the developing brain. NSCs have potential to give rise to many neuronal types in the early telencephalon, and the potential decreases as embryonic development proceeds. Nepro, which encodes a unique nucleolar protein and is activated downstream of Notch, is essential for maintaining NSCs in the early telencephalon. Nepro is also expressed at basal levels and required for maintaining the preimplantation embryo, by repressing mitochondria-associated p53 apoptotic signaling. Notch signaling also controls dendritic complexity in mitral cells, major projection neurons in the olfactory bulb, showing that many steps of neural development involve Notch signaling.
Collapse
Affiliation(s)
- Tetsuichiro Saito
- Department of Developmental Biology, Graduate School of Medicine, Chiba University, Chiba, Japan.
| |
Collapse
|
38
|
Edens BM, Vissers C, Su J, Arumugam S, Xu Z, Shi H, Miller N, Rojas Ringeling F, Ming GL, He C, Song H, Ma YC. FMRP Modulates Neural Differentiation through m 6A-Dependent mRNA Nuclear Export. Cell Rep 2019; 28:845-854.e5. [PMID: 31340148 PMCID: PMC6687293 DOI: 10.1016/j.celrep.2019.06.072] [Citation(s) in RCA: 203] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 04/09/2019] [Accepted: 06/20/2019] [Indexed: 12/21/2022] Open
Abstract
N6-methyladenosine (m6A) modification of mRNA is emerging as a vital mechanism regulating RNA function. Here, we show that fragile X mental retardation protein (FMRP) reads m6A to promote nuclear export of methylated mRNA targets during neural differentiation. Fmr1 knockout (KO) mice show delayed neural progenitor cell cycle progression and extended maintenance of proliferating neural progenitors into postnatal stages, phenocopying methyltransferase Mettl14 conditional KO (cKO) mice that have no m6A modification. RNA-seq and m6A-seq reveal that both Mettl14cKO and Fmr1KO lead to the nuclear retention of m6A-modified FMRP target mRNAs regulating neural differentiation, indicating that both m6A and FMRP are required for the nuclear export of methylated target mRNAs. FMRP preferentially binds m6A-modified RNAs to facilitate their nuclear export through CRM1. The nuclear retention defect can be mitigated by wild-type but not nuclear export-deficient FMRP, establishing a critical role for FMRP in mediating m6A-dependent mRNA nuclear export during neural differentiation.
Collapse
Affiliation(s)
- Brittany M Edens
- Departments of Pediatrics, Neurology, and Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Caroline Vissers
- Biochemistry, Molecular and Cellular Biology Training Program, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jing Su
- Departments of Pediatrics, Neurology, and Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Saravanan Arumugam
- Departments of Pediatrics, Neurology, and Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Zhaofa Xu
- Departments of Pediatrics, Neurology, and Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Han Shi
- Departments of Pediatrics, Neurology, and Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Nimrod Miller
- Departments of Pediatrics, Neurology, and Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | | | - Guo-Li Ming
- Department of Neuroscience and Mahoney Institute for Neuroscience, Department of Cell and Developmental Biology, Institute for Regeneration, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Chuan He
- Departments of Chemistry and Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Hongjun Song
- Department of Neuroscience and Mahoney Institute for Neuroscience, Department of Cell and Developmental Biology, Institute for Regeneration, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Yongchao C Ma
- Departments of Pediatrics, Neurology, and Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA.
