1
|
Gaweda-Walerych K, Aragona V, Lodato S, Sitek EJ, Narożańska E, Buratti E. Progranulin deficiency in the brain: the interplay between neuronal and non-neuronal cells. Transl Neurodegener 2025; 14:18. [PMID: 40234992 PMCID: PMC12001433 DOI: 10.1186/s40035-025-00475-8] [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: 09/20/2024] [Accepted: 02/21/2025] [Indexed: 04/17/2025] Open
Abstract
Heterozygous mutations in GRN gene lead to insufficient levels of the progranulin (PGRN) protein, resulting in frontotemporal dementia (FTD) with TAR DNA-binding protein 43 (TDP-43) inclusions, classified pathologically as frontotemporal lobar degeneration (FTLD-TDP). Homozygous GRN mutations are exceedingly rare and cause neuronal ceroid lipofuscinosis 11, a lysosomal storage disease with onset in young adulthood, or an FTD syndrome with late-onset manifestations. In this review, we highlight the broad spectrum of clinical phenotypes associated with PGRN deficiency, including primary progressive aphasia and behavioral variant of frontotemporal dementia. We explore these phenotypes alongside relevant rodent and in vitro human models, ranging from the induced pluripotent stem cell-derived neural progenitors, neurons, microglia, and astrocytes to genetically engineered heterotypic organoids containing both neurons and astrocytes. We summarize advantages and limitations of these models in recapitulating the main FTLD-GRN hallmarks, highlighting the role of non-cell-autonomous mechanisms in the formation of TDP-43 pathology, neuroinflammation, and neurodegeneration. Data obtained from patients' brain tissues and biofluids, in parallel with single-cell transcriptomics, demonstrate the complexity of interactions among the highly heterogeneous cellular clusters present in the brain, including neurons, astrocytes, microglia, oligodendroglia, endothelial cells, and pericytes. Emerging evidence has revealed that PGRN deficiency is associated with cell cluster-specific, often conserved, genetic and molecular phenotypes in the central nervous system. In this review, we focus on how these distinct cellular populations and their dysfunctional crosstalk contribute to neurodegeneration and neuroinflammation in FTD-GRN. Specifically, we characterize the phenotypes of lipid droplet-accumulating microglia and alterations of myelin lipid content resulting from lysosomal dysfunction caused by PGRN deficiency. Additionally, we consider how the deregulation of glia-neuron communication affects the exchange of organelles such as mitochondria, and the removal of excess toxic products such as protein aggregates, in PGRN-related neurodegeneration.
Collapse
Affiliation(s)
- Katarzyna Gaweda-Walerych
- Department of Neurogenetics and Functional Genomics, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106, Warsaw, Poland.
| | - Vanessa Aragona
- Department of Biomedical Sciences, Humanitas University, Via Levi Montalicini 4, Pieve Emanuele, 20072, Milan, Italy
- Neurodevelopment Biology Lab, IRCCS Humanitas Research Hospital, via Manzoni, 56, Rozzano, 20089, Milan, Italy
| | - Simona Lodato
- Department of Biomedical Sciences, Humanitas University, Via Levi Montalicini 4, Pieve Emanuele, 20072, Milan, Italy
- Neurodevelopment Biology Lab, IRCCS Humanitas Research Hospital, via Manzoni, 56, Rozzano, 20089, Milan, Italy
| | - Emilia J Sitek
- Division of Neurological and Psychiatric Nursing, Laboratory of Clinical Neuropsychology, Neurolinguistics, and Neuropsychotherapy, Faculty of Health Sciences, Medical University of Gdansk, 80-210, Gdansk, Poland.
- Neurology Department, St. Adalbert Hospital, Copernicus PL, 80-462, Gdansk, Poland.
| | - Ewa Narożańska
- Neurology Department, St. Adalbert Hospital, Copernicus PL, 80-462, Gdansk, Poland
| | - Emanuele Buratti
- Molecular Pathology Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), AREA Science Park, 34149, Trieste, Italy
| |
Collapse
|
2
|
Vidyawan V, Puspita L, Juwono VB, Deline M, Pieknell K, Chang MY, Lee SH, Shim JW. Autophagy controls neuronal differentiation by regulating the WNT-DVL signaling pathway. Autophagy 2025; 21:719-736. [PMID: 39385328 DOI: 10.1080/15548627.2024.2407707] [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: 02/01/2024] [Revised: 09/10/2024] [Accepted: 09/18/2024] [Indexed: 10/12/2024] Open
Abstract
Macroautophagy/autophagy dysregulation is associated with various neurological diseases, including Vici syndrome. We aimed to determine the role of autophagy in early brain development. We generated neurons from human embryonic stem cells and developed a Vici syndrome model by introducing a loss-of-function mutation in the EPG5 gene. Autophagy-related genes were upregulated at the neuronal progenitor cell stage. Inhibition of autolysosome formation with bafilomycin A1 treatment at the neuronal progenitor cell stage delayed neuronal differentiation. Notably, WNT (Wnt family member) signaling may be part of the underlying mechanism, which is negatively regulated by autophagy-mediated DVL2 (disheveled segment polarity protein 2) degradation. Disruption of autolysosome formation may lead to failure in the downregulation of WNT signaling, delaying neuronal differentiation. EPG5 mutations disturbed autolysosome formation, subsequently inducing defects in progenitor cell differentiation and cortical layer generation in organoids. Disrupted autophagy leads to smaller organoids, recapitulating Vici syndrome-associated microcephaly, and validating the disease relevance of our study.Abbreviations: BafA1: bafilomycin A1; co-IP: co-immunoprecipitation; DVL2: dishevelled segment polarity protein 2; EPG5: ectopic P-granules 5 autophagy tethering factor; gRNA, guide RNA; hESC: human embryonic stem cells; KO: knockout; mDA, midbrain dopamine; NIM: neural induction media; NPC: neuronal progenitor cell; qPCR: quantitative polymerase chain reaction; UPS: ubiquitin-proteasome system; WNT: Wnt family member; WT: wild type.
Collapse
Affiliation(s)
- Vincencius Vidyawan
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-Si, Korea
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan-Si, Korea
| | - Lesly Puspita
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-Si, Korea
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan-Si, Korea
| | - Virginia Blessy Juwono
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-Si, Korea
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan-Si, Korea
| | - Magdalena Deline
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-Si, Korea
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan-Si, Korea
| | - Kelvin Pieknell
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea
- Biomedical Research Institute, Hanyang University, Seoul, Korea
| | - Mi-Yoon Chang
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea
- Biomedical Research Institute, Hanyang University, Seoul, Korea
- Department of Premedicine, College of Medicine, Hanyang University, Seoul, Korea
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul, Korea
| | - Sang-Hun Lee
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea
- Biomedical Research Institute, Hanyang University, Seoul, Korea
- Department of Biochemistry & Molecular Biology, College of Medicine, Hanyang University, Seoul, Korea
| | - Jae-Won Shim
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-Si, Korea
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan-Si, Korea
| |
Collapse
|
3
|
Cark O, Katkat E, Aydogdu I, Iscan E, Oktay Y, Ozhan G. tubg1 Somatic Mutants Show Tubulinopathy-Associated Neurodevelopmental Phenotypes in a Zebrafish Model. Mol Neurobiol 2025; 62:3024-3039. [PMID: 39215931 DOI: 10.1007/s12035-024-04448-2] [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: 05/11/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
Development of the multilayered cerebral cortex relies on precise orchestration of neurogenesis, neuronal migration, and differentiation, processes tightly regulated by microtubule dynamics. Mutations in tubulin superfamily genes have been associated with tubulinopathies, encompassing a spectrum of cortical malformations including microcephaly and lissencephaly. Here, we focus on γ-tubulin, a pivotal regulator of microtubule nucleation encoded by TUBG1. We investigate its role in brain development using a zebrafish model with somatic tubg1 mutation, recapitulating features of TUBG1-associated tubulinopathies in patients and mouse disease models. We demonstrate that γ-tubulin deficiency disrupts neurogenesis and brain development, mirroring microcephaly phenotypes. Furthermore, we uncover a novel potential regulatory link between γ-tubulin and canonical Wnt/β-catenin signaling, with γ-tubulin deficiency impairing Wnt activity. Our findings provide insights into the pathogenesis of cortical defects and suggest that γ-tubulin could be a potential target for further research in neurodevelopmental disorders, although challenges such as mode of action, specificity, and potential side effects must be addressed.
Collapse
Affiliation(s)
- Ozge Cark
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Inciralti-Balcova 35340, Izmir, Türkiye
- Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, Inciralti-Balcova 35340, Izmir, Türkiye
- Center for Regenerative Therapies at the TU Dresden, Technische Universität Dresden, 01307, Dresden, Germany
| | - Esra Katkat
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Inciralti-Balcova 35340, Izmir, Türkiye
- Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, Inciralti-Balcova 35340, Izmir, Türkiye
| | - Ipek Aydogdu
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Inciralti-Balcova 35340, Izmir, Türkiye
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Urla, 35430, Izmir, Türkiye
| | - Evin Iscan
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Inciralti-Balcova 35340, Izmir, Türkiye
- Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, Inciralti-Balcova 35340, Izmir, Türkiye
| | - Yavuz Oktay
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Inciralti-Balcova 35340, Izmir, Türkiye
- Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, Inciralti-Balcova 35340, Izmir, Türkiye
- Department of Medical Biology, School of Medicine, Dokuz Eylul University, Izmir, 35340, Türkiye
| | - Gunes Ozhan
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Inciralti-Balcova 35340, Izmir, Türkiye.
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Urla, 35430, Izmir, Türkiye.
| |
Collapse
|
4
|
Cezar LC, da Fonseca CCN, Klein MO, Kirsten TB, Felicio LF. Prenatal Valproic Acid Induces Autistic-Like Behaviors in Rats via Dopaminergic Modulation in Nigrostriatal and Mesocorticolimbic Pathways. J Neurochem 2025; 169:e16282. [PMID: 39801243 DOI: 10.1111/jnc.16282] [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: 07/08/2024] [Revised: 11/19/2024] [Accepted: 11/26/2024] [Indexed: 05/02/2025]
Abstract
Autism spectrum disorder (ASD) is a complex developmental disorder characterized by several behavioral impairments, especially in socialization, communication, and the occurrence of stereotyped behaviors. In rats, prenatal exposure to valproic acid (VPA) induces autistic-like behaviors. Previous studies by our group have suggested that the autistic-like phenotype is possibly related to dopaminergic system modulation because tyrosine hydroxylase (TH) expression was affected. The objective of the present study was to understand the dopaminergic role in autism. Wistar rats on gestational day 12.5 received VPA (400 mg/kg) and behaviors related to rat models of ASD were evaluated in juvenile offspring. Neurochemical and genetic dopaminergic components were studied in different brain areas of both juvenile and adult rats. Prenatal VPA-induced autistic-like behaviors in comparison to a control group: decreased maternal solicitations by ultrasonic vocalizations, cognitive inflexibility and stereotyped behavior in the T-maze test, decreased social interaction and play behavior, as well as motor hyperactivity. Prenatal VPA also decreased dopamine synthesis and activity in the striatum and prefrontal cortex, as well as dopamine transporter, D1 and D2 receptors, and TH expressions. Moreover, prenatal VPA increased TH+ immunoreactive neurons of the ventral tegmental area-substantia nigra complex. In conclusion, the dopaminergic hypoactivity associated with the behavioral impairments exhibited by the rats that received prenatal VPA suggests the important role of this system in the establishment of the characteristic symptoms of ASD in juvenile and adult males. Dopamine was demonstrated to be an important biomarker and a potential pharmacological target for ASD.
Collapse
Affiliation(s)
- Luana C Cezar
- Department of Pathology, School of Veterinary Medicine, University of São Paulo, Sao Paulo, Brazil
| | | | - Marianne O Klein
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, Sao Paulo, Brazil
| | - Thiago B Kirsten
- Psychoneuroimmunology Laboratory, Program in Environmental and Experimental Pathology, Paulista University, Sao Paulo, Brazil
| | - Luciano F Felicio
- Department of Pathology, School of Veterinary Medicine, University of São Paulo, Sao Paulo, Brazil
| |
Collapse
|
5
|
Li J, Hu M, Liu Y, Lu R, Feng W. Lead exposure leads to premature neural differentiation via inhibiting Wnt signaling. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 363:125232. [PMID: 39489322 DOI: 10.1016/j.envpol.2024.125232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/10/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
Heavy metals, such as Lead (Pb), are ubiquitous environmental pollutants that is a considerable problem worldwide. Increasing evidences suggest that Pb exposure negatively impact central nervous system. However, the exact toxic mechanism of Pb on early human brain development remain unclear due to the limitations of animal models and 2D cell models. In this study, we used human cortical organoids to reveal that Pb had specific early neurodevelopmental toxicity during the neural differentiation stage. We observed that short-term Pb exposure (10 days) is sufficient to induce premature neuronal differentiation. Mechanistically, Pb exposure downregulates the Wnt signaling in cortical organoids, and the activation of Wnt signaling reverses the neurodevelopmental phenotype. In support, Pb exposure during pregnancy lead to premature neuronal differentiation and reduced neurogenesis in mice. In conclusion, our study reveals the neuropathogenesis of Pb exposure and uncovers the potential intervention role of Wnt activation.
Collapse
Affiliation(s)
- Jun Li
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Meixin Hu
- Department of Child Health Care, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Yingying Liu
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Rongrong Lu
- Department of Neurosurgery, Children's Hospital of Fudan University, National Children's Medical Center (Shanghai), Shanghai, 201102, China
| | - Weijun Feng
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China; Fujian Key Laboratory of Neonatal Diseases, Xiamen Key Laboratory of Neonatal Diseases, Xiamen Children's Hospital, Children's Hospital of Fudan University at Xiamen, Xiamen, 361006, China.
| |
Collapse
|
6
|
Dorsey SG, Mocci E, Lane MV, Krueger BK. Rapid effects of valproic acid on the fetal brain transcriptome: implications for brain development and autism. Transl Psychiatry 2024; 14:482. [PMID: 39632793 PMCID: PMC11618798 DOI: 10.1038/s41398-024-03179-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 10/27/2024] [Accepted: 11/05/2024] [Indexed: 12/07/2024] Open
Abstract
There is an increased incidence of autism among the children of women who take the anti-epileptic, mood-stabilizing drug, valproic acid (VPA) during pregnancy; moreover, exposure to VPA in utero causes autistic-like symptoms in rodents and non-human primates. Analysis of RNA-seq data obtained from E12.5 fetal mouse brains 3 hours after VPA administration to the pregnant dam revealed that VPA rapidly and significantly increased or decreased the expression of approximately 7,300 genes. No significant sex differences in VPA-induced gene expression were observed. Expression of 399 autism risk genes was significantly altered by VPA as was expression of 258 genes that have been reported to modulate fetal brain development but are not otherwise linked to autism. Expression of genes associated with intracellular signaling pathways, neurogenesis, and excitation-inhibition balance as well as synaptogenesis, neuronal fate determination, axon and dendritic development, neuroinflammation, circadian rhythms, and epigenetic modulation of gene expression was dysregulated by VPA. Notably, at least 40 genes that are known to regulate embryonic neurogenesis were dysregulated by VPA. The goal of this study was to identify mouse genes that are: (a) significantly up- or downregulated by VPA in the fetal brain and (b) associated with autism and/or known to play a role in embryonic neurodevelopmental processes, perturbation of which has the potential to alter brain connectivity and, consequently behavior, in the adult. The genes meeting these criteria provide potential targets for future hypothesis-driven studies to elucidate the proximal causes of errors in brain connectivity underlying neurodevelopmental disorders such as autism.