| |
Collapse
|
39
|
Little JN, Dwyer ND. p53 deletion rescues lethal microcephaly in a mouse model with neural stem cell abscission defects. Hum Mol Genet 2019; 28:434-447. [PMID: 30304535 DOI: 10.1093/hmg/ddy350] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 09/28/2018] [Indexed: 12/17/2022] Open
Abstract
Building a cerebral cortex of the proper size involves balancing rates and timing of neural stem cell (NSC) proliferation, neurogenesis and cell death. The cellular mechanisms connecting genetic mutations to brain malformation phenotypes are still poorly understood. Microcephaly may result when NSC divisions are too slow, produce neurons too early or undergo apoptosis but the relative contributions of these cellular mechanisms to various types of microcephaly are not understood. We previously showed that mouse mutants in Kif20b (formerly called Mphosph1, Mpp1 or KRMP1) have small cortices that show elevated apoptosis and defects in maturation of NSC midbodies, which mediate cytokinetic abscission. Here we test the contribution of intrinsic NSC apoptosis to brain size reduction in this lethal microcephaly model. By making double mutants with the pro-apoptotic genes Bax and Trp53 (p53), we find that p53-dependent apoptosis of cortical NSCs accounts for most of the microcephaly, but that there is a significant apoptosis-independent contribution as well. Remarkably, heterozygous p53 deletion is sufficient to fully rescue survival of the Kif20b mutant into adulthood. In addition, the NSC midbody maturation defects are not rescued by p53 deletion, showing that they are either upstream of p53 activation, or in a parallel pathway. Accumulation of p53 in the nucleus of mutant NSCs at midbody stage suggests the possibility of a novel midbody-mediated pathway for p53 activation. This work elucidates both NSC apoptosis and abscission mechanisms that could underlie human microcephaly or other brain malformations.
Collapse
Affiliation(s)
- Jessica Neville Little
- Department of Cell Biology.,Cell and Developmental Biology Graduate Program.,Medical Scientist Training Program, University of Virginia School of Medicine, Charlottesville, VA, USA
| | | |
Collapse
|
40
|
Wu HN, Cao XL, Fang Z, Zhang YF, Han WJ, Yue KY, Cao Y, Zheng MH, Wang LL, Han H. Deficiency of Ttyh1 downstream to Notch signaling results in precocious differentiation of neural stem cells. Biochem Biophys Res Commun 2019; 514:842-847. [PMID: 31079925 DOI: 10.1016/j.bbrc.2019.04.181] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 04/27/2019] [Indexed: 12/18/2022]
Abstract
Mammalian neural stem cells (NSCs) are not only responsible for normal development of the central nervous system (CNS), but also participate in brain homeostasis and repair, thus hold promising clinical potentials in the treatment of neurodegenerative diseases and trauma. However the molecular networks regulating the stemness and differentiation of NSCs have not been fully understood. In this study, we show that Tweety-homolog 1 (Ttyh1), a five-pass transmembrane protein specifically expressed in mouse brain, is involved in maintaining stemness of murine NSCs. Blocking or activating Notch signal led to downregulation and upregulation of Ttyh1 in cultured NSCs, respectively, suggesting that Ttyh1 is under the control of Notch signaling. Knockdown of Ttyh1 in cultured NSCs resulted in a transient increase in the number and size of neurospheres, followed by a decrease of stemness as manifested by compromised neurosphere formation, downregulated stem cell markers, and increased neuronal differentiation. We generated Ttyh1 knockout mice by deleting its exon 4 using the CRISPR-Cas9 technology. Surprisingly, in contrast to a previous report, Ttyh1 knockout did not result in embryonic lethality. NSCs derived from Ttyh1 knockout mice phenocopied NSCs transfected with Ttyh1 siRNA. Immunofluorescence showed that loss of Ttyh1 leads to the increase of neurogenesis in adult mice. Taken together, these findings indicate that Ttyh1, which is likely downstream to Notch signaling, plays an important role in regulating NSCs.
Collapse
Affiliation(s)
- Hai-Ning Wu
- State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, 710032, China; Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Xiu-Li Cao
- State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, 710032, China; Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Zheng Fang
- State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, 710032, China; Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Yu-Fei Zhang
- State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, 710032, China; Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Wen-Juan Han
- State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, 710032, China; Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Kang-Yi Yue
- State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, 710032, China; Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Yuan Cao
- State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, 710032, China; Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Min-Hua Zheng
- State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, 710032, China; Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, 710032, China.
| | - Li-Li Wang
- Key Laboratory of Synthetic and Natural Functional Molecular Chemistry of Ministry of Education, Institute of Modern Separation Science, Northwest University, Shaanxi Key Laboratory of Modern Separation Science, Xi'an, 710069, China
| | - Hua Han
- State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, 710032, China; Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
41
|
O'Neill RS, Schoborg TA, Rusan NM. Same but different: pleiotropy in centrosome-related microcephaly. Mol Biol Cell 2019; 29:241-246. [PMID: 29382806 PMCID: PMC5996963 DOI: 10.1091/mbc.e17-03-0192] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 11/22/2017] [Accepted: 11/30/2017] [Indexed: 01/19/2023] Open
Abstract
An intimate link between centrosome function and neurogenesis is revealed by the identification of many genes with centrosome-associated functions that are mutated in microcephaly disorders. Consistent with the major role of the centrosome in mitosis, mutations in these centrosome-related microcephaly (CRM) genes are thought to affect neurogenesis by depleting the pool of neural progenitor cells, primarily through apoptosis as a consequence of mitotic failure or premature differentiation as a consequence of cell cycle delay and randomization of spindle orientation. However, as suggested by the wide range of microcephaly phenotypes and the multifunctional nature of many CRM proteins, this picture of CRM gene function is incomplete. Here, we explore several examples of CRM genes pointing to additional functions that contribute to microcephaly, including regulation of cell cycle signaling, actin cytoskeleton, and Hippo pathway proteins, as well as functions in postmitotic neurons and glia. As these examples are likely just the tip of the iceberg, further exploration of the roles of microcephaly-related genes are certain to reveal additional unforeseen functions important for neurodevelopment.
Collapse
Affiliation(s)
- Ryan S O'Neill
- Cell Biology and Physiology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Todd A Schoborg
- Cell Biology and Physiology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Nasser M Rusan
- Cell Biology and Physiology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
42
|
Kalogeropoulou A, Lygerou Z, Taraviras S. Cortical Development and Brain Malformations: Insights From the Differential Regulation of Early Events of DNA Replication. Front Cell Dev Biol 2019; 7:29. [PMID: 30915332 PMCID: PMC6421272 DOI: 10.3389/fcell.2019.00029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 02/20/2019] [Indexed: 12/27/2022] Open
Abstract
During the development of the cortex distinct populations of Neural Stem Cells (NSCs) are defined by differences in their cell cycle duration, self-renewal capacity and transcriptional profile. A key difference across the distinct populations of NSCs is the length of G1 phase, where the licensing of the DNA replication origins takes place by the assembly of a pre-replicative complex. Licensing of DNA replication is a process that is adapted accordingly to the cell cycle length of NSCs to secure the timed duplication of the genome. Moreover, DNA replication should be efficiently coordinated with ongoing transcription for the prevention of conflicts that would impede the progression of both processes, compromising the normal course of development. In the present review we discuss how the differential regulation of the licensing and initiation of DNA replication in different cortical NSCs populations is integrated with the properties of these stem cells populations. Moreover, we examine the implication of the initial steps of DNA replication in the pathogenetic mechanisms of neurodevelopmental defects and Zika virus-related microcephaly, highlighting the significance of the differential regulation of DNA replication during brain development.
Collapse
Affiliation(s)
| | - Zoi Lygerou
- Department of General Biology, Medical School, University of Patras, Patras, Greece
| | - Stavros Taraviras
- Department of Physiology, Medical School, University of Patras, Patras, Greece
| |
Collapse
|
43
|
Aubid NN, Liu Y, Vidal JMP, Hall VJ. Isolation and culture of porcine primary fetal progenitors and neurons from the developing dorsal telencephalon. J Vet Sci 2019; 20:e3. [PMID: 30944526 PMCID: PMC6441812 DOI: 10.4142/jvs.2019.20.e3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 11/26/2018] [Accepted: 12/04/2018] [Indexed: 01/20/2023] Open
Abstract
The development of long-term surviving fetal cell cultures from primary cell tissue from the developing brain is important for facilitating studies investigating neural development and for modelling neural disorders and brain congenital defects. The field faces current challenges in co-culturing both progenitors and neurons long-term. Here, we culture for the first time, porcine fetal cells from the dorsal telencephalon at embryonic day (E) 50 and E60 in conditions that promoted both the survival of progenitor cells and young neurons. We applied a novel protocol designed to collect, isolate and promote survival of both progenitors and young neurons. Herein, we used a combination of low amount of fetal bovine serum, together with pro-survival factors, including basic fibroblast growth factor and retinoic acid, together with arabinofuranosylcytosine and could maintain progenitors and facilitate in vitro differentiation into calbindin 1+ neurons and reelin+ interneurons for a period of 7 days. Further improvements to the protocol that might extend the survival of the fetal primary neural cells would be beneficial. The development of new porcine fetal culture methods is of value for the field, given the pig's neuroanatomical and developmental similarities to the human brain.