Collapse
Affiliation(s)
- Susan G Dorsey
- Department of Pain and Translational Symptom Science University of Maryland School of Nursing, Baltimore, MD, 21201, USA
| | - Evelina Mocci
- Department of Pain and Translational Symptom Science University of Maryland School of Nursing, Baltimore, MD, 21201, USA
- Institute for Genome Sciences University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Malcolm V Lane
- Translational Toxicology/Department of Epidemiology and Public Health University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Bruce K Krueger
- Departments of Physiology and Psychiatry University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| |
Collapse
|
7
|
Moffat A, Schuurmans C. The Control of Cortical Folding: Multiple Mechanisms, Multiple Models. Neuroscientist 2024; 30:704-722. [PMID: 37621149 PMCID: PMC11558946 DOI: 10.1177/10738584231190839] [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] [Indexed: 08/26/2023]
Abstract
The cerebral cortex develops through a carefully conscripted series of cellular and molecular events that culminate in the production of highly specialized neuronal and glial cells. During development, cortical neurons and glia acquire a precise cellular arrangement and architecture to support higher-order cognitive functioning. Decades of study using rodent models, naturally gyrencephalic animal models, human pathology specimens, and, recently, human cerebral organoids, reveal that rodents recapitulate some but not all the cellular and molecular features of human cortices. Whereas rodent cortices are smooth-surfaced or lissencephalic, larger mammals, including humans and nonhuman primates, have highly folded/gyrencephalic cortices that accommodate an expansion in neuronal mass and increase in surface area. Several genes have evolved to drive cortical gyrification, arising from gene duplications or de novo origins, or by alterations to the structure/function of ancestral genes or their gene regulatory regions. Primary cortical folds arise in stereotypical locations, prefigured by a molecular "blueprint" that is set up by several signaling pathways (e.g., Notch, Fgf, Wnt, PI3K, Shh) and influenced by the extracellular matrix. Mutations that affect neural progenitor cell proliferation and/or neurogenesis, predominantly of upper-layer neurons, perturb cortical gyrification. Below we review the molecular drivers of cortical folding and their roles in disease.
Collapse
Affiliation(s)
- Alexandra Moffat
- Sunnybrook Research Institute, Biological Sciences Platform, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Carol Schuurmans
- Sunnybrook Research Institute, Biological Sciences Platform, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
8
|
Morey RA, Zheng Y, Bayly H, Sun D, Garrett ME, Gasperi M, Maihofer AX, Baird CL, Grasby KL, Huggins AA, Haswell CC, Thompson PM, Medland S, Gustavson DE, Panizzon MS, Kremen WS, Nievergelt CM, Ashley-Koch AE, Logue MW. Genomic structural equation modeling reveals latent phenotypes in the human cortex with distinct genetic architecture. Transl Psychiatry 2024; 14:451. [PMID: 39448598 PMCID: PMC11502831 DOI: 10.1038/s41398-024-03152-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/30/2024] [Accepted: 10/03/2024] [Indexed: 10/26/2024] Open
Abstract
Genetic contributions to human cortical structure manifest pervasive pleiotropy. This pleiotropy may be harnessed to identify unique genetically-informed parcellations of the cortex that are neurobiologically distinct from functional, cytoarchitectural, or other cortical parcellation schemes. We investigated genetic pleiotropy by applying genomic structural equation modeling (SEM) to map the genetic architecture of cortical surface area (SA) and cortical thickness (CT) for 34 brain regions recently reported in the ENIGMA cortical GWAS. Genomic SEM uses the empirical genetic covariance estimated from GWAS summary statistics with LD score regression (LDSC) to discover factors underlying genetic covariance, which we are denoting genetically informed brain networks (GIBNs). Genomic SEM can fit a multivariate GWAS from summary statistics for each of the GIBNs, which can subsequently be used for LD score regression (LDSC). We found the best-fitting model of cortical SA identified 6 GIBNs and CT identified 4 GIBNs, although sensitivity analyses indicated that other structures were plausible. The multivariate GWASs of the GIBNs identified 74 genome-wide significant (GWS) loci (p < 5 × 10-8), including many previously implicated in neuroimaging phenotypes, behavioral traits, and psychiatric conditions. LDSC of GIBN GWASs found that SA-derived GIBNs had a positive genetic correlation with bipolar disorder (BPD), and cannabis use disorder, indicating genetic predisposition to a larger SA in the specific GIBN is associated with greater genetic risk of these disorders. A negative genetic correlation was observed between attention deficit hyperactivity disorder (ADHD) and major depressive disorder (MDD). CT GIBNs displayed a negative genetic correlation with alcohol dependence. Even though we observed model instability in our application of genomic SEM to high-dimensional data, jointly modeling the genetic architecture of complex traits and investigating multivariate genetic links across neuroimaging phenotypes offers new insights into the genetics of cortical structure and relationships to psychopathology.
Collapse
Affiliation(s)
- Rajendra A Morey
- Brain Imaging and Analysis Center, Duke University, Durham, NC, 27710, USA
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC, USA
- VISN 6 MIRECC, VA Health Care System, Croasdaile Drive, Durham, NC, 27705, USA
| | - Yuanchao Zheng
- National Center for PTSD, VA Boston Healthcare System, Boston, MA, 02130, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, 02118, USA
| | - Henry Bayly
- National Center for PTSD, VA Boston Healthcare System, Boston, MA, 02130, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, 02118, USA
| | - Delin Sun
- Brain Imaging and Analysis Center, Duke University, Durham, NC, 27710, USA
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC, USA
- VISN 6 MIRECC, VA Health Care System, Croasdaile Drive, Durham, NC, 27705, USA
| | - Melanie E Garrett
- VISN 6 MIRECC, VA Health Care System, Croasdaile Drive, Durham, NC, 27705, USA
- Department of Medicine, Duke Molecular Physiology Institute, Carmichael Building, Duke University Medical Center, Durham, NC, 27701, USA
| | - Marianna Gasperi
- VA Center of Excellence for Stress and Mental Health, VA San Diego Healthcare System, San Diego, CA, 92161, USA
- Research Service VA, San Diego Healthcare System, San Diego, CA, 92161, USA
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
| | - Adam X Maihofer
- Research Service VA, San Diego Healthcare System, San Diego, CA, 92161, USA
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
| | - C Lexi Baird
- Brain Imaging and Analysis Center, Duke University, Durham, NC, 27710, USA
| | - Katrina L Grasby
- Psychiatric Genetics, QIMR, Berghofer Medical Research Institute, 4006, Brisbane, QLD, Australia
| | - Ashley A Huggins
- Brain Imaging and Analysis Center, Duke University, Durham, NC, 27710, USA
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC, USA
- VISN 6 MIRECC, VA Health Care System, Croasdaile Drive, Durham, NC, 27705, USA
| | - Courtney C Haswell
- Brain Imaging and Analysis Center, Duke University, Durham, NC, 27710, USA
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC, USA
| | - Paul M Thompson
- Imaging Genetics Center, Stevens Neuroimaging & Informatics Institute Keck School of Medicine University of Southern California, Los Angeles, CA, 90033, USA
| | - Sarah Medland
- Queensland Institute for Medical Research, Berghofer Medical Research Institute, 4006, Brisbane, QLD, Australia
| | - Daniel E Gustavson
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, 80303, USA
| | - Matthew S Panizzon
- Stein Institute for Research on Aging, University of California San Diego, La Jolla, CA, 92093, USA
| | - William S Kremen
- Stein Institute for Research on Aging, University of California San Diego, La Jolla, CA, 92093, USA
| | - Caroline M Nievergelt
- VA Center of Excellence for Stress and Mental Health, VA San Diego Healthcare System, San Diego, CA, 92161, USA
- Research Service VA, San Diego Healthcare System, San Diego, CA, 92161, USA
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
| | - Allison E Ashley-Koch
- VISN 6 MIRECC, VA Health Care System, Croasdaile Drive, Durham, NC, 27705, USA
- Department of Medicine, Duke Molecular Physiology Institute, Carmichael Building, Duke University Medical Center, Durham, NC, 27701, USA
| | - Mark W Logue
- National Center for PTSD, VA Boston Healthcare System, Boston, MA, 02130, USA.
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, 02118, USA.
- Department of Psychiatry, Boston University School of Medicine, Boston, MA, 02118, USA.
- Biomedical Genetics, Boston University School of Medicine, Boston, MA, 02118-2526, USA.
| |
Collapse
|
9
|
Todorov H, Weißbach S, Schlichtholz L, Mueller H, Hartwich D, Gerber S, Winter J. Stage-specific expression patterns and co-targeting relationships among miRNAs in the developing mouse cerebral cortex. Commun Biol 2024; 7:1366. [PMID: 39433948 PMCID: PMC11493953 DOI: 10.1038/s42003-024-07092-7] [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: 05/14/2024] [Accepted: 10/16/2024] [Indexed: 10/23/2024] Open
Abstract
microRNAs are crucial regulators of brain development, however, miRNA regulatory networks are not sufficiently well characterized. By performing small RNA-seq of the mouse embryonic cortex at E14, E17, and P0 as well as in neural progenitor cells and neurons, here we detected clusters of miRNAs that were co-regulated at distinct developmental stages. miRNAs such as miR-92a/b acted as hubs during early, and miR-124 and miR-137 during late neurogenesis. Notably, validated targets of P0 hub miRNAs were enriched for downregulated genes related to stem cell proliferation, negative regulation of neuronal differentiation and RNA splicing, among others, suggesting that miRNAs are particularly important for modulating transcriptional programs of crucial factors that guide the switch to neuronal differentiation. As most genes contain binding sites for more than one miRNA, we furthermore constructed a co-targeting network where numerous miRNAs shared more targets than expected by chance. Using luciferase reporter assays, we demonstrated that simultaneous binding of miRNA pairs to neurodevelopmentally relevant genes exerted an enhanced transcriptional silencing effect compared to single miRNAs. Taken together, we provide a comprehensive resource of miRNA longitudinal expression changes during murine corticogenesis. Furthermore, we highlight several potential mechanisms through which miRNA regulatory networks can shape embryonic brain development.
Collapse
Affiliation(s)
- Hristo Todorov
- Institute of Human Genetics, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Stephan Weißbach
- Institute of Human Genetics, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Institute of Developmental Biology and Neurobiology (iDN), Johannes Gutenberg University Mainz, Mainz, Germany
| | - Laura Schlichtholz
- Institute of Human Genetics, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Focus Program of Translational Neurosciences, University Medical Center Mainz, Mainz, Germany
| | - Hanna Mueller
- Institute of Human Genetics, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Dewi Hartwich
- Institute of Human Genetics, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Susanne Gerber
- Institute of Human Genetics, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.
| | - Jennifer Winter
- Institute of Human Genetics, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.
| |
Collapse
|
10
|
Shaw NC, Chen K, Farley KO, Hedges M, Forbes C, Baynam G, Lassmann T, Fear VS. Identifying SETBP1 haploinsufficiency molecular pathways to improve patient diagnosis using induced pluripotent stem cells and neural disease modelling. Mol Autism 2024; 15:42. [PMID: 39350244 PMCID: PMC11443744 DOI: 10.1186/s13229-024-00625-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND SETBP1 Haploinsufficiency Disorder (SETBP1-HD) is characterised by mild to moderate intellectual disability, speech and language impairment, mild motor developmental delay, behavioural issues, hypotonia, mild facial dysmorphisms, and vision impairment. Despite a clear link between SETBP1 mutations and neurodevelopmental disorders the precise role of SETBP1 in neural development remains elusive. We investigate the functional effects of three SETBP1 genetic variants including two pathogenic mutations p.Glu545Ter and SETBP1 p.Tyr1066Ter, resulting in removal of SKI and/or SET domains, and a point mutation p.Thr1387Met in the SET domain. METHODS Genetic variants were introduced into induced pluripotent stem cells (iPSCs) and subsequently differentiated into neurons to model the disease. We measured changes in cellular differentiation, SETBP1 protein localisation, and gene expression changes. RESULTS The data indicated a change in the WNT pathway, RNA polymerase II pathway and identified GATA2 as a central transcription factor in disease perturbation. In addition, the genetic variants altered the expression of gene sets related to neural forebrain development matching characteristics typical of the SETBP1-HD phenotype. LIMITATIONS The study investigates changes in cellular function in differentiation of iPSC to neural progenitor cells as a human model of SETBP1 HD disorder. Future studies may provide additional information relevant to disease on further neural cell specification, to derive mature neurons, neural forebrain cells, or brain organoids. CONCLUSIONS We developed a human SETBP1-HD model and identified perturbations to the WNT and POL2RA pathway, genes regulated by GATA2. Strikingly neural cells for both the SETBP1 truncation mutations and the single nucleotide variant displayed a SETBP1-HD-like phenotype.
Collapse
Affiliation(s)
- Nicole C Shaw
- The Kids Research Institute of Australia, The University of Western Australia, Nedlands, WA, Australia
| | - Kevin Chen
- The Kids Research Institute of Australia, The University of Western Australia, Nedlands, WA, Australia
| | - Kathryn O Farley
- The Kids Research Institute of Australia, The University of Western Australia, Nedlands, WA, Australia
| | - Mitchell Hedges
- The Kids Research Institute of Australia, The University of Western Australia, Nedlands, WA, Australia
| | - Catherine Forbes
- The Kids Research Institute of Australia, The University of Western Australia, Nedlands, WA, Australia
| | - Gareth Baynam
- Rare Care Centre, Perth Children's Hospital, Nedlands, WA, Australia
| | - Timo Lassmann
- The Kids Research Institute of Australia, The University of Western Australia, Nedlands, WA, Australia
| | - Vanessa S Fear
- The Kids Research Institute of Australia, The University of Western Australia, Nedlands, WA, Australia.
| |
Collapse
|
11
|
Boonsawat P, Asadollahi R, Niedrist D, Steindl K, Begemann A, Joset P, Bhoj EJ, Li D, Zackai E, Vetro A, Barba C, Guerrini R, Whalen S, Keren B, Khan A, Jing D, Palomares Bralo M, Rikeros Orozco E, Hao Q, Schlott Kristiansen B, Zheng B, Donnelly D, Clowes V, Zweier M, Papik M, Siegel G, Sabatino V, Mocera M, Horn AHC, Sticht H, Rauch A. Deleterious ZNRF3 germline variants cause neurodevelopmental disorders with mirror brain phenotypes via domain-specific effects on Wnt/β-catenin signaling. Am J Hum Genet 2024; 111:1994-2011. [PMID: 39168120 PMCID: PMC11393693 DOI: 10.1016/j.ajhg.2024.07.016] [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: 04/29/2024] [Revised: 07/24/2024] [Accepted: 07/24/2024] [Indexed: 08/23/2024] Open
Abstract
Zinc and RING finger 3 (ZNRF3) is a negative-feedback regulator of Wnt/β-catenin signaling, which plays an important role in human brain development. Although somatically frequently mutated in cancer, germline variants in ZNRF3 have not been established as causative for neurodevelopmental disorders (NDDs). We identified 12 individuals with ZNRF3 variants and various phenotypes via GeneMatcher/Decipher and evaluated genotype-phenotype correlation. We performed structural modeling and representative deleterious and control variants were assessed using in vitro transcriptional reporter assays with and without Wnt-ligand Wnt3a and/or Wnt-potentiator R-spondin (RSPO). Eight individuals harbored de novo missense variants and presented with NDD. We found missense variants associated with macrocephalic NDD to cluster in the RING ligase domain. Structural modeling predicted disruption of the ubiquitin ligase function likely compromising Wnt receptor turnover. Accordingly, the functional assays showed enhanced Wnt/β-catenin signaling for these variants in a dominant negative manner. Contrarily, an individual with microcephalic NDD harbored a missense variant in the RSPO-binding domain predicted to disrupt binding affinity to RSPO and showed attenuated Wnt/β-catenin signaling in the same assays. Additionally, four individuals harbored de novo truncating or de novo or inherited large in-frame deletion variants with non-NDD phenotypes, including heart, adrenal, or nephrotic problems. In contrast to NDD-associated missense variants, the effects on Wnt/β-catenin signaling were comparable between the truncating variant and the empty vector and between benign variants and the wild type. In summary, we provide evidence for mirror brain size phenotypes caused by distinct pathomechanisms in Wnt/β-catenin signaling through protein domain-specific deleterious ZNRF3 germline missense variants.