Collapse
Affiliation(s)
- Niroch Nawzad Aubid
- Department of Veterinary and Animal Sciences, Faculty of Health Sciences, University of Copenhagen, Frederiksberg C, DK-1870, Denmark
| | - Yong Liu
- Department of Veterinary and Animal Sciences, Faculty of Health Sciences, University of Copenhagen, Frederiksberg C, DK-1870, Denmark
| | - Juan Miguel Peralvo Vidal
- Department of Veterinary and Animal Sciences, Faculty of Health Sciences, University of Copenhagen, Frederiksberg C, DK-1870, Denmark
| | - Vanessa Jane Hall
- Department of Veterinary and Animal Sciences, Faculty of Health Sciences, University of Copenhagen, Frederiksberg C, DK-1870, Denmark
| |
Collapse
|
44
|
Saili KS, Zurlinden TJ, Schwab AJ, Silvin A, Baker NC, Hunter ES, Ginhoux F, Knudsen TB. Blood-brain barrier development: Systems modeling and predictive toxicology. Birth Defects Res 2018; 109:1680-1710. [PMID: 29251840 DOI: 10.1002/bdr2.1180] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 11/12/2017] [Indexed: 01/17/2023]
Abstract
The blood-brain barrier (BBB) serves as a gateway for passage of drugs, chemicals, nutrients, metabolites, and hormones between vascular and neural compartments in the brain. Here, we review BBB development with regard to the microphysiology of the neurovascular unit (NVU) and the impact of BBB disruption on brain development. Our focus is on modeling these complex systems. Extant in silico models are available as tools to predict the probability of drug/chemical passage across the BBB; in vitro platforms for high-throughput screening and high-content imaging provide novel data streams for profiling chemical-biological interactions; and engineered human cell-based microphysiological systems provide empirical models with which to investigate the dynamics of NVU function. Computational models are needed that bring together kinetic and dynamic aspects of NVU function across gestation and under various physiological and toxicological scenarios. This integration will inform adverse outcome pathways to reduce uncertainty in translating in vitro data and in silico models for use in risk assessments that aim to protect neurodevelopmental health.
Collapse
Affiliation(s)
- Katerine S Saili
- National Center for Computational Toxicology (NCCT); U.S. Environmental Protection Agency, Office of Research and Development, Research Triangle Park, North Carolina 27711
| | - Todd J Zurlinden
- National Center for Computational Toxicology (NCCT); U.S. Environmental Protection Agency, Office of Research and Development, Research Triangle Park, North Carolina 27711
| | - Andrew J Schwab
- National Health and Environmental Effects Research Laboratory (NHEERL), U.S. Environmental Protection Agency, Office of Research and Development, Research Triangle Park, North Carolina 27711
| | - Aymeric Silvin
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), 138648, Singapore
| | - Nancy C Baker
- Leidos, contractor to NCCT, Research Triangle Park, North Carolina 27711
| | - E Sidney Hunter
- National Health and Environmental Effects Research Laboratory (NHEERL), U.S. Environmental Protection Agency, Office of Research and Development, Research Triangle Park, North Carolina 27711
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), 138648, Singapore
| | - Thomas B Knudsen
- National Center for Computational Toxicology (NCCT); U.S. Environmental Protection Agency, Office of Research and Development, Research Triangle Park, North Carolina 27711
| |
Collapse
|
45
|
Wilson C, Muñoz-Palma E, González-Billault C. From birth to death: A role for reactive oxygen species in neuronal development. Semin Cell Dev Biol 2018; 80:43-49. [DOI: 10.1016/j.semcdb.2017.09.012] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 09/07/2017] [Accepted: 09/08/2017] [Indexed: 02/07/2023]
|
46
|
Nganou G, Silva CG, Gladwyn-Ng I, Engel D, Coumans B, Delgado-Escueta AV, Tanaka M, Nguyen L, Grisar T, de Nijs L, Lakaye B. Importin-8 Modulates Division of Apical Progenitors, Dendritogenesis and Tangential Migration During Development of Mouse Cortex. Front Mol Neurosci 2018; 11:234. [PMID: 30042658 PMCID: PMC6048241 DOI: 10.3389/fnmol.2018.00234] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 06/13/2018] [Indexed: 01/18/2023] Open
Abstract
The building of the brain is a multistep process that requires the coordinate expression of thousands of genes and an intense nucleocytoplasmic transport of RNA and proteins. This transport is mediated by karyopherins that comprise importins and exportins. Here, we investigated the role of the ß-importin, importin-8 (IPO8) during mouse cerebral corticogenesis as several of its cargoes have been shown to be essential during this process. First, we showed that Ipo8 mRNA is expressed in mouse brain at various embryonic ages with a clear signal in the sub-ventricular/ventricular zone (SVZ/VZ), the cerebral cortical plate (CP) and the ganglionic eminences. We found that acute knockdown of IPO8 in cortical progenitors reduced both their proliferation and cell cycle exit leading to the increase in apical progenitor pool without influencing the number of basal progenitors (BPs). Projection neurons ultimately reached their appropriate cerebral cortical layer, but their dendritogenesis was specifically affected, resulting in neurons with reduced dendrite complexity. IPO8 knockdown also slowed the migration of cortical interneurons. Together, our data demonstrate that IPO8 contribute to the coordination of several critical steps of cerebral cortex development. These results suggest that the impairment of IPO8 function might be associated with some diseases of neuronal migration defects.
Collapse
Affiliation(s)
- Gerry Nganou
- GIGA-Neurosciences, University of Liege, Liege, Belgium.,GENESS International Consortium, Los Angeles, CA, United States
| | - Carla G Silva
- GIGA-Neurosciences, University of Liege, Liege, Belgium
| | | | | | - Bernard Coumans
- GIGA-Neurosciences, University of Liege, Liege, Belgium.,GENESS International Consortium, Los Angeles, CA, United States
| | - Antonio V Delgado-Escueta
- GENESS International Consortium, Los Angeles, CA, United States.,Epilepsy Genetics/Genomics Lab, Neurology and Research Services, VA Greater Los Angeles Healthcare System (VA GLAHS), University of California, Los Angeles, Los Angeles, CA, United States
| | - Miyabi Tanaka
- GENESS International Consortium, Los Angeles, CA, United States.,Epilepsy Genetics/Genomics Lab, Neurology and Research Services, VA Greater Los Angeles Healthcare System (VA GLAHS), University of California, Los Angeles, Los Angeles, CA, United States
| | | | - Thierry Grisar
- GIGA-Neurosciences, University of Liege, Liege, Belgium.,GENESS International Consortium, Los Angeles, CA, United States
| | - Laurence de Nijs
- GENESS International Consortium, Los Angeles, CA, United States.,MHeNS, Maastricht University, Maastricht, Netherlands
| | - Bernard Lakaye
- GIGA-Neurosciences, University of Liege, Liege, Belgium.,GENESS International Consortium, Los Angeles, CA, United States
| |
Collapse
|
47
|
Uzquiano A, Gladwyn-Ng I, Nguyen L, Reiner O, Götz M, Matsuzaki F, Francis F. Cortical progenitor biology: key features mediating proliferation versus differentiation. J Neurochem 2018; 146:500-525. [PMID: 29570795 DOI: 10.1111/jnc.14338] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 02/26/2018] [Accepted: 03/08/2018] [Indexed: 12/18/2022]
Abstract
The cerebral cortex is a highly organized structure whose development depends on diverse progenitor cell types, namely apical radial glia, intermediate progenitors, and basal radial glia cells, which are responsible for the production of the correct neuronal output. In recent years, these progenitor cell types have been deeply studied, particularly basal radial glia and their role in cortical expansion and gyrification. We review here a broad series of factors that regulate progenitor behavior and daughter cell fate. We first describe the different neuronal progenitor types, emphasizing the differences between lissencephalic and gyrencephalic species. We then review key factors shown to influence progenitor proliferation versus differentiation, discussing their roles in progenitor dynamics, neuronal production, and potentially brain size and complexity. Although spindle orientation has been considered a critical factor for mode of division and daughter cell output, we discuss other features that are emerging as crucial for these processes such as organelle and cell cycle dynamics. Additionally, we highlight the importance of adhesion molecules and the polarity complex for correct cortical development. Finally, we briefly discuss studies assessing progenitor multipotency and its possible contribution to the production of specific neuronal populations. This review hence summarizes recent aspects of cortical progenitor cell biology, and pinpoints emerging features critical for their behavior.