Collapse
Affiliation(s)
| | - Reza Asadollahi
- Institute of Medical Genetics, University of Zurich, Zurich, Switzerland; Faculty of Engineering and Science, University of Greenwich London, Medway Campus, Chatham Maritime ME4 4TB, UK
| | - Dunja Niedrist
- Institute of Medical Genetics, University of Zurich, Zurich, Switzerland
| | - Katharina Steindl
- Institute of Medical Genetics, University of Zurich, Zurich, Switzerland
| | - Anaïs Begemann
- Institute of Medical Genetics, University of Zurich, Zurich, Switzerland
| | - Pascal Joset
- Medical Genetics, University Hospital Basel, Basel, Switzerland
| | - Elizabeth J Bhoj
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Dong Li
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Elaine Zackai
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Annalisa Vetro
- Neuroscience Department, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Carmen Barba
- Neuroscience Department, Meyer Children's Hospital IRCCS, Florence, Italy; University of Florence, Florence, Italy
| | - Renzo Guerrini
- Neuroscience Department, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Sandra Whalen
- Unité Fonctionnelle de Génétique Odellin, Hôpital Armand Trousseau, Paris, France
| | - Boris Keren
- Département de Génétique, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Amjad Khan
- Faculty of Science, Department of Biological Science (Zoology), University of Lakki Marwat, Khyber Pakhtunkhwa 28420, Pakistan
| | - Duan Jing
- Shenzhen Children's Hospital, Shenzhen, Guangdong, China
| | - María Palomares Bralo
- Instituto de Genética Médica y Molecular (INGEMM), Unidad de Trastornos Del Neurodesarrollo, Hospital Universitario La Paz, Madrid, Spain
| | - Emi Rikeros Orozco
- Instituto de Genética Médica y Molecular (INGEMM), Unidad de Trastornos Del Neurodesarrollo, Hospital Universitario La Paz, Madrid, Spain
| | - Qin Hao
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
| | | | - Bixia Zheng
- Nanjing Key Laboratory of Pediatrics Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Deirdre Donnelly
- Northern Ireland Regional Genetics Centre, Belfast Health & Social Care Trust, Belfast, Northern Ireland
| | - Virginia Clowes
- Thames Regional Genetics Service, North West University Healthcare NHS Trust, London, UK
| | - Markus Zweier
- Institute of Medical Genetics, University of Zurich, Zurich, Switzerland
| | - Michael Papik
- Institute of Medical Genetics, University of Zurich, Zurich, Switzerland
| | - Gabriele Siegel
- Institute of Medical Genetics, University of Zurich, Zurich, Switzerland
| | - Valeria Sabatino
- Institute of Medical Genetics, University of Zurich, Zurich, Switzerland
| | - Martina Mocera
- Institute of Medical Genetics, University of Zurich, Zurich, Switzerland
| | - Anselm H C Horn
- Institute of Medical Genetics, University of Zurich, Zurich, Switzerland; Institute of Biochemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Heinrich Sticht
- Institute of Biochemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Anita Rauch
- Institute of Medical Genetics, University of Zurich, Zurich, Switzerland; Pediatric University Hospital Zurich, Zurich, Switzerland.
| |
Collapse
|
12
|
Moskalenko AM, Ikrin AN, Kozlova AV, Mukhamadeev RR, de Abreu MS, Riga V, Kolesnikova TO, Kalueff AV. Decoding Molecular Bases of Rodent Social Hetero-Grooming Behavior Using in Silico Analyses and Bioinformatics Tools. Neuroscience 2024; 554:146-155. [PMID: 38876356 DOI: 10.1016/j.neuroscience.2024.06.004] [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: 05/04/2024] [Revised: 06/03/2024] [Accepted: 06/06/2024] [Indexed: 06/16/2024]
Abstract
Highly prevalent in laboratory rodents, 'social' hetero-grooming behavior is translationally relevant to modeling a wide range of neuropsychiatric disorders. Here, we comprehensively evaluated all known to date mouse genes linked to aberrant hetero-grooming phenotype, and applied bioinformatics tools to construct a network of their established protein-protein interactions (PPI). We next identified several distinct molecular clusters within this complex network, including neuronal differentiation, cytoskeletal, WNT-signaling and synapsins-associated pathways. Using additional bioinformatics analyses, we further identified 'central' (hub) proteins within these molecular clusters, likely key for mouse hetero-grooming behavior. Overall, a more comprehensive characterization of intricate molecular pathways linked to aberrant rodent grooming may markedly advance our understanding of underlying cellular mechanisms and related neurological disorders, eventually helping discover novel targets for their pharmacological or gene therapy interventions.
Collapse
Affiliation(s)
- Anastasia M Moskalenko
- Graduate Program in Genetics and Genetic Technologies, Sirius University of Science and Technology, Sochi 354340, Russia; Neuroscience Department, Sirius University of Science and Technology, Sochi 354340, Russia
| | - Aleksey N Ikrin
- Graduate Program in Genetics and Genetic Technologies, Sirius University of Science and Technology, Sochi 354340, Russia; Neuroscience Department, Sirius University of Science and Technology, Sochi 354340, Russia
| | - Alena V Kozlova
- Graduate Program in Genetics and Genetic Technologies, Sirius University of Science and Technology, Sochi 354340, Russia
| | - Radmir R Mukhamadeev
- Graduate Program in Bioinformatics and Genomics, Sirius University of Science and Technology, Sochi 354340, Russia; Neuroscience Department, Sirius University of Science and Technology, Sochi 354340, Russia
| | - Murilo S de Abreu
- Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre 90050, Brazil.
| | - Vyacheslav Riga
- Neuroscience Department, Sirius University of Science and Technology, Sochi 354340, Russia
| | - Tatiana O Kolesnikova
- Neuroscience Department, Sirius University of Science and Technology, Sochi 354340, Russia
| | - Allan V Kalueff
- Neuroscience Department, Sirius University of Science and Technology, Sochi 354340, Russia; Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg 199034, Russia; Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg 194021, Russia; Suzhou Key Laboratory of Neurobiology and Cell Signaling, Department of Biological Sciences, School of Science, Xi'an Jiaotong-Liverpool University (XJTLU), Suzhou 215123, China.
| |
Collapse
|
13
|
Bhatt RR, Gadewar SP, Shetty A, Ba Gari I, Haddad E, Javid S, Ramesh A, Nourollahimoghadam E, Zhu AH, de Leeuw C, Thompson PM, Medland SE, Jahanshad N. The Genetic Architecture of the Human Corpus Callosum and its Subregions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.22.603147. [PMID: 39091796 PMCID: PMC11291056 DOI: 10.1101/2024.07.22.603147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
The corpus callosum (CC) is the largest set of white matter fibers connecting the two hemispheres of the brain. In humans, it is essential for coordinating sensorimotor responses, performing associative/executive functions, and representing information in multiple dimensions. Understanding which genetic variants underpin corpus callosum morphometry, and their shared influence on cortical structure and susceptibility to neuropsychiatric disorders, can provide molecular insights into the CC's role in mediating cortical development and its contribution to neuropsychiatric disease. To characterize the morphometry of the midsagittal corpus callosum, we developed a publicly available artificial intelligence based tool to extract, parcellate, and calculate its total and regional area and thickness. Using the UK Biobank (UKB) and the Adolescent Brain Cognitive Development study (ABCD), we extracted measures of midsagittal corpus callosum morphometry and performed a genome-wide association study (GWAS) meta-analysis of European participants (combined N = 46,685). We then examined evidence for generalization to the non-European participants of the UKB and ABCD cohorts (combined N = 7,040). Post-GWAS analyses implicate prenatal intracellular organization and cell growth patterns, and high heritability in regions of open chromatin, suggesting transcriptional activity regulation in early development. Results suggest programmed cell death mediated by the immune system drives the thinning of the posterior body and isthmus. Global and local genetic overlap, along with causal genetic liability, between the corpus callosum, cerebral cortex, and neuropsychiatric disorders such as attention-deficit/hyperactivity and bipolar disorders were identified. These results provide insight into variability of corpus callosum development, its genetic influence on the cerebral cortex, and biological mechanisms related to neuropsychiatric dysfunction.
Collapse
Affiliation(s)
- Ravi R Bhatt
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Shruti P Gadewar
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Ankush Shetty
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Iyad Ba Gari
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Elizabeth Haddad
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Shayan Javid
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Abhinaav Ramesh
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Elnaz Nourollahimoghadam
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Alyssa H Zhu
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Christiaan de Leeuw
- Department of Complex Trait Genetics, Centre for Neurogenomics and Cognitive Research, VU University, Amsterdam, The Netherlands
| | - Paul M Thompson
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Sarah E Medland
- Psychiatric Genetics, QIMR Berghofer Medical Research Institute, Brisbane 4006, Australia
| | - Neda Jahanshad
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| |
Collapse
|
14
|
Su L, Zhang M, Ji F, Zhao J, Wang Y, Wang W, Zhang S, Ma H, Wang Y, Jiao J. Microglia homeostasis mediated by epigenetic ARID1A regulates neural progenitor cells response and leads to autism-like behaviors. Mol Psychiatry 2024; 29:1595-1609. [PMID: 35858990 DOI: 10.1038/s41380-022-01703-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 07/01/2022] [Accepted: 07/05/2022] [Indexed: 01/26/2023]
Abstract
Microglia are resident macrophages of the central nervous system that selectively emerge in embryonic cortical proliferative zones and regulate neurogenesis by altering molecular and phenotypic states. Despite their important roles in inflammatory phagocytosis and neurodegenerative diseases, microglial homeostasis during early brain development has not been fully elucidated. Here, we demonstrate a notable interplay between microglial homeostasis and neural progenitor cell signal transduction during embryonic neurogenesis. ARID1A, an epigenetic subunit of the SWI/SNF chromatin-remodeling complex, disrupts genome-wide H3K9me3 occupancy in microglia and changes the epigenetic chromatin landscape of regulatory elements that influence the switching of microglial states. Perturbation of microglial homeostasis impairs the release of PRG3, which regulates neural progenitor cell self-renewal and differentiation during embryonic development. Furthermore, the loss of microglia-driven PRG3 alters the downstream cascade of the Wnt/β-catenin signaling pathway through its interaction with the neural progenitor receptor LRP6, which leads to misplaced regulation in neuronal development and causes autism-like behaviors at later stages. Thus, during early fetal brain development, microglia progress toward a more homeostatic competent phenotype, which might render neural progenitor cells respond to environmental cross-talk perturbations.
Collapse
Affiliation(s)
- Libo Su
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
| | - Mengtian Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
| | - Fen Ji
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
| | - Jinyue Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
| | - Yuanyuan Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
| | - Wenwen Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- School of Life Sciences, University of Science and Technology of China, Hefei, 230026, China
| | - Shukui Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- College of Life Sciences, Yantai University, Yantai, 264005, Shandong, China
| | - Hongyan Ma
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
| | - Yanyan Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
| | - Jianwei Jiao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China.
- University of Chinese Academy of Sciences, 100049, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China.
| |
Collapse
|
15
|
Knol MJ, Poot RA, Evans TE, Satizabal CL, Mishra A, Sargurupremraj M, van der Auwera S, Duperron MG, Jian X, Hostettler IC, van Dam-Nolen DHK, Lamballais S, Pawlak MA, Lewis CE, Carrion-Castillo A, van Erp TGM, Reinbold CS, Shin J, Scholz M, Håberg AK, Kämpe A, Li GHY, Avinun R, Atkins JR, Hsu FC, Amod AR, Lam M, Tsuchida A, Teunissen MWA, Aygün N, Patel Y, Liang D, Beiser AS, Beyer F, Bis JC, Bos D, Bryan RN, Bülow R, Caspers S, Catheline G, Cecil CAM, Dalvie S, Dartigues JF, DeCarli C, Enlund-Cerullo M, Ford JM, Franke B, Freedman BI, Friedrich N, Green MJ, Haworth S, Helmer C, Hoffmann P, Homuth G, Ikram MK, Jack CR, Jahanshad N, Jockwitz C, Kamatani Y, Knodt AR, Li S, Lim K, Longstreth WT, Macciardi F, Mäkitie O, Mazoyer B, Medland SE, Miyamoto S, Moebus S, Mosley TH, Muetzel R, Mühleisen TW, Nagata M, Nakahara S, Palmer ND, Pausova Z, Preda A, Quidé Y, Reay WR, Roshchupkin GV, Schmidt R, Schreiner PJ, Setoh K, Shapland CY, Sidney S, St Pourcain B, Stein JL, Tabara Y, Teumer A, Uhlmann A, van der Lugt A, Vernooij MW, Werring DJ, Windham BG, Witte AV, Wittfeld K, Yang Q, Yoshida K, Brunner HG, Le Grand Q, et alKnol MJ, Poot RA, Evans TE, Satizabal CL, Mishra A, Sargurupremraj M, van der Auwera S, Duperron MG, Jian X, Hostettler IC, van Dam-Nolen DHK, Lamballais S, Pawlak MA, Lewis CE, Carrion-Castillo A, van Erp TGM, Reinbold CS, Shin J, Scholz M, Håberg AK, Kämpe A, Li GHY, Avinun R, Atkins JR, Hsu FC, Amod AR, Lam M, Tsuchida A, Teunissen MWA, Aygün N, Patel Y, Liang D, Beiser AS, Beyer F, Bis JC, Bos D, Bryan RN, Bülow R, Caspers S, Catheline G, Cecil CAM, Dalvie S, Dartigues JF, DeCarli C, Enlund-Cerullo M, Ford JM, Franke B, Freedman BI, Friedrich N, Green MJ, Haworth S, Helmer C, Hoffmann P, Homuth G, Ikram MK, Jack CR, Jahanshad N, Jockwitz C, Kamatani Y, Knodt AR, Li S, Lim K, Longstreth WT, Macciardi F, Mäkitie O, Mazoyer B, Medland SE, Miyamoto S, Moebus S, Mosley TH, Muetzel R, Mühleisen TW, Nagata M, Nakahara S, Palmer ND, Pausova Z, Preda A, Quidé Y, Reay WR, Roshchupkin GV, Schmidt R, Schreiner PJ, Setoh K, Shapland CY, Sidney S, St Pourcain B, Stein JL, Tabara Y, Teumer A, Uhlmann A, van der Lugt A, Vernooij MW, Werring DJ, Windham BG, Witte AV, Wittfeld K, Yang Q, Yoshida K, Brunner HG, Le Grand Q, Sim K, Stein DJ, Bowden DW, Cairns MJ, Hariri AR, Cheung CL, Andersson S, Villringer A, Paus T, Cichon S, Calhoun VD, Crivello F, Launer LJ, White T, Koudstaal PJ, Houlden H, Fornage M, Matsuda F, Grabe HJ, Ikram MA, Debette S, Thompson PM, Seshadri S, Adams HHH. Genetic variants for head size share genes and pathways with cancer. Cell Rep Med 2024; 5:101529. [PMID: 38703765 PMCID: PMC11148644 DOI: 10.1016/j.xcrm.2024.101529] [Show More Authors] [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: 11/30/2021] [Revised: 09/18/2023] [Accepted: 04/04/2024] [Indexed: 05/06/2024]
Abstract
The size of the human head is highly heritable, but genetic drivers of its variation within the general population remain unmapped. We perform a genome-wide association study on head size (N = 80,890) and identify 67 genetic loci, of which 50 are novel. Neuroimaging studies show that 17 variants affect specific brain areas, but most have widespread effects. Gene set enrichment is observed for various cancers and the p53, Wnt, and ErbB signaling pathways. Genes harboring lead variants are enriched for macrocephaly syndrome genes (37-fold) and high-fidelity cancer genes (9-fold), which is not seen for human height variants. Head size variants are also near genes preferentially expressed in intermediate progenitor cells, neural cells linked to evolutionary brain expansion. Our results indicate that genes regulating early brain and cranial growth incline to neoplasia later in life, irrespective of height. This warrants investigation of clinical implications of the link between head size and cancer.
Collapse
Affiliation(s)
- Maria J Knol
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Raymond A Poot
- Department of Cell Biology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Tavia E Evans
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, the Netherlands; Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Claudia L Satizabal
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX, USA; The Framingham Heart Study, Framingham, MA, USA; Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Aniket Mishra
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, team VINTAGE, UMR 1219, Bordeaux, France
| | - Muralidharan Sargurupremraj
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX, USA
| | - Sandra van der Auwera
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany; German Centre of Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, Greifswald, Germany
| | - Marie-Gabrielle Duperron
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, team VINTAGE, UMR 1219, Bordeaux, France
| | - Xueqiu Jian
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Isabel C Hostettler
- Stroke Research Centre, University College London, Institute of Neurology, London, UK; Department of Neurosurgery, Klinikum rechts der Isar, University of Munich, Munich, Germany; Neurosurgical Department, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Dianne H K van Dam-Nolen
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Sander Lamballais
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Mikolaj A Pawlak
- Department of Neurology, Poznań University of Medical Sciences, Poznań, Poland; Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Cora E Lewis
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham School of Medicine, Birmingham, AL, USA
| | - Amaia Carrion-Castillo
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, the Netherlands
| | - Theo G M van Erp
- Clinical Translational Neuroscience Laboratory, Department of Psychiatry and Human Behavior, University of California, Irvine, Irvine, CA, USA; Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, CA, USA
| | - Céline S Reinbold
- Department of Biomedicine, University of Basel, Basel, Switzerland; Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland; Institute of Computational Life Sciences, Zurich University of Applied Sciences, Wädenswil, Switzerland
| | - Jean Shin
- The Hospital for Sick Children, University of Toronto, Toronto, Canada; Departments of Physiology and Nutritional Sciences, University of Toronto, Toronto, Canada
| | - Markus Scholz
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany; LIFE Research Center for Civilization Disease, Leipzig, Germany
| | - Asta K Håberg
- Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology (NTNU), Trondheim, Norway; Department of Radiology and Nuclear Medicine, St. Olavs University Hospital, Trondheim, Norway
| | - Anders Kämpe
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
| | - Gloria H Y Li
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Reut Avinun
- Laboratory of NeuroGenetics, Department of Psychology & Neuroscience, Duke University, Durham, NC, USA
| | - Joshua R Atkins
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia; Centre for Brain and Mental Health Research, Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Fang-Chi Hsu
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Alyssa R Amod
- Department of Child and Adolescent Psychiatry, TU Dresden, Dresden, Germany
| | - Max Lam
- North Region, Institute of Mental Health, Singapore, Singapore; Population and Global Health, LKC Medicine, Nanyang Technological University, Singapore, Singapore
| | - Ami Tsuchida
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, team VINTAGE, UMR 1219, Bordeaux, France; Groupe d'imagerie neurofonctionnelle, Institut des Maladies Neurodégénératives, UMR 5293, CNRS, CEA, Université de Bordeaux, Bordeaux, France
| | - Mariël W A Teunissen
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Neurology, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Nil Aygün
- Department of Genetics UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yash Patel
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - Dan Liang
- Department of Genetics UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alexa S Beiser
- The Framingham Heart Study, Framingham, MA, USA; Department of Neurology, Boston University School of Medicine, Boston, MA, USA; Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Frauke Beyer
- Department of Neurology, Max Planck Institute for Cognitive and Brain Sciences, Leipzig, Germany; Collaborative Research Center 1052 Obesity Mechanisms, Faculty of Medicine, University of Leipzig, Leipzig, Germany; Day Clinic for Cognitive Neurology, University Hospital Leipzig, Leipzig, Germany
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Daniel Bos
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands; Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - R Nick Bryan
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Robin Bülow
- Institute of Diagnostic Radiology and Neuroradiology, University Medicine Greifswald, Greifswald, Germany
| | - Svenja Caspers
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany; Institute for Anatomy I, Medical Faculty & University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Gwenaëlle Catheline
- University of Bordeaux, CNRS, INCIA, UMR 5287, team NeuroImagerie et Cognition Humaine, Bordeaux, France; EPHE-PSL University, Bordeaux, France
| | - Charlotte A M Cecil
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands; Department of Child and Adolescent Psychiatry, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Shareefa Dalvie
- Department of Child and Adolescent Psychiatry, TU Dresden, Dresden, Germany
| | - Jean-François Dartigues
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, team SEPIA, UMR 1219, Bordeaux, France
| | - Charles DeCarli
- Department of Neurology and Center for Neuroscience, University of California at Davis, Sacramento, CA, USA
| | - Maria Enlund-Cerullo
- Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Folkhälsan Research Center, Helsinki, Finland
| | - Judith M Ford
- San Francisco Veterans Administration Medical Center, San Francisco, CA, USA; University of California, San Francisco, San Francisco, CA, USA
| | - Barbara Franke
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Psychiatry, Radboud University Medical Center, Nijmegen, the Netherlands; Donders Institute for Brain, Cognition, and Behaviour, Radboud University, Nijmegen, the Netherlands
| | - Barry I Freedman
- Department of Internal Medicine, Section on Nephrology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Nele Friedrich
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Melissa J Green
- School of Clinical Medicine, University of New South Wales, Sydney, NSW, Australia; Neuroscience Research Australia, Sydney, NSW, Australia
| | - Simon Haworth
- Bristol Dental School, University of Bristol, Bristol, UK
| | - Catherine Helmer
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, team LEHA, UMR 1219, Bordeaux, France
| | - Per Hoffmann
- Department of Biomedicine, University of Basel, Basel, Switzerland; Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland; Institute of Human Genetics, University of Bonn Medical School, Bonn, Germany
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - M Kamran Ikram
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands; Department of Neurology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | | | - Neda Jahanshad
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging & Informatics Institute, Keck USC School of Medicine, Los Angeles, CA, USA
| | - Christiane Jockwitz
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany; Department of Psychiatry, Psychotherapy and Psychosomatics, RWTH Aachen University, Medical Faculty, Aachen, Germany
| | - Yoichiro Kamatani
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Annchen R Knodt
- Laboratory of NeuroGenetics, Department of Psychology & Neuroscience, Duke University, Durham, NC, USA
| | - Shuo Li
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Keane Lim
- Research Division, Institute of Mental Health, Singapore, Singapore
| | - W T Longstreth
- Department of Neurology, University of Washington, Seattle, WA, USA; Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Fabio Macciardi
- Laboratory of Molecular Psychiatry, Department of Psychiatry and Human Behavior, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Outi Mäkitie
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden; Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Folkhälsan Research Center, Helsinki, Finland
| | - Bernard Mazoyer
- Groupe d'imagerie neurofonctionnelle, Institut des Maladies Neurodégénératives, UMR 5293, CNRS, CEA, Université de Bordeaux, Bordeaux, France; Centre Hospitalo-Universitaire de Bordeaux, Bordeaux, France
| | - Sarah E Medland
- Psychiatric Genetics, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia; School of Psychology, University of Queensland, Brisbane, QLD, Australia; Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Susumu Miyamoto
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Susanne Moebus
- Institute for Urban Public Health, University of Duisburg-Essen, Essen, Germany
| | - Thomas H Mosley
- Department of Medicine, Division of Geriatrics, University of Mississippi Medical Center, Jackson, MS, USA; Memory Impairment and Neurodegenerative Dementia (MIND) Center, Jackson, MS, USA
| | - Ryan Muetzel
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands; Department of Child and Adolescent Psychiatry, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Thomas W Mühleisen
- Department of Biomedicine, University of Basel, Basel, Switzerland; Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany; C. and O. Vogt Institute for Brain Research, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Manabu Nagata
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Soichiro Nakahara
- Clinical Translational Neuroscience Laboratory, Department of Psychiatry and Human Behavior, University of California, Irvine, Irvine, CA, USA; Unit 2, Candidate Discovery Science Labs, Drug Discovery Research, Astellas Pharma Inc, 21 Miyukigaoka, Tsukuba, Ibaraki 305-8585, Japan
| | - Nicholette D Palmer
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Zdenka Pausova
- The Hospital for Sick Children, University of Toronto, Toronto, Canada; Departments of Physiology and Nutritional Sciences, University of Toronto, Toronto, Canada
| | - Adrian Preda
- Department of Psychiatry, University of California, Irvine, Irvine, CA, USA
| | - Yann Quidé
- School of Clinical Medicine, University of New South Wales, Sydney, NSW, Australia; Neuroscience Research Australia, Sydney, NSW, Australia
| | - William R Reay
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia; Centre for Brain and Mental Health Research, Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Gennady V Roshchupkin
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands; Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Reinhold Schmidt
- Clinical Division of Neurogeriatrics, Department of Neurology, Medical University of Graz, Graz, Austria
| | | | - Kazuya Setoh
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Chin Yang Shapland
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, the Netherlands; MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK; Population Health Sciences, University of Bristol, Bristol, UK
| | - Stephen Sidney
- Kaiser Permanente Division of Research, Oakland, CA, USA
| | - Beate St Pourcain
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, the Netherlands; Donders Institute for Brain, Cognition, and Behaviour, Radboud University, Nijmegen, the Netherlands; MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Jason L Stein
- Department of Genetics UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yasuharu Tabara
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Alexander Teumer
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany; Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Anne Uhlmann
- Department of Child and Adolescent Psychiatry, TU Dresden, Dresden, Germany
| | - Aad van der Lugt
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Meike W Vernooij
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands; Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - David J Werring
- Stroke Research Centre, University College London, Institute of Neurology, London, UK
| | - B Gwen Windham
- Department of Medicine, Division of Geriatrics, University of Mississippi Medical Center, Jackson, MS, USA; Memory Impairment and Neurodegenerative Dementia (MIND) Center, Jackson, MS, USA
| | - A Veronica Witte
- Department of Neurology, Max Planck Institute for Cognitive and Brain Sciences, Leipzig, Germany; Collaborative Research Center 1052 Obesity Mechanisms, Faculty of Medicine, University of Leipzig, Leipzig, Germany; Day Clinic for Cognitive Neurology, University Hospital Leipzig, Leipzig, Germany
| | - Katharina Wittfeld
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany; German Centre of Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, Greifswald, Germany
| | - Qiong Yang
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Kazumichi Yoshida
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Han G Brunner
- Department of Human Genetics, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Clinical Genetics MUMC+, GROW School of Oncology and Developmental Biology, and MHeNs School of Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Quentin Le Grand
- Bordeaux Population Health, University of Bordeaux, INSERM U1219, Bordeaux, France
| | - Kang Sim
- West Region, Institute of Mental Health, Singapore, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Dan J Stein
- Department of Child and Adolescent Psychiatry, TU Dresden, Dresden, Germany; SAMRC Unit on Risk and Resilience, University of Cape Town, Cape Town, South Africa
| | - Donald W Bowden
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Murray J Cairns
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia; Centre for Brain and Mental Health Research, Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Ahmad R Hariri
- Laboratory of NeuroGenetics, Department of Psychology & Neuroscience, Duke University, Durham, NC, USA
| | - Ching-Lung Cheung
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; Centre for Genomic Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Sture Andersson
- Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Arno Villringer
- Department of Neurology, Max Planck Institute for Cognitive and Brain Sciences, Leipzig, Germany; Day Clinic for Cognitive Neurology, University Hospital Leipzig, Leipzig, Germany
| | - Tomas Paus
- Departments of Psychiatry and Neuroscience, Faculty of Medicine and Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, Montreal, QC, Canada; Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Sven Cichon
- Department of Biomedicine, University of Basel, Basel, Switzerland; Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland; Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany
| | - Vince D Calhoun
- Tri-institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS) {Georgia State, Georgia Tech, Emory}, Atlanta, GA, USA
| | - Fabrice Crivello
- Groupe d'imagerie neurofonctionnelle, Institut des Maladies Neurodégénératives, UMR 5293, CNRS, CEA, Université de Bordeaux, Bordeaux, France
| | - Lenore J Launer
- Laboratory of Epidemiology, Demography, and Biometry, Intramural Research Program, National Institute of Aging, The National Institutes of Health, Bethesda, MD, USA
| | - Tonya White
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, the Netherlands; Department of Child and Adolescent Psychiatry, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Peter J Koudstaal
- Department of Neurology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Henry Houlden
- Stroke Research Centre, University College London, Institute of Neurology, London, UK
| | - Myriam Fornage
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA; Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Fumihiko Matsuda
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hans J Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Stéphanie Debette
- Bordeaux Population Health, University of Bordeaux, INSERM U1219, Bordeaux, France; Department of Neurology, Bordeaux University Hospital, Bordeaux, France
| | - Paul M Thompson
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging & Informatics Institute, Keck USC School of Medicine, Los Angeles, CA, USA
| | - Sudha Seshadri
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX, USA; The Framingham Heart Study, Framingham, MA, USA; Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Hieab H H Adams
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands; Latin American Brain Health (BrainLat), Universidad Adolfo Ibáñez, Santiago, Chile.
| |
Collapse
|
16
|
Wang D, Ruan Z, Wang R, Ma L, Tang S, Wang X, Ma A. Decoding the mechanism of earthworm extract against wounds: an integrated metabolomics and network pharmacology study. Mol Divers 2024; 28:631-647. [PMID: 36705857 DOI: 10.1007/s11030-023-10609-7] [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: 10/06/2022] [Accepted: 01/19/2023] [Indexed: 01/28/2023]
Abstract
Earthworms are used to cure wounds in Chinese villages for thousands of years. Recently, scientists realized their extracts could promote wound healing and they have anti-inflammatory, antioxidant, anti-apoptosis, and anti-microbial properties, but its mechanism of promoting wound healing remains unclear. In the presented study, electronic literature databases and LC-MS/MS were used to determine earthworms' ingredients and differential metabolites. Swiss Target Prediction database was used for ingredients' target prediction and wound disease-relevant genes were found from GeneCards, OMIM, and DrugBank databases. Network pharmacology was conducted to demonstrate filtering hub targets, biological functions, and the signaling pathways of earthworms extract against wounds. Molecular docking and metabolism analysis were used to look for core target genes and key bioactive molecules from earthworms. Finally, the investigation shows 5 most important signal pathways, 5 core genes, and 6 bioactive ingredients-related cell-cell adhesion, cell proliferation, and cell migration processes could be affected by earthworms' extract. On 3rd day, the extract could regulate HIF1A and EGFR targets to make the differences of quantities of 4-pyridoxate, tetradecanoic acid, and L-kynurenine. While on 7th day, the regulation refers 6 earthworms' bioactive ingredients, 4 core genes (CTNNB1, EGFR, SRC, and CASP3), and 4 differential metabolites (4-hydoxy-2-quinolinecarboxylic acid, urocanate, deoxyinosine, creatine, and sn-glycerol-3-phosphocholine). on 14th day, 2 core genes (EGFR, SRC) are influenced in the biological processes. Briefly, we found that 6 ingredients from earthworms have most bioactive and 5 core genes play an important role in promoting wound-healing processes. These discovers indicates earthworms could against wound via AGE-RAGE, PI3K-Akt, HIF1A, MAPK, and Axon guidance pathways.
Collapse
Affiliation(s)
- Dong Wang
- Medical Research and Experiment Center, Shaanxi University of Chinese Medicine, Xianyang, China.
- Shaanxi Key Laboratory of Research on TCM Physical Constitution and Disease Prevention and Treatment, Xianyang, China.
| | - Zhen Ruan
- Xianyang Central Hospital, Xianyang, China
| | - Ruihui Wang
- Medical Research and Experiment Center, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Li Ma
- Medical Research and Experiment Center, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Saiqing Tang
- Second Clinical Medical School, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Xuejing Wang
- Medical Research and Experiment Center, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Axue Ma
- Second Clinical Medical School, Shaanxi University of Chinese Medicine, Xianyang, China
| |
Collapse
|
17
|
Suresh V, Bhattacharya B, Tshuva RY, Danan Gotthold M, Olender T, Bose M, Pradhan SJ, Zeev BB, Smith RS, Tole S, Galande S, Harwell CC, Baizabal JM, Reiner O. PRDM16 co-operates with LHX2 to shape the human brain. OXFORD OPEN NEUROSCIENCE 2024; 3:kvae001. [PMID: 38595939 PMCID: PMC10914218 DOI: 10.1093/oons/kvae001] [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: 08/12/2023] [Revised: 11/24/2023] [Accepted: 12/15/2023] [Indexed: 04/11/2024]
Abstract
PRDM16 is a dynamic transcriptional regulator of various stem cell niches, including adipocytic, hematopoietic, cardiac progenitors, and neural stem cells. PRDM16 has been suggested to contribute to 1p36 deletion syndrome, one of the most prevalent subtelomeric microdeletion syndromes. We report a patient with a de novo nonsense mutation in the PRDM16 coding sequence, accompanied by lissencephaly and microcephaly features. Human stem cells were genetically modified to mimic this mutation, generating cortical organoids that exhibited altered cell cycle dynamics. RNA sequencing of cortical organoids at day 32 unveiled changes in cell adhesion and WNT-signaling pathways. ChIP-seq of PRDM16 identified binding sites in postmortem human fetal cortex, indicating the conservation of PRDM16 binding to developmental genes in mice and humans, potentially at enhancer sites. A shared motif between PRDM16 and LHX2 was identified and further examined through comparison with LHX2 ChIP-seq data from mice. These results suggested a collaborative partnership between PRDM16 and LHX2 in regulating a common set of genes and pathways in cortical radial glia cells, possibly via their synergistic involvement in cortical development.
Collapse
Affiliation(s)
- Varun Suresh
- Department of Molecular Genetics, Weizmann Institute of Science, 234 Herzl St., Rehovot 7610001, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, 234 Herzl St., Rehovot 7610001, Israel
- Department of Biological Sciences, Tata Institute of Fundamental Research, Homi Bhabha Road, Navy Nagar, Colaba, Mumbai 400005, India
| | - Bidisha Bhattacharya
- Department of Molecular Genetics, Weizmann Institute of Science, 234 Herzl St., Rehovot 7610001, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, 234 Herzl St., Rehovot 7610001, Israel
| | - Rami Yair Tshuva
- Department of Molecular Genetics, Weizmann Institute of Science, 234 Herzl St., Rehovot 7610001, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, 234 Herzl St., Rehovot 7610001, Israel
| | - Miri Danan Gotthold
- Department of Molecular Genetics, Weizmann Institute of Science, 234 Herzl St., Rehovot 7610001, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, 234 Herzl St., Rehovot 7610001, Israel
| | - Tsviya Olender
- Department of Molecular Genetics, Weizmann Institute of Science, 234 Herzl St., Rehovot 7610001, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, 234 Herzl St., Rehovot 7610001, Israel
| | - Mahima Bose
- Department of Biological Sciences, Tata Institute of Fundamental Research, Homi Bhabha Road, Navy Nagar, Colaba, Mumbai 400005, India
| | - Saurabh J Pradhan
- Chromatin Biology and Epigenetics Laboratory, Biology Department, Indian Institute of Science Education and Research Pune, Dr. Homi Bhabha Road, Pune 411008, India
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna Biocenter, 3 Dr. Bohr-Gasse, 1030 Vienna, Austria
| | - Bruria Ben Zeev
- Edmond and Lily Safra Pediatric Hospital, Sheba Medical Center and Tel Aviv School of Medicine, Tel Aviv University, Ramat Aviv 69978, Israel
| | - Richard Scott Smith
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, 320 E. Superior St., Chicago, IL 60611, USA
| | - Shubha Tole
- Department of Biological Sciences, Tata Institute of Fundamental Research, Homi Bhabha Road, Navy Nagar, Colaba, Mumbai 400005, India
| | - Sanjeev Galande
- Chromatin Biology and Epigenetics Laboratory, Biology Department, Indian Institute of Science Education and Research Pune, Dr. Homi Bhabha Road, Pune 411008, India
- Department of Life Sciences, Center of Excellence in Epigenetics, Shiv Nadar University, Shiv Nadar IoE, Gautam Buddha Nagar, Uttar Pradesh - 201314, India
| | - Corey C Harwell
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, 35 Medical Center Way, San Francisco, CA 94143, USA
- Weill Institute for Neuroscience, 1651 4th St, San Francisco, CA94158, USA
- Department of Neurology, University of California, San Francisco, 505 Parnassus Ave, San Francisco, CA 94143, USA
| | - José-Manuel Baizabal
- Department of Biology, Indiana University, 1001 E 3rd St., Bloomington, IN 47405, USA
| | - Orly Reiner
- Department of Molecular Genetics, Weizmann Institute of Science, 234 Herzl St., Rehovot 7610001, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, 234 Herzl St., Rehovot 7610001, Israel
| |
Collapse
|
18
|
Dorsey SG, Mocci E, Lane MV, Krueger BK. Rapid effects of valproic acid on the fetal brain transcriptome: Implications for brain development and autism. RESEARCH SQUARE 2024:rs.3.rs-3684653. [PMID: 38260618 PMCID: PMC10802704 DOI: 10.21203/rs.3.rs-3684653/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
There is an increased incidence of autism among the children of women who take the anti-epileptic, mood-stabilizing drug, valproic acid (VPA) during pregnancy; moreover, exposure to VPA in utero causes autistic-like symptoms in rodents and non-human primates. Analysis of RNA-seq data obtained from E12.5 fetal mouse brains 3 hours after VPA administration to the pregnant dam revealed that VPA rapidly and significantly increased or decreased the expression of approximately 7,300 genes. No significant sex differences in VPA-induced gene expression were observed. Expression of 399 autism risk genes was significantly altered by VPA as was expression of 255 genes that have been reported to play fundamental roles in fetal brain development but are not otherwise linked to autism. Expression of genes associated with intracellular signaling pathways, neurogenesis, and excitation-inhibition balance as well as synaptogenesis, neuronal fate determination, axon and dendritic development, neuroinflammation, circadian rhythms, and epigenetic modulation of gene expression was dysregulated by VPA. The goal of this study was to identify mouse genes that are: (a) significantly up- or down-regulated by VPA in the fetal brain and (b) known to be associated with autism and/or to play a role in embryonic neurodevelopmental processes, perturbation of which has the potential to alter brain connectivity and, consequently behavior, in the adult. The set of genes meeting these criteria provides potential targets for future hypothesis-driven studies to elucidate the proximal causes of errors in brain connectivity underlying neurodevelopmental disorders such as autism.
Collapse
Affiliation(s)
- Susan G. Dorsey
- Department of Pain and Translational Symptom Sciences, University of Maryland School of Nursing, Baltimore, MD 21201
| | - Evelina Mocci
- Department of Pain and Translational Symptom Sciences, University of Maryland School of Nursing, Baltimore, MD 21201
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Malcolm V. Lane
- Translational Toxicology/Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Bruce K. Krueger
- Departments of Physiology and Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21201
| |
Collapse
|
19
|
Sun LWH, Asana Marican HT, Beh LK, Shen H. Imaging the radioprotective effect of amifostine in the developing brain using an apoptosis-reporting transgenic zebrafish. Int J Radiat Biol 2023; 100:433-444. [PMID: 37922446 DOI: 10.1080/09553002.2023.2280011] [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: 05/09/2023] [Accepted: 10/03/2023] [Indexed: 11/05/2023]
Abstract
PURPOSE Normal tissue radioprotectants alleviate radiation-induced damages and preserve critical organ functions. Investigating their efficacy in vivo remains challenging, especially in enclosed organs like the brain. An animal model that enables direct visualization of radiation-induced apoptosis while possessing the structural complexity of a vertebrate brain facilitates these studies in a precise and effective manner. MATERIALS AND METHODS We employed a secA5 transgenic zebrafish expressing secreted Annexin V fused with a yellow fluorescent protein to visualize radiation-induced apoptosis in vivo. We developed a semi-automated imaging method for standardized acquisition of apoptosis signals in batches of zebrafish larvae. Using these approaches, we studied the protective effect of amifostine (WR-2721) in the irradiated zebrafish larval brain. RESULTS Upon 2 Gy total-body 137Cs irradiation, increased apoptosis could be visualized at high resolution in the secA5 brain at 2, 24, and 48 hour post irradiation (hpi). Amifostine treatment (4 mM) during irradiation reduced apoptosis significantly at 24 hpi and preserved Wnt active cells in the larval brain. When the 2 Gy irradiation was delivered in combination with cisplatin treatment (0.1 mM), the radioprotective effect of amifostine was also observed. CONCLUSIONS Our study reveals the radioprotective effect of amifostine in the developing zebrafish larval brain, and highlights the utility of secA5 transgenic zebrafish as a novel system for investigating normal tissue radioprotectants in vivo.
Collapse
Affiliation(s)
- Lucas W H Sun
- Singapore Nuclear Research and Safety Initiative, National University of Singapore, Singapore
| | | | - Lih Khiang Beh
- Singapore Nuclear Research and Safety Initiative, National University of Singapore, Singapore
| | - Hongyuan Shen
- Singapore Nuclear Research and Safety Initiative, National University of Singapore, Singapore
| |
Collapse
|
20
|
Fasham J, Huebner AK, Liebmann L, Khalaf-Nazzal R, Maroofian R, Kryeziu N, Wortmann SB, Leslie JS, Ubeyratna N, Mancini GMS, van Slegtenhorst M, Wilke M, Haack TB, Shamseldin HE, Gleeson JG, Almuhaizea M, Dweikat I, Abu-Libdeh B, Daana M, Zaki MS, Wakeling MN, McGavin L, Turnpenny PD, Alkuraya FS, Houlden H, Schlattmann P, Kaila K, Crosby AH, Baple EL, Hübner CA. SLC4A10 mutation causes a neurological disorder associated with impaired GABAergic transmission. Brain 2023; 146:4547-4561. [PMID: 37459438 PMCID: PMC10629776 DOI: 10.1093/brain/awad235] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/19/2023] [Accepted: 06/06/2023] [Indexed: 11/09/2023] Open
Abstract
SLC4A10 is a plasma-membrane bound transporter that utilizes the Na+ gradient to drive cellular HCO3- uptake, thus mediating acid extrusion. In the mammalian brain, SLC4A10 is expressed in principal neurons and interneurons, as well as in epithelial cells of the choroid plexus, the organ regulating the production of CSF. Using next generation sequencing on samples from five unrelated families encompassing nine affected individuals, we show that biallelic SLC4A10 loss-of-function variants cause a clinically recognizable neurodevelopmental disorder in humans. The cardinal clinical features of the condition include hypotonia in infancy, delayed psychomotor development across all domains and intellectual impairment. Affected individuals commonly display traits associated with autistic spectrum disorder including anxiety, hyperactivity and stereotyped movements. In two cases isolated episodes of seizures were reported in the first few years of life, and a further affected child displayed bitemporal epileptogenic discharges on EEG without overt clinical seizures. While occipitofrontal circumference was reported to be normal at birth, progressive postnatal microcephaly evolved in 7 out of 10 affected individuals. Neuroradiological features included a relative preservation of brain volume compared to occipitofrontal circumference, characteristic narrow sometimes 'slit-like' lateral ventricles and corpus callosum abnormalities. Slc4a10 -/- mice, deficient for SLC4A10, also display small lateral brain ventricles and mild behavioural abnormalities including delayed habituation and alterations in the two-object novel object recognition task. Collapsed brain ventricles in both Slc4a10-/- mice and affected individuals suggest an important role of SLC4A10 in the production of the CSF. However, it is notable that despite diverse roles of the CSF in the developing and adult brain, the cortex of Slc4a10-/- mice appears grossly intact. Co-staining with synaptic markers revealed that in neurons, SLC4A10 localizes to inhibitory, but not excitatory, presynapses. These findings are supported by our functional studies, which show the release of the inhibitory neurotransmitter GABA is compromised in Slc4a10-/- mice, while the release of the excitatory neurotransmitter glutamate is preserved. Manipulation of intracellular pH partially rescues GABA release. Together our studies define a novel neurodevelopmental disorder associated with biallelic pathogenic variants in SLC4A10 and highlight the importance of further analyses of the consequences of SLC4A10 loss-of-function for brain development, synaptic transmission and network properties.
Collapse
Affiliation(s)
- James Fasham
- RILD Wellcome Wolfson Centre, University of Exeter Medical School, Royal Devon University Healthcare NHS Foundation Trust, Exeter EX2 5DW, UK
- Peninsula Clinical Genetics Service, Royal Devon University Healthcare NHS Foundation Trust, Exeter EX2 5DW, UK
| | - Antje K Huebner
- Institute of Human Genetics, Jena University Hospital, Friedrich Schiller Universität, 07747 Jena, Germany
| | - Lutz Liebmann
- Institute of Human Genetics, Jena University Hospital, Friedrich Schiller Universität, 07747 Jena, Germany
| | - Reham Khalaf-Nazzal
- Department of Biomedical Sciences, Faculty of Medicine, Arab American University of Palestine, Jenin, P227, Palestine
| | - Reza Maroofian
- Molecular and Clinical Sciences Institute, St. George’s University of London, London SW17 0RE, UK
| | - Nderim Kryeziu
- Institute of Human Genetics, Jena University Hospital, Friedrich Schiller Universität, 07747 Jena, Germany
| | - Saskia B Wortmann
- University Children’s Hospital, Salzburger Landeskliniken (SALK) and Paracelsus Medical University (PMU), 5020 Salzburg, Austria
- Amalia Children’s Hospital, Radboudumc, 6525 GA Nijmegen, The Netherlands
- Institute of Human Genetics, Technische Universität München, 80333 Munich, Germany
| | - Joseph S Leslie
- RILD Wellcome Wolfson Centre, University of Exeter Medical School, Royal Devon University Healthcare NHS Foundation Trust, Exeter EX2 5DW, UK
| | - Nishanka Ubeyratna
- RILD Wellcome Wolfson Centre, University of Exeter Medical School, Royal Devon University Healthcare NHS Foundation Trust, Exeter EX2 5DW, UK
| | - Grazia M S Mancini
- Department of Clinical Genetics, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands
| | | | - Martina Wilke
- Department of Clinical Genetics, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Tobias B Haack
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, 72076 Tübingen, Germany
| | - Hanan E Shamseldin
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh 11564, Saudi Arabia
| | - Joseph G Gleeson
- Rady Children’s Institute for Genomic Medicine, San Diego, CA 92123, USA
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Mohamed Almuhaizea
- Department of Neuroscience, King Faisal Specialist Hospital and Research Center, Riyadh 11564, Saudi Arabia
| | - Imad Dweikat
- Department of Biomedical Sciences, Faculty of Medicine, Arab American University of Palestine, Jenin, P227, Palestine
| | - Bassam Abu-Libdeh
- Department of Pediatrics and Genetics, Makassed Hospital and Al-Quds University, East Jerusalem, 95908, Palestine
| | - Muhannad Daana
- Department of Pediatrics, Arab Women’s Union Hospital, Nablus, P400, Palestine
| | - Maha S Zaki
- Clinical Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Dokki, Cairo 12622, Egypt
| | - Matthew N Wakeling
- RILD Wellcome Wolfson Centre, University of Exeter Medical School, Royal Devon University Healthcare NHS Foundation Trust, Exeter EX2 5DW, UK
| | - Lucy McGavin
- Department of Radiology, Derriford Hospital, Plymouth PL6 8DH, UK
| | - Peter D Turnpenny
- RILD Wellcome Wolfson Centre, University of Exeter Medical School, Royal Devon University Healthcare NHS Foundation Trust, Exeter EX2 5DW, UK
- Peninsula Clinical Genetics Service, Royal Devon University Healthcare NHS Foundation Trust, Exeter EX2 5DW, UK
| | - Fowzan S Alkuraya
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh 11564, Saudi Arabia
| | - Henry Houlden
- Molecular and Clinical Sciences Institute, St. George’s University of London, London SW17 0RE, UK
| | - Peter Schlattmann
- Institute for Medical Statistics, Computer Science and Data Science, Jena University Hospital, 07747 Jena, Germany
| | - Kai Kaila
- Molecular and Integrative Biosciences, University of Helsinki, 00014 Helsinki, Finland
- Neuroscience Center, Helsinki Institute of Life Science, University of Helsinki, 00014 Helsinki, Finland
| | - Andrew H Crosby
- RILD Wellcome Wolfson Centre, University of Exeter Medical School, Royal Devon University Healthcare NHS Foundation Trust, Exeter EX2 5DW, UK
| | - Emma L Baple
- RILD Wellcome Wolfson Centre, University of Exeter Medical School, Royal Devon University Healthcare NHS Foundation Trust, Exeter EX2 5DW, UK
- Peninsula Clinical Genetics Service, Royal Devon University Healthcare NHS Foundation Trust, Exeter EX2 5DW, UK
| | - Christian A Hübner
- Institute of Human Genetics, Jena University Hospital, Friedrich Schiller Universität, 07747 Jena, Germany
- Center for Rare Diseases, Jena University Hospital, Friedrich Schiller Universität, 07747 Jena, Germany
| |
Collapse
|
21
|
Zhang B, Zhao C, Shen W, Li W, Zheng Y, Kong X, Wang J, Wu X, Zeng T, Liu Y, Zhou Y. KDM2B regulates hippocampal morphogenesis by transcriptionally silencing Wnt signaling in neural progenitors. Nat Commun 2023; 14:6489. [PMID: 37838801 PMCID: PMC10576813 DOI: 10.1038/s41467-023-42322-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 10/06/2023] [Indexed: 10/16/2023] Open
Abstract
The hippocampus plays major roles in learning and memory, and its formation requires precise coordination of patterning, cell proliferation, differentiation, and migration. Here we removed the chromatin-association capability of KDM2B in the progenitors of developing dorsal telencephalon (Kdm2b∆CxxC) to discover that Kdm2b∆CxxC hippocampus, particularly the dentate gyrus, became drastically smaller with disorganized cellular components and structure. Kdm2b∆CxxC mice display prominent defects in spatial memory, motor learning and fear conditioning, resembling patients with KDM2B mutations. The migration and differentiation of neural progenitor cells is greatly impeded in the developing Kdm2b∆CxxC hippocampus. Mechanism studies reveal that Wnt signaling genes in developing Kdm2b∆CxxC hippocampi are de-repressed due to reduced enrichment of repressive histone marks by polycomb repressive complexes. Activating the Wnt signaling disturbs hippocampal neurogenesis, recapitulating the effect of KDM2B loss. Together, we unveil a previously unappreciated gene repressive program mediated by KDM2B that controls progressive fate specifications and cell migration, hence morphogenesis of the hippocampus.
Collapse
Affiliation(s)
- Bo Zhang
- Department of Neurosurgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Chen Zhao
- Department of Neurosurgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Wenchen Shen
- Department of Neurosurgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Wei Li
- Department of Neurosurgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Yue Zheng
- Department of Neurosurgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Xiangfei Kong
- Department of Neurosurgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Junbao Wang
- Department of Neurosurgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Xudong Wu
- Department of Cell Biology, Tianjin Medical University, Tianjin, China
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Tao Zeng
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China.
| | - Ying Liu
- Department of Neurosurgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China.
| | - Yan Zhou
- Department of Neurosurgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China.
| |
Collapse
|
22
|
Morey R, Zheng Y, Sun D, Garrett M, Gasperi M, Maihofer A, Baird CL, Grasby K, Huggins A, Haswell C, Thompson P, Medland S, Gustavson D, Panizzon M, Kremen W, Nievergelt C, Ashley-Koch A, Logue L. Genomic Structural Equation Modeling Reveals Latent Phenotypes in the Human Cortex with Distinct Genetic Architecture. RESEARCH SQUARE 2023:rs.3.rs-3253035. [PMID: 37886496 PMCID: PMC10602057 DOI: 10.21203/rs.3.rs-3253035/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
Genetic contributions to human cortical structure manifest pervasive pleiotropy. This pleiotropy may be harnessed to identify unique genetically-informed parcellations of the cortex that are neurobiologically distinct from functional, cytoarchitectural, or other cortical parcellation schemes. We investigated genetic pleiotropy by applying genomic structural equation modeling (SEM) to map the genetic architecture of cortical surface area (SA) and cortical thickness (CT) for the 34 brain regions recently reported in the ENIGMA cortical GWAS. Genomic SEM uses the empirical genetic covariance estimated from GWAS summary statistics with LD score regression (LDSC) to discover factors underlying genetic covariance, which we are denoting genetically informed brain networks (GIBNs). Genomic SEM can fit a multivariate GWAS from summary statistics for each of the GIBNs, which can subsequently be used for LD score regression (LDSC). We found the best-fitting model of cortical SA identified 6 GIBNs and CT identified 4 GIBNs. The multivariate GWASs of these GIBNs identified 74 genome-wide significant (GWS) loci (p<5×10-8), including many previously implicated in neuroimaging phenotypes, behavioral traits, and psychiatric conditions. LDSC of GIBN GWASs found that SA-derived GIBNs had a positive genetic correlation with bipolar disorder (BPD), and cannabis use disorder, indicating genetic predisposition to a larger SA in the specific GIBN is associated with greater genetic risk of these disorders. A negative genetic correlation was observed with attention deficit hyperactivity disorder (ADHD), major depressive disorder (MDD), and insomnia, indicating genetic predisposition to a larger SA in the specific GIBN is associated with lower genetic risk of these disorders. CT GIBNs displayed a negative genetic correlation with alcohol dependence. Jointly modeling the genetic architecture of complex traits and investigating multivariate genetic links across phenotypes offers a new vantage point for mapping the cortex into genetically informed networks.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Paul Thompson
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging & Informatics Institute, Keck School of Medicine of the University of Southern California, Marina del Rey, California, USA
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Suresh V, Bhattacharya B, Tshuva RY, Danan Gotthold M, Olender T, Bose M, Pradhan SJ, Ben Zeev B, Smith RS, Tole S, Galande S, Harwell C, Baizabal JM, Reiner O. PRDM16 co-operates with LHX2 to shape the human brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.12.553065. [PMID: 37609127 PMCID: PMC10441425 DOI: 10.1101/2023.08.12.553065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
PRDM16 is a dynamic transcriptional regulator of various stem cell niches, including adipocytic, hematopoietic, cardiac progenitors, and neural stem cells. PRDM16 has been suggested to contribute to 1p36 deletion syndrome, one of the most prevalent subtelomeric microdeletion syndromes. We report a patient with a de novo nonsense mutation in the PRDM16 coding sequence, accompanied by lissencephaly and microcephaly features. Human stem cells were genetically modified to mimic this mutation, generating cortical organoids that exhibited altered cell cycle dynamics. RNA sequencing of cortical organoids at day 32 unveiled changes in cell adhesion and WNT-signaling pathways. ChIP-seq of PRDM16 identified binding sites in postmortem human fetal cortex, indicating the conservation of PRDM16 binding to developmental genes in mice and humans, potentially at enhancer sites. A shared motif between PRDM16 and LHX2 was identified and further examined through comparison with LHX2 ChIP-seq data from mice. These results suggested a collaborative partnership between PRDM16 and LHX2 in regulating a common set of genes and pathways in cortical radial glia cells, possibly via their synergistic involvement in cortical development.
Collapse
|
24
|
Da Silva F, Niehrs C. Multimodal Wnt signalling in the mouse neocortex. Cells Dev 2023; 174:203838. [PMID: 37060946 DOI: 10.1016/j.cdev.2023.203838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/30/2023] [Accepted: 04/08/2023] [Indexed: 04/17/2023]
Abstract
The neocortex is the site of higher cognitive functions and its development is tightly regulated by cell signalling pathways. Wnt signalling is inexorably linked with neocortex development but its precise role remains unclear. Most studies demonstrate that Wnt/β-catenin regulates neural progenitor self-renewal but others suggest it can also promote differentiation. Wnt/STOP signalling is a novel branch of the Wnt pathway that stabilizes proteins during G2/M by inhibiting glycogen synthase kinase 3 (GSK3)-mediated protein degradation. Recent data from Da Silva et al. (2021) demonstrate that Wnt/STOP is involved in neocortex development where, by stabilizing the neurogenic transcription factors Sox4 and Sox11, it promotes neural progenitor differentiation. The authors also show that Wnt/STOP regulates asymmetric cell division and cell cycle dynamics in apical and basal progenitors, respectively. This study reveals a division of labour in the Wnt signalling pathway by suggesting that Wnt/STOP is the primary driver of cortical neurogenesis while Wnt/β-catenin is mainly responsible for self-renewal. These results resolve a decades-old question on the role of Wnt signalling in cortical neural progenitors.
Collapse
Affiliation(s)
- Fabio Da Silva
- Division of Molecular Embryology, DKFZ-ZMBH Alliance, Deutsches Krebsforschungszentrum (DKFZ), 69120 Heidelberg, Germany
| | - Christof Niehrs
- Division of Molecular Embryology, DKFZ-ZMBH Alliance, Deutsches Krebsforschungszentrum (DKFZ), 69120 Heidelberg, Germany; Institute of Molecular Biology (IMB), 55128 Mainz, Germany.
| |
Collapse
|
25
|
Sun XL, Chen ZH, Guo X, Wang J, Ge M, Wong SZH, Wang T, Li S, Yao M, Johnston LA, Wu QF. Stem cell competition driven by the Axin2-p53 axis controls brain size during murine development. Dev Cell 2023; 58:744-759.e11. [PMID: 37054704 DOI: 10.1016/j.devcel.2023.03.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 01/08/2023] [Accepted: 03/20/2023] [Indexed: 04/15/2023]
Abstract
Cell competition acts as a quality-control mechanism that eliminates cells less fit than their neighbors to optimize organ development. Whether and how competitive interactions occur between neural progenitor cells (NPCs) in the developing brain remains unknown. Here, we show that endogenous cell competition occurs and intrinsically correlates with the Axin2 expression level during normal brain development. Induction of genetic mosaicism predisposes Axin2-deficient NPCs to behave as "losers" in mice and undergo apoptotic elimination, but homogeneous ablation of Axin2 does not promote cell death. Mechanistically, Axin2 suppresses the p53 signaling pathway at the post-transcriptional level to maintain cell fitness, and Axin2-deficient cell elimination requires p53-dependent signaling. Furthermore, mosaic Trp53 deletion confers a "winner" status to p53-deficient cells that outcompete their neighbors. Conditional loss of both Axin2 and Trp53 increases cortical area and thickness, suggesting that the Axin2-p53 axis may coordinate to survey cell fitness, regulate natural cell competition, and optimize brain size during neurodevelopment.
Collapse
Affiliation(s)
- Xue-Lian Sun
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100101, China
| | - Zhen-Hua Chen
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100101, China
| | - Xize Guo
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100101, China
| | - Jingjing Wang
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Mengmeng Ge
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Samuel Zheng Hao Wong
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ting Wang
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100101, China
| | - Si Li
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100101, China
| | - Mingze Yao
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, China
| | - Laura A Johnston
- Department of Genetics and Development, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Qing-Feng Wu
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100101, China; Beijing Children's Hospital, Capital Medical University, Beijing 100045, China; Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
26
|
Inada H, Corales LG, Osumi N. A novel feature of the ancient organ: A possible involvement of the subcommissural organ in neurogenic/gliogenic potential in the adult brain. Front Neurosci 2023; 17:1141913. [PMID: 36960167 PMCID: PMC10027738 DOI: 10.3389/fnins.2023.1141913] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 02/20/2023] [Indexed: 03/09/2023] Open
Abstract
The subcommissural organ (SCO) is a circumventricular organ highly conserved in vertebrates from Cyclostomata such as lamprey to mammals including human. The SCO locates in the boundary between the third ventricle and the entrance of the aqueduct of Sylvius. The SCO functions as a secretory organ producing a variety of proteins such as SCO-spondin, transthyretin, and basic fibroblast growth factor (FGF) into the cerebrospinal fluid (CSF). A significant contribution of the SCO has been thought to maintain the homeostasis of CSF dynamics. However, evidence has shown a possible role of SCO on neurogenesis in the adult brain. This review highlights specific features of the SCO related to adult neurogenesis, suggested by the progress of understanding SCO functions. We begin with a brief history of the SCO discovery and continue to structural features, gene expression, and a possible role in adult neurogenesis suggested by the SCO transplant experiment.
Collapse
Affiliation(s)
- Hitoshi Inada
- Laboratory of Health and Sports Sciences, Division of Biomedical Engineering for Health and Welfare, Graduate School of Biomedical Engineering, Tohoku University, Sendai, Japan
- Department of Developmental Neuroscience, Graduate School of Medicine, Tohoku University, Sendai, Japan
- *Correspondence: Hitoshi Inada,
| | - Laarni Grace Corales
- Department of Developmental Neuroscience, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Noriko Osumi
- Department of Developmental Neuroscience, Graduate School of Medicine, Tohoku University, Sendai, Japan
| |
Collapse
|
27
|
Yeh H, Woodbury ME, Ingraham Dixie KL, Ikezu T, Ikezu S. Microglial WNT5A supports dendritic spines maturation and neuronal firing. Brain Behav Immun 2023; 107:403-413. [PMID: 36395958 PMCID: PMC10588768 DOI: 10.1016/j.bbi.2022.11.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 10/13/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022] Open
Abstract
There is increasing evidence showing that microglia play a critical role in mediating synapse formation and spine growth, although the molecular mechanism remains elusive. Here, we demonstrate that the secreted morphogen WNT family member 5A (WNT5A) is the most abundant WNT expressed in microglia and that it promotes neuronal maturation. Co-culture of microglia with Thy1-YFP+ differentiated neurons significantly increased neuronal spine density and reduced dendritic spine turnover rate, which was diminished by silencing microglial Wnt5a in vitro. Co-cultured microglia increased post-synaptic marker PSD95 and synaptic density as determined by the co-localization of PSD95 with pre-synaptic marker VGLUT2 in vitro. The silencing of Wnt5a expression in microglia partially reduced both PSD95 and synaptic densities. Co-culture of differentiated neurons with microglia significantly enhanced neuronal firing rate as measured by multiple electrode array, which was significantly reduced by silencing microglial Wnt5a at 23 days differentiation in vitro. These findings demonstrate that microglia can mediate spine maturation and regulate neuronal excitability via WNT5A secretion indicating possible pathological roles of dysfunctional microglia in developmental disorders.
Collapse
Affiliation(s)
- Hana Yeh
- Graduate Program in Neuroscience, Boston University, United States; Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
| | - Maya E Woodbury
- Graduate Program in Neuroscience, Boston University, United States; Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
| | - Kaitlin L Ingraham Dixie
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States; Center for Education Innovation and Learning in the Sciences, University of California, Los Angeles, CA, United States
| | - Tsuneya Ikezu
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States; Department of Neuroscience, Molecular Neurotherapeutics Laboratory, Mayo Clinic, Jacksonville, FL, United States.
| | - Seiko Ikezu
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States; Department of Neuroscience, Molecular Neurotherapeutics Laboratory, Mayo Clinic, Jacksonville, FL, United States.
| |
Collapse
|
28
|
Geng S, Paul F, Kowalczyk I, Raimundo S, Sporbert A, Mamo TM, Hammes A. Balancing WNT signalling in early forebrain development: The role of LRP4 as a modulator of LRP6 function. Front Cell Dev Biol 2023; 11:1173688. [PMID: 37091972 PMCID: PMC10119419 DOI: 10.3389/fcell.2023.1173688] [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: 02/24/2023] [Accepted: 03/28/2023] [Indexed: 04/25/2023] Open
Abstract
The specification of the forebrain relies on the precise regulation of WNT/ß-catenin signalling to support neuronal progenitor cell expansion, patterning, and morphogenesis. Imbalances in WNT signalling activity in the early neuroepithelium lead to congenital disorders, such as neural tube defects (NTDs). LDL receptor-related protein (LRP) family members, including the well-studied receptors LRP5 and LRP6, play critical roles in modulating WNT signalling capacity through tightly regulated interactions with their co-receptor Frizzled, WNT ligands, inhibitors and intracellular WNT pathway components. However, little is known about the function of LRP4 as a potential modulator of WNT signalling in the central nervous system. In this study, we investigated the role of LRP4 in the regulation of WNT signalling during early mouse forebrain development. Our results demonstrate that LRP4 can modulate LRP5- and LRP6-mediated WNT signalling in the developing forebrain prior to the onset of neurogenesis at embryonic stage 9.5 and is therefore essential for accurate neural tube morphogenesis. Specifically, LRP4 functions as a genetic modifier for impaired mitotic activity and forebrain hypoplasia, but not for NTDs in LRP6-deficient mutants. In vivo and in vitro data provide evidence that LRP4 is a key player in fine-tuning WNT signalling capacity and mitotic activity of mouse neuronal progenitors and of human retinal pigment epithelial (hTERT RPE-1) cells. Our data demonstrate the crucial roles of LRP4 and LRP6 in regulating WNT signalling and forebrain development and highlight the need to consider the interaction between different signalling pathways to understand the underlying mechanisms of disease. The findings have significant implications for our mechanistic understanding of how LRPs participate in controlling WNT signalling.
Collapse
Affiliation(s)
- Shuang Geng
- Neuroscience, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Institute for Biology, Free University of Berlin, Berlin, Germany
| | - Fabian Paul
- Neuroscience, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Institute for Biology, Free University of Berlin, Berlin, Germany
| | - Izabela Kowalczyk
- Neuroscience, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Institute for Biology, Free University of Berlin, Berlin, Germany
| | - Sandra Raimundo
- Advanced Light Microscopy Technology Platform, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Anje Sporbert
- Advanced Light Microscopy Technology Platform, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Tamrat Meshka Mamo
- Neuroscience, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- *Correspondence: Tamrat Meshka Mamo, ; Annette Hammes,
| | - Annette Hammes
- Neuroscience, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- *Correspondence: Tamrat Meshka Mamo, ; Annette Hammes,
| |
Collapse
|
29
|
Zang Z, Yin H, Du Z, Xie R, Yang L, Cai Y, Wang L, Zhang D, Li X, Liu T, Gong H, Gao J, Yang H, Warner M, Gustafsson JA, Xu H, Fan X. Valproic acid exposure decreases neurogenic potential of outer radial glia in human brain organoids. Front Mol Neurosci 2022; 15:1023765. [PMID: 36523605 PMCID: PMC9744776 DOI: 10.3389/fnmol.2022.1023765] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 11/08/2022] [Indexed: 07/29/2023] Open
Abstract
Valproic acid (VPA) exposure during pregnancy leads to a higher risk of autism spectrum disorder (ASD) susceptibility in offspring. Human dorsal forebrain organoids were used to recapitulate course of cortical neurogenesis in the developing human brain. Combining morphological characterization with massive parallel RNA sequencing (RNA-seq) on organoids to analyze the pathogenic effects caused by VPA exposure and critical signaling pathway. We found that VPA exposure in organoids caused a reduction in the size and impairment in the proliferation and expansion of neural progenitor cells (NPCs) in a dose-dependent manner. VPA exposure typically decreased the production of outer radial glia-like cells (oRGs), a subtype of NPCs contributing to mammalian neocortical expansion and delayed their fate toward upper-layer neurons. Transcriptomics analysis revealed that VPA exposure influenced ASD risk gene expression in organoids, which markedly overlapped with irregulated genes in brains or organoids originating from ASD patients. We also identified that VPA-mediated Wnt/β-catenin signaling pathway activation is essential for sustaining cortical neurogenesis and oRGs output. Taken together, our study establishes the use of dorsal forebrain organoids as an effective platform for modeling VPA-induced teratogenic pathways involved in the cortical neurogenesis and oRGs output, which might contribute to ASD pathogenesis in the developing brain.
Collapse
Affiliation(s)
- Zhenle Zang
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Huachun Yin
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Zhulin Du
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Ruxin Xie
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Ling Yang
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yun Cai
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Liuyongwei Wang
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Dandan Zhang
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xin Li
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Tianyao Liu
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hong Gong
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Junwei Gao
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hui Yang
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Margaret Warner
- Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX, United States
| | - Jan-Ake Gustafsson
- Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX, United States
- Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden
| | - Haiwei Xu
- Southwest Hospital and Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xiaotang Fan
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
30
|
Sun H, Wu M, Wang M, Zhang X, Zhu J. The regulatory role of endoplasmic reticulum chaperone proteins in neurodevelopment. Front Neurosci 2022; 16:1032607. [DOI: 10.3389/fnins.2022.1032607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/25/2022] [Indexed: 11/16/2022] Open
Abstract
The endoplasmic reticulum (ER) is the largest tubular reticular organelle spanning the cell. As the main site of protein synthesis, Ca2+ homeostasis maintenance and lipid metabolism, the ER plays a variety of essential roles in eukaryotic cells, with ER molecular chaperones participate in all these processes. In recent years, it has been reported that the abnormal expression of ER chaperones often leads to a variety of neurodevelopmental disorders (NDDs), including abnormal neuronal migration, neuronal morphogenesis, and synaptic function. Neuronal development is a complex and precisely regulated process. Currently, the mechanism by which neural development is regulated at the ER level remains under investigation. Therefore, in this work, we reviewed the recent advances in the roles of ER chaperones in neural development and developmental disorders caused by the deficiency of these molecular chaperones.
Collapse
|
31
|
Alkailani MI, Aittaleb M, Tissir F. WNT signaling at the intersection between neurogenesis and brain tumorigenesis. Front Mol Neurosci 2022; 15:1017568. [PMID: 36267699 PMCID: PMC9577257 DOI: 10.3389/fnmol.2022.1017568] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 09/13/2022] [Indexed: 11/23/2022] Open
Abstract
Neurogenesis and tumorigenesis share signaling molecules/pathways involved in cell proliferation, differentiation, migration, and death. Self-renewal of neural stem cells is a tightly regulated process that secures the accuracy of cell division and eliminates cells that undergo mitotic errors. Abnormalities in the molecular mechanisms controlling this process can trigger aneuploidy and genome instability, leading to neoplastic transformation. Mutations that affect cell adhesion, polarity, or migration enhance the invasive potential and favor the progression of tumors. Here, we review recent evidence of the WNT pathway’s involvement in both neurogenesis and tumorigenesis and discuss the experimental progress on therapeutic opportunities targeting components of this pathway.
Collapse
Affiliation(s)
- Maisa I. Alkailani
- College of Health and Life Sciences, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Mohamed Aittaleb
- College of Health and Life Sciences, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Fadel Tissir
- College of Health and Life Sciences, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
- Institute of Neuroscience, Université catholique de Louvain, Brussels, Belgium
- *Correspondence: Fadel Tissir,
| |
Collapse
|
32
|
Huang L, Liu M, Jiang W, Ding H, Han Y, Wen M, Li Y, Liu X, Zeng H. Bradykinin/bradykinin 1 receptor promotes brain microvascular endothelial cell permeability and proinflammatory cytokine release by downregulating Wnt3a. J Biochem Mol Toxicol 2022; 36:e23213. [PMID: 36111657 PMCID: PMC10078380 DOI: 10.1002/jbt.23213] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 06/29/2022] [Accepted: 08/30/2022] [Indexed: 11/10/2022]
Abstract
Stroke is a life-threatening disease with limited therapeutic options. Damage to the blood-brain barrier (BBB) is the key pathological feature of ischemic stroke. This study explored the role of the bradykinin (BK)/bradykinin 1 receptor (B1R) and its mechanism of action in the BBB. Human brain microvascular endothelial cells (BMECs) were used to test for cellular responses to BK by using the Cell Counting Kit-8 assay, 5-ethynyl-2'-deoxyuridine staining, enzyme-linked immunosorbent assay, flow cytometry, immunofluorescence, cellular permeability assays, and western blotting to evaluate cell viability, cytokine production, and reactive oxygen species (ROS) levels in vitro. A BBB induced by middle cerebral artery occlusion was used to evaluate BBB injuries, and the role played by BK/B1R in ischemic/reperfusion (I/R) was explored in a rat model. Results showed that BK reduced the viability of BMECs and increased the levels of proinflammatory cytokines (interleukin 6 [IL-6], IL-18, and monocyte chemoattractant protein-1) and ROS. Additionally, cellular permeability was increased by BK treatment, and the expression of tight junction proteins (claudin-5 and occludin) was decreased. Interestingly, Wnt3a expression was inhibited by BK and exogenous Wnt3a restored the effects of BK on BMECs. In an in vivo I/R rat model, knockdown of B1R significantly decreased infarct volume and inflammation in I/R rats. Our results suggest that BK might be a key inducer of BBB injury and B1R knockdown might provide a beneficial effect by upregulating Wnt3a.
Collapse
Affiliation(s)
- Linqiang Huang
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital Guangdong Academy of Medical Sciences Guangzhou Guangdong China
| | - Mengting Liu
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital Guangdong Academy of Medical Sciences Guangzhou Guangdong China
- Clinical Medical Division, The Second School of Clinical Medicine Southern Medical University Guangzhou China
| | - Wenqiang Jiang
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital Guangdong Academy of Medical Sciences Guangzhou Guangdong China
| | - Hongguang Ding
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital Guangdong Academy of Medical Sciences Guangzhou Guangdong China
| | - Yongli Han
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital Guangdong Academy of Medical Sciences Guangzhou Guangdong China
| | - Miaoyun Wen
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital Guangdong Academy of Medical Sciences Guangzhou Guangdong China
| | - Ya Li
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital Guangdong Academy of Medical Sciences Guangzhou Guangdong China
- Clinical Medical Division, School of Medicine South China University of Technology Guangzhou China
| | - Xiaoyu Liu
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital Guangdong Academy of Medical Sciences Guangzhou Guangdong China
- Clinical Medical Division, The Second School of Clinical Medicine Southern Medical University Guangzhou China
| | - Hongke Zeng
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital Guangdong Academy of Medical Sciences Guangzhou Guangdong China
| |
Collapse
|
33
|
Wang J, Wang A, Tian K, Hua X, Zhang B, Zheng Y, Kong X, Li W, Xu L, Wang J, Li Z, Liu Y, Zhou Y. A Ctnnb1 enhancer regulates neocortical neurogenesis by controlling the abundance of intermediate progenitors. Cell Discov 2022; 8:74. [PMID: 35915089 PMCID: PMC9343459 DOI: 10.1038/s41421-022-00421-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 05/05/2022] [Indexed: 11/09/2022] Open
Abstract
β-catenin-dependent canonical Wnt signaling plays a plethora of roles in neocortex (Ncx) development, but its function in regulating the abundance of intermediate progenitors (IPs) is elusive. Here we identified neCtnnb1, an evolutionarily conserved cis-regulatory element with typical enhancer features in developing Ncx. neCtnnb1 locates 55 kilobase upstream of and spatially close to the promoter of Ctnnb1, the gene encoding β-catenin. CRISPR/Cas9-mediated activation or interference of the neCtnnb1 locus enhanced or inhibited transcription of Ctnnb1. neCtnnb1 drove transcription predominantly in the subventricular zone of developing Ncx. Knock-out of neCtnnb1 in mice resulted in compromised expression of Ctnnb1 and the Wnt reporter in developing Ncx. Importantly, knock-out of neCtnnb1 lead to reduced production and transit-amplification of IPs, which subsequently generated fewer upper-layer Ncx projection neurons (PNs). In contrast, enhancing the canonical Wnt signaling by stabilizing β-catenin in neCtnnb1-active cells promoted the production of IPs and upper-layer Ncx PNs. ASH2L was identified as the key trans-acting factor that associates with neCtnnb1 and Ctnnb1’s promoter to maintain Ctnnb1’s transcription in both mouse and human Ncx progenitors. These findings advance understanding of transcriptional regulation of Ctnnb1, and provide insights into mechanisms underlying Ncx expansion during development.
Collapse
Affiliation(s)
- Junbao Wang
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University; Frontier Science Center for Immunology and Metabolism, Medical Research Institute at School of Medicine; The RNA Institute, College of Life Sciences; Wuhan University, Wuhan, Hubei, China
| | - Andi Wang
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University; Frontier Science Center for Immunology and Metabolism, Medical Research Institute at School of Medicine; The RNA Institute, College of Life Sciences; Wuhan University, Wuhan, Hubei, China
| | - Kuan Tian
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University; Frontier Science Center for Immunology and Metabolism, Medical Research Institute at School of Medicine; The RNA Institute, College of Life Sciences; Wuhan University, Wuhan, Hubei, China
| | - Xiaojiao Hua
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University; Frontier Science Center for Immunology and Metabolism, Medical Research Institute at School of Medicine; The RNA Institute, College of Life Sciences; Wuhan University, Wuhan, Hubei, China
| | - Bo Zhang
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University; Frontier Science Center for Immunology and Metabolism, Medical Research Institute at School of Medicine; The RNA Institute, College of Life Sciences; Wuhan University, Wuhan, Hubei, China
| | - Yue Zheng
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University; Frontier Science Center for Immunology and Metabolism, Medical Research Institute at School of Medicine; The RNA Institute, College of Life Sciences; Wuhan University, Wuhan, Hubei, China
| | - Xiangfei Kong
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University; Frontier Science Center for Immunology and Metabolism, Medical Research Institute at School of Medicine; The RNA Institute, College of Life Sciences; Wuhan University, Wuhan, Hubei, China
| | - Wei Li
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University; Frontier Science Center for Immunology and Metabolism, Medical Research Institute at School of Medicine; The RNA Institute, College of Life Sciences; Wuhan University, Wuhan, Hubei, China
| | - Lichao Xu
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University; Frontier Science Center for Immunology and Metabolism, Medical Research Institute at School of Medicine; The RNA Institute, College of Life Sciences; Wuhan University, Wuhan, Hubei, China
| | - Juan Wang
- Department of Neurology, Wuhan Central Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhiqiang Li
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University; Frontier Science Center for Immunology and Metabolism, Medical Research Institute at School of Medicine; The RNA Institute, College of Life Sciences; Wuhan University, Wuhan, Hubei, China
| | - Ying Liu
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University; Frontier Science Center for Immunology and Metabolism, Medical Research Institute at School of Medicine; The RNA Institute, College of Life Sciences; Wuhan University, Wuhan, Hubei, China.
| | - Yan Zhou
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University; Frontier Science Center for Immunology and Metabolism, Medical Research Institute at School of Medicine; The RNA Institute, College of Life Sciences; Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
34
|
Abstract
While intraventricular hemorrhage (IVH) predominantly damages the periventricular white matter, it induces substantial injury to the cerebral gray matter. IVH destroys the germinal matrix, suppresses neurogenesis, and disrupts corticogenesis, thereby reducing the number of neurons in the upper cortical layer and volume of the cerebral gray matter. The pathogenesis of gray matter injury is attributed to IVH-induced oxidative stress, inflammation, and mass effect damaging the germinal matrix as well as to post-hemorrhagic ventricular dilation (PHVD). The IVH-induced cerebral gray matter injury and PHVD contribute to cognitive deficits and neurobehavioral disorders. Neuroimaging has enhanced our understanding of cerebral gray matter injury and is a valuable predictor of neurodevelopmental outcomes. Evidence from therapies tested in preclinical models and clinical trials suggests that strategies to promote neurogenesis, reduce cerebral inflammation and oxidative stress, and remove blood clots from the ventricles might enhance the outcome of these infants. This review offers an integrated view of new insights into the mechanisms underlying gray matter injury in premature infants with IVH and highlights the imminent therapies to restore neurodevelopmental dysfunction in IVH survivors.
Collapse
Affiliation(s)
- Deep Sharma
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY
| | - Alex Agyemang
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY
| | - Praveen Ballabh
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY; Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
35
|
Reis L, Raciti M, Rodriguez PG, Joseph B, Al Rayyes I, Uhlén P, Falk A, da Cunha Lima ST, Ceccatelli S. Glyphosate-based herbicide induces long-lasting impairment in neuronal and glial differentiation. ENVIRONMENTAL TOXICOLOGY 2022; 37:2044-2057. [PMID: 35485992 PMCID: PMC9541419 DOI: 10.1002/tox.23549] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 04/14/2022] [Accepted: 04/16/2022] [Indexed: 05/09/2023]
Abstract
Glyphosate-based herbicides (GBH) are among the most sold pesticides in the world. There are several formulations based on the active ingredient glyphosate (GLY) used along with other chemicals to improve the absorption and penetration in plants. The final composition of commercial GBH may modify GLY toxicological profile, potentially enhancing its neurotoxic properties. The developing nervous system is particularly susceptible to insults occurring during the early phases of development, and exposure to chemicals in this period may lead to persistent impairments on neurogenesis and differentiation. The aim of this study was to evaluate the long-lasting effects of a sub-cytotoxic concentration, 2.5 parts per million of GBH and GLY, on the differentiation of human neuroepithelial stem cells (NES) derived from induced pluripotent stem cells (iPSC). We treated NES cells with each compound and evaluated the effects on key cellular processes, such as proliferation and differentiation in daughter cells never directly exposed to the toxicants. We found that GBH induced a more immature neuronal profile associated to increased PAX6, NESTIN and DCX expression, and a shift in the differentiation process toward glial cell fate at the expense of mature neurons, as shown by an increase in the glial markers GFAP, GLT1, GLAST and a decrease in MAP2. Such alterations were associated to dysregulation of key genes critically involved in neurogenesis, including PAX6, HES1, HES5, and DDK1. Altogether, the data indicate that subtoxic concentrations of GBH, but not of GLY, induce long-lasting impairments on the differentiation potential of NES cells.
Collapse
Affiliation(s)
- Luã Reis
- Department of NeuroscienceKarolinska InstitutetStockholmSweden
| | - Marilena Raciti
- Department of NeuroscienceKarolinska InstitutetStockholmSweden
| | | | - Bertrand Joseph
- Institute of Environmental MedicineKarolinska InstitutetStockholmSweden
| | - Ibrahim Al Rayyes
- Department of Medical Biochemistry and BiophysicsKarolinska InstitutetStockholmSweden
| | - Per Uhlén
- Department of Medical Biochemistry and BiophysicsKarolinska InstitutetStockholmSweden
| | - Anna Falk
- Department of NeuroscienceKarolinska InstitutetStockholmSweden
| | - Suzana Telles da Cunha Lima
- Laboratório de Bioprospecção e Biotecnologia, Instituto de BiologiaUniversidade Federal da Bahia (UFBA)SalvadorBrazil
| | | |
Collapse
|
36
|
Dou R, Liu X, Kan X, Shen X, Mao J, Shen H, Wu J, Chen H, Xu W, Li S, Wu T, Hong Y. Dendrobium officinale polysaccharide-induced neuron-like cells from bone marrow mesenchymal stem cells improve neuronal function a rat stroke model. Tissue Cell 2021; 73:101649. [PMID: 34583247 DOI: 10.1016/j.tice.2021.101649] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 08/25/2021] [Accepted: 09/13/2021] [Indexed: 01/15/2023]
Abstract
Various methods have been used to induce the neuronal differentiation of marrow mesenchymal stem cells (MSCs). However, the limited induction efficiency of cells in vitro has restricted their use. Therefore, identifying a simple and efficient treatment method is necessary. Dendrobium officinale is an important traditional Chinese medicine, and its main component, polysaccharides, has many pharmacological activities. However, the effects of D. officinale polysaccharide (DOP) on the neuronal differentiation of bone marrow mesenchymal stem cells (BMSCs) and treatment of ischaemic stroke remain unknown. We found that DOP promoted the neuronal differentiation of BMSCs by increasing the expression levels of neural markers, and the optimal concentration of DOP was 25 μg/mL. Additionally, the Notch signalling pathway was inhibited during the neuronal differentiation of BMSCs induced by DOP, and this effect was strengthened using an inhibitor of this pathway. The Wnt signalling pathway was activated during the differentiation of BMSCs, and inhibition of the Wnt signalling pathway downregulated the expression of neuronal genes. Furthermore, the transplantation of neuron-like cells induced by DOP improved neuronal recovery, as the brain infarct volume, neurologic severity scores and levels of inflammatory factors were all significantly reduced in vivo. In conclusion, DOP is an effective inducer of the neuronal differentiation of BMSCs and treatment option for ischaemic stroke.
Collapse
Affiliation(s)
- Rengang Dou
- Department of Rehabilitation Medicine, the Second Hospital of Anhui Medical University, No. 678 Furong Road, Economic and Technological Development Zone, Hefei, Anhui, 230061, China.
| | - Xue Liu
- Department of Rehabilitation Medicine, the Second Hospital of Anhui Medical University, No. 678 Furong Road, Economic and Technological Development Zone, Hefei, Anhui, 230061, China.
| | - Xiuli Kan
- Department of Rehabilitation Medicine, the Second Hospital of Anhui Medical University, No. 678 Furong Road, Economic and Technological Development Zone, Hefei, Anhui, 230061, China.
| | - Xianshan Shen
- Department of Rehabilitation Medicine, the Second Hospital of Anhui Medical University, No. 678 Furong Road, Economic and Technological Development Zone, Hefei, Anhui, 230061, China.
| | - Jing Mao
- Department of Rehabilitation Medicine, the Second Hospital of Anhui Medical University, No. 678 Furong Road, Economic and Technological Development Zone, Hefei, Anhui, 230061, China.
| | - Hongtao Shen
- Department of Rehabilitation Medicine, the Second Hospital of Anhui Medical University, No. 678 Furong Road, Economic and Technological Development Zone, Hefei, Anhui, 230061, China.
| | - Jianxian Wu
- Department of Rehabilitation Medicine, the Second Hospital of Anhui Medical University, No. 678 Furong Road, Economic and Technological Development Zone, Hefei, Anhui, 230061, China.
| | - Hanlin Chen
- Stomatologic Hospital & College, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, No. 69 Meishan Road, Shushan District, Hefei, Anhui, 230001, China.
| | - Wanting Xu
- Stomatologic Hospital & College, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, No. 69 Meishan Road, Shushan District, Hefei, Anhui, 230001, China.
| | - Shasha Li
- Stomatologic Hospital & College, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, No. 69 Meishan Road, Shushan District, Hefei, Anhui, 230001, China.
| | - Tingting Wu
- Stomatologic Hospital & College, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, No. 69 Meishan Road, Shushan District, Hefei, Anhui, 230001, China.
| | - Yongfeng Hong
- Department of Rehabilitation Medicine, the Second Hospital of Anhui Medical University, No. 678 Furong Road, Economic and Technological Development Zone, Hefei, Anhui, 230061, China.
| |
Collapse
|
37
|
Abstract
The human brain is characterized by the large size and intricate folding of its cerebral cortex, which are fundamental for our higher cognitive function and frequently altered in pathological dysfunction. Cortex folding is not unique to humans, nor even to primates, but is common across mammals. Cortical growth and folding are the result of complex developmental processes that involve neural stem and progenitor cells and their cellular lineages, the migration and differentiation of neurons, and the genetic programs that regulate and fine-tune these processes. All these factors combined generate mechanical stress and strain on the developing neural tissue, which ultimately drives orderly cortical deformation and folding. In this review we examine and summarize the current knowledge on the molecular, cellular, histogenic and mechanical mechanisms that are involved in and influence folding of the cerebral cortex, and how they emerged and changed during mammalian evolution. We discuss the main types of pathological malformations of human cortex folding, their specific developmental origin, and how investigating their genetic causes has illuminated our understanding of key events involved. We close our review by presenting the state-of-the-art animal and in vitro models of cortex folding that are currently used to study these devastating developmental brain disorders in children, and what are the main challenges that remain ahead of us to fully understand brain folding.
Collapse
Affiliation(s)
- Lucia Del Valle Anton
- Instituto de Neurociencias, Agencia Estatal Consejo Superior de Investigaciones Científicas, San Juan de Alicante, Alicante, Spain
| | - Victor Borrell
- Instituto de Neurociencias, Agencia Estatal Consejo Superior de Investigaciones Científicas, San Juan de Alicante, Alicante, Spain
| |
Collapse
|
38
|
Jiménez S, Moreno N. Analysis of the Expression Pattern of Cajal-Retzius Cell Markers in the Xenopus laevis Forebrain. BRAIN, BEHAVIOR AND EVOLUTION 2021; 96:263-282. [PMID: 34614492 DOI: 10.1159/000519025] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 08/09/2021] [Indexed: 01/26/2023]
Abstract
Cajal-Retzius cells are essential for cortical development in mammals, and their involvement in the evolution of this structure has been widely postulated, but very little is known about their progenitor domains in non-mammalian vertebrates. Using in situhybridization and immunofluorescence techniques we analyzed the expression of some of the main Cajal-Retzius cell markers such as Dbx1, Ebf3, ER81, Lhx1, Lhx5, p73, Reelin, Wnt3a, Zic1, and Zic2 in the forebrain of the anuran Xenopus laevis, because amphibians are the only class of anamniote tetrapods and show a tetrapartite evaginated pallium, but no layered or nuclear organization. Our results suggested that the Cajal-Retzius cell progenitor domains were comparable to those previously described in amniotes. Thus, at dorsomedial telencephalic portions a region comparable to the cortical hem was defined in Xenopus based on the expression of Wnt3a, p73, Reelin, Zic1, and Zic2. In the septum, two different domains were observed: a periventricular dorsal septum, at the limit between the pallium and the subpallium, expressing Reelin, Zic1, and Zic2, and a related septal domain, expressing Ebf3, Zic1, and Zic2. In the lateral telencephalon, the ventral pallium next to the pallio-subpallial boundary, the lack of Dbx1 and the unique expression of Reelin during development defined this territory as the most divergent with respect to mammals. Finally, we also analyzed the expression of these markers at the prethalamic eminence region, suggested as Cajal-Retzius progenitor domain in amniotes, observing there Zic1, Zic2, ER81, and Lhx1 expression. Our data show that in anurans there are different subtypes and progenitor domains of Cajal-Retzius cells, which probably contribute to the cortical regional specification and territory-specific properties. This supports the notion that the basic organization of pallial derivatives in vertebrates follows a comparable fundamental arrangement, even in those that do not have a sophisticated stratified cortical structure like the mammalian cerebral cortex.
Collapse
Affiliation(s)
- Sara Jiménez
- Department of Cell Biology, Faculty of Biology, University Complutense, Madrid, Spain
| | - Nerea Moreno
- Department of Cell Biology, Faculty of Biology, University Complutense, Madrid, Spain
| |
Collapse
|
39
|
Da Silva F, Zhang K, Pinson A, Fatti E, Wilsch‐Bräuninger M, Herbst J, Vidal V, Schedl A, Huttner WB, Niehrs C. Mitotic WNT signalling orchestrates neurogenesis in the developing neocortex. EMBO J 2021; 40:e108041. [PMID: 34431536 PMCID: PMC8488556 DOI: 10.15252/embj.2021108041] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 08/05/2021] [Accepted: 08/10/2021] [Indexed: 02/06/2023] Open
Abstract
The role of WNT/β-catenin signalling in mouse neocortex development remains ambiguous. Most studies demonstrate that WNT/β-catenin regulates progenitor self-renewal but others suggest it can also promote differentiation. Here we explore the role of WNT/STOP signalling, which stabilizes proteins during G2/M by inhibiting glycogen synthase kinase (GSK3)-mediated protein degradation. We show that mice mutant for cyclin Y and cyclin Y-like 1 (Ccny/l1), key regulators of WNT/STOP signalling, display reduced neurogenesis in the developing neocortex. Specifically, basal progenitors, which exhibit delayed cell cycle progression, were drastically decreased. Ccny/l1-deficient apical progenitors show reduced asymmetric division due to an increase in apical-basal astral microtubules. We identify the neurogenic transcription factors Sox4 and Sox11 as direct GSK3 targets that are stabilized by WNT/STOP signalling in basal progenitors during mitosis and that promote neuron generation. Our work reveals that WNT/STOP signalling drives cortical neurogenesis and identifies mitosis as a critical phase for neural progenitor fate.
Collapse
Affiliation(s)
| | - Kaiqing Zhang
- Division of Molecular EmbryologyDKFZHeidelbergGermany
| | - Anneline Pinson
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
| | - Edoardo Fatti
- Division of Molecular EmbryologyDKFZHeidelbergGermany
- Present address:
Department of BiologyInstitute of BiochemistryETH (Eidgenössische Technische Hochschule)ZürichSwitzerland
| | | | | | | | | | - Wieland B Huttner
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
| | - Christof Niehrs
- Division of Molecular EmbryologyDKFZHeidelbergGermany
- Institute of Molecular Biology (IMB)MainzGermany
| |
Collapse
|
40
|
Ciptasari U, van Bokhoven H. The phenomenal epigenome in neurodevelopmental disorders. Hum Mol Genet 2021; 29:R42-R50. [PMID: 32766754 PMCID: PMC7530535 DOI: 10.1093/hmg/ddaa175] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 07/16/2020] [Accepted: 07/30/2020] [Indexed: 12/12/2022] Open
Abstract
Disruption of chromatin structure due to epimutations is a leading genetic etiology of neurodevelopmental disorders, collectively known as chromatinopathies. We show that there is an increasing level of convergence from the high diversity of genes that are affected by mutations to the molecular networks and pathways involving the respective proteins, the disrupted cellular and subcellular processes, and their consequence for higher order cellular network function. This convergence is ultimately reflected by specific phenotypic features shared across the various chromatinopathies. Based on these observations, we propose that the commonly disrupted molecular and cellular anomalies might provide a rational target for the development of symptomatic interventions for defined groups of genetically distinct neurodevelopmental disorders.
Collapse
Affiliation(s)
- Ummi Ciptasari
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behavior, Radboud university medical center, 6500 HB Nijmegen, The Netherlands
| | - Hans van Bokhoven
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behavior, Radboud university medical center, 6500 HB Nijmegen, The Netherlands.,Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud university medical center, 6500 HB Nijmegen, The Netherlands
| |
Collapse
|
41
|
Bhagat R, Rajpara P, Kaur G, Gupta K, Seth P. Zika virus E protein dysregulate mir-204/WNT2 signalling in human fetal neural stem cells. Brain Res Bull 2021; 176:93-102. [PMID: 34425198 DOI: 10.1016/j.brainresbull.2021.08.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 08/17/2021] [Accepted: 08/18/2021] [Indexed: 12/17/2022]
Abstract
Zika Virus (ZIKV) belongs to the family of flaviviruses, and is neurotrophic. It has been known to cause severe congenital disabilities including microcephaly in neonates. The virus has a specific preference towards neural stem cells (NSCs). ZIKV impairs proliferation and differentiation of NSCs during in-utero brain development of the fetus. However, molecular pathways involved in ZIKV induced alteration in NSCs are yet to be explored. In our previous study, we have described that ZIKV E protein dysregulates microRNA circuitry in NSCs and also impairs their proliferative and differentiation abilities. WNT signalling was found to be the target of differentially expressed miRNAs as suggested by PANTHER PATHWAY analysis of differentially expressed miRNA targets. In our current follow-up study, we investigate that WNT2 is downregulated in response to ZIKV E protein in human fetal NSCs and WNT2 is the molecular target of microRNA miR-204-5p. We provide pieces of evidences that miR-204-5p/WNT2 axis is involved in ZIKV induced impairment in the proliferation and immature differentiation of neural stem cells.
Collapse
Affiliation(s)
- Reshma Bhagat
- Department of Cellular and Molecular Neuroscience, Neurovirology Section, National Brain Research Centre, Manesar, Gurgaon, Haryana, India.
| | - Prateek Rajpara
- Department of Cellular and Molecular Neuroscience, Neurovirology Section, National Brain Research Centre, Manesar, Gurgaon, Haryana, India.
| | - Guneet Kaur
- Department of Cellular and Molecular Neuroscience, Neurovirology Section, National Brain Research Centre, Manesar, Gurgaon, Haryana, India.
| | - Karnika Gupta
- Department of Cellular and Molecular Neuroscience, Neurovirology Section, National Brain Research Centre, Manesar, Gurgaon, Haryana, India.
| | - Pankaj Seth
- Department of Cellular and Molecular Neuroscience, Neurovirology Section, National Brain Research Centre, Manesar, Gurgaon, Haryana, India.
| |
Collapse
|
42
|
Godoy JA, Espinoza-Caicedo J, Inestrosa NC. Morphological neurite changes induced by porcupine inhibition are rescued by Wnt ligands. Cell Commun Signal 2021; 19:87. [PMID: 34399774 PMCID: PMC8369806 DOI: 10.1186/s12964-021-00709-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 01/18/2021] [Indexed: 11/13/2022] Open
Abstract
Background Wnt signaling plays key roles in cellular and physiological processes, including cell proliferation, differentiation and migration during development and tissue homeostasis in adults. This pathway can be defined as Wnt/β-catenin-dependent or β-catenin-independent or “non-canonical”, both signaling are involved in neurite and synapse development/maintenance. Porcupine (PORCN), an acylase that o-acylates Wnt ligands, a major modification in secretion and interaction with its receptors. We use Wnt-C59, a specific PORCN inhibitor, to block the secretion of endogenous Wnts in embryonic hippocampal neurons (DIV 4). Under these conditions, the activity of exogenous Wnt ligands on the complexity of the dendritic tree and axonal polarity were evaluated Methods Cultured primary embryonic hippocampal neurons obtained from Sprague–Dawley rat fetuses (E18), were cultured until day in vitro (DIV) 4 (according to Banker´s protocol) and treated with Wnt-C59 for 24 h, Wnt ligands were added to the cultures on DIV 3 for 24 h. Dendritic arbors and neurites were analysis by fluorescence microscopy. Transfection with Lipofectamine 2000 on DIV 2 of plasmid expressing eGFP and KIF5-Cherry was carried out to evaluate neuronal polarity. Immunostaining was performed with MAP1B and Tau protein. Immunoblot analysis was carried out with Wnt3a, β-catenin and GSK-3β (p-Ser9). Quantitative analysis of dendrite morphology was carried out with ImageJ (NIH) software with Neuron J Plugin. Results We report, here, that Wnt-C59 treatment changed the morphology of the dendritic arbors and neurites of embryonic hippocampal neurons, with decreases β-catenin and Wnt3a and an apparent increase in GSK-3β (p-Ser9) levels. No effect was observed on axonal polarity. In sister cultures, addition of exogenous Wnt3a, 5a and 7a ligands rescued the changes in neuronal morphology. Wnt3a restored the length of neurites to near that of the control, but Wnt7a increased the neurite length beyond that of the control. Wnt5a also restored the length of neurites relative to Wnt concentrations. Conclusions Results indicated that Wnt ligands, added exogenously, restored dendritic arbor complexity in embryonic hippocampal neurons, previously treated with a high affinity specific Porcupine inhibitor. We proposed that PORCN is an emerging molecular target of interest in the search for preclinical options to study and treat Wnt-related diseases. Video Abstract
Supplementary Information The online version contains supplementary material available at 10.1186/s12964-021-00709-y.
Collapse
Affiliation(s)
- Juan A Godoy
- Centro de Envejecimiento y Regeneración (CARE-UC), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O`Higgins 340, Santiago de Chile, Chile
| | - Jasson Espinoza-Caicedo
- Centro de Envejecimiento y Regeneración (CARE-UC), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O`Higgins 340, Santiago de Chile, Chile
| | - Nibaldo C Inestrosa
- Centro de Envejecimiento y Regeneración (CARE-UC), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O`Higgins 340, Santiago de Chile, Chile. .,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile.
| |
Collapse
|
43
|
Tang X, Zuo C, Fang P, Liu G, Qiu Y, Huang Y, Tang R. Targeting Glioblastoma Stem Cells: A Review on Biomarkers, Signal Pathways and Targeted Therapy. Front Oncol 2021; 11:701291. [PMID: 34307170 PMCID: PMC8297686 DOI: 10.3389/fonc.2021.701291] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 06/25/2021] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma (GBM) remains the most lethal and common primary brain tumor, even after treatment with multiple therapies, such as surgical resection, chemotherapy, and radiation. Although great advances in medical development and improvements in therapeutic methods of GBM have led to a certain extension of the median survival time of patients, prognosis remains poor. The primary cause of its dismal outcomes is the high rate of tumor recurrence, which is closely related to its resistance to standard therapies. During the last decade, glioblastoma stem cells (GSCs) have been successfully isolated from GBM, and it has been demonstrated that these cells are likely to play an indispensable role in the formation, maintenance, and recurrence