Collapse
Affiliation(s)
- Ana Uzquiano
- INSERM, UMR-S 839, Paris, France.,Sorbonne Université, Université Pierre et Marie Curie, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Ivan Gladwyn-Ng
- GIGA-Neurosciences, Interdisciplinary Cluster for Applied Genoproteomics (GIGA-R), University of Liège, C.H.U. Sart Tilman, Liège, Belgium
| | - Laurent Nguyen
- GIGA-Neurosciences, Interdisciplinary Cluster for Applied Genoproteomics (GIGA-R), University of Liège, C.H.U. Sart Tilman, Liège, Belgium
| | - Orly Reiner
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Magdalena Götz
- Physiological Genomics, Biomedical Center, Ludwig Maximilians University Munich, Planegg/Munich, Germany.,Institute for Stem Cell Research, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany.,SYNERGY, Excellence Cluster of Systems Neurology, Biomedical Center, Ludwig-Maximilian University Munich, Planegg/Munich, Germany
| | - Fumio Matsuzaki
- Laboratory for Cell Asymmetry, Center for Developmental Biology, RIKEN Kobe Institute, Kobe, Hyogo, Japan
| | - Fiona Francis
- INSERM, UMR-S 839, Paris, France.,Sorbonne Université, Université Pierre et Marie Curie, Paris, France.,Institut du Fer à Moulin, Paris, France
| |
Collapse
|
48
|
Adams KV, Morshead CM. Neural stem cell heterogeneity in the mammalian forebrain. Prog Neurobiol 2018; 170:2-36. [PMID: 29902499 DOI: 10.1016/j.pneurobio.2018.06.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 05/23/2018] [Accepted: 06/07/2018] [Indexed: 12/21/2022]
Abstract
The brain was long considered an organ that underwent very little change after development. It is now well established that the mammalian central nervous system contains neural stem cells that generate progeny that are capable of making new neurons, astrocytes, and oligodendrocytes throughout life. The field has advanced rapidly as it strives to understand the basic biology of these precursor cells, and explore their potential to promote brain repair. The purpose of this review is to present current knowledge about the diversity of neural stem cells in vitro and in vivo, and highlight distinctions between neural stem cell populations, throughout development, and within the niche. A comprehensive understanding of neural stem cell heterogeneity will provide insights into the cellular and molecular regulation of neural development and lifelong neurogenesis, and will guide the development of novel strategies to promote regeneration and neural repair.
Collapse
Affiliation(s)
- Kelsey V Adams
- Institute of Medical Science, Terrence Donnelly Centre, University of Toronto, Toronto ON, M5S 3E2, Canada.
| | - Cindi M Morshead
- Institute of Medical Science, Terrence Donnelly Centre, University of Toronto, Toronto ON, M5S 3E2, Canada; Department of Surgery, Division of Anatomy, Canada; Institute of Biomaterials and Biomedical Engineering, Canada; Rehabilitation Science Institute, University of Toronto, Canada.
| |
Collapse
|
49
|
Kowiański P, Lietzau G, Czuba E, Waśkow M, Steliga A, Moryś J. BDNF: A Key Factor with Multipotent Impact on Brain Signaling and Synaptic Plasticity. Cell Mol Neurobiol 2018; 38:579-593. [PMID: 28623429 PMCID: PMC5835061 DOI: 10.1007/s10571-017-0510-4] [Citation(s) in RCA: 866] [Impact Index Per Article: 123.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 06/08/2017] [Indexed: 12/15/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is one of the most widely distributed and extensively studied neurotrophins in the mammalian brain. Among its prominent functions, one can mention control of neuronal and glial development, neuroprotection, and modulation of both short- and long-lasting synaptic interactions, which are critical for cognition and memory. A wide spectrum of processes are controlled by BDNF, and the sometimes contradictory effects of its action can be explained based on its specific pattern of synthesis, comprising several intermediate biologically active isoforms that bind to different types of receptor, triggering several signaling pathways. The functions of BDNF must be discussed in close relation to the stage of brain development, the different cellular components of nervous tissue, as well as the molecular mechanisms of signal transduction activated under physiological and pathological conditions. In this review, we briefly summarize the current state of knowledge regarding the impact of BDNF on regulation of neurophysiological processes. The importance of BDNF for future studies aimed at disclosing mechanisms of activation of signaling pathways, neuro- and gliogenesis, as well as synaptic plasticity is highlighted.
Collapse
Affiliation(s)
- Przemysław Kowiański
- Department of Anatomy and Neurobiology, Medical University of Gdansk, 1 Debinki Street, 80-211, Gdańsk, Poland.
- Department of Health Sciences, Pomeranian University of Slupsk, 64 Bohaterów Westerplatte Str., 76-200, Słupsk, Poland.
| | - Grażyna Lietzau
- Department of Anatomy and Neurobiology, Medical University of Gdansk, 1 Debinki Street, 80-211, Gdańsk, Poland
| | - Ewelina Czuba
- Department of Anatomy and Neurobiology, Medical University of Gdansk, 1 Debinki Street, 80-211, Gdańsk, Poland
| | - Monika Waśkow
- Department of Health Sciences, Pomeranian University of Slupsk, 64 Bohaterów Westerplatte Str., 76-200, Słupsk, Poland
| | - Aleksandra Steliga
- Department of Health Sciences, Pomeranian University of Slupsk, 64 Bohaterów Westerplatte Str., 76-200, Słupsk, Poland
| | - Janusz Moryś
- Department of Anatomy and Neurobiology, Medical University of Gdansk, 1 Debinki Street, 80-211, Gdańsk, Poland
| |
Collapse
|
50
|
Abstract
A hundred years after Lhx2 ortholog apterous was identified as a critical regulator of wing development in Drosophila, LIM-HD gene family members have proved to be versatile and powerful components of the molecular machinery that executes the blueprint of embryogenesis across vertebrate and invertebrate species. Here, we focus on the spatio-temporally varied functions of LIM-homeodomain transcription factor LHX2 in the developing mouse forebrain. Right from its earliest known role in telencephalic and eye field patterning, to the control of the neuron-glia cell fate switch, and the regulation of axon pathfinding and dendritic arborization in late embryonic stages, LHX2 has been identified as a fundamental, temporally dynamic, always necessary, and often sufficient factor in a range of critical developmental phenomena. While Lhx2 mutant phenotypes have been characterized in detail in multiple brain structures, only recently have we advanced in our understanding of the molecular mechanisms by which this factor acts. Common themes emerge from how this multifunctional molecule controls a range of developmental steps in distinct forebrain structures. Examining these shared features, and noting unique aspects of LHX2 function is likely to inform our understanding of how a single factor can bring about a diversity of effects and play central and critical roles across systems and stages. The parallels in LHX2 and APTEROUS functions, and the protein complexes they participate in, offer insights into evolutionary strategies that conserve tool kits and deploy them to play new, yet familiar roles in species separated by hundreds of millions of years.
Collapse
Affiliation(s)
- Shen-Ju Chou
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Shubha Tole
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India.
| |
Collapse
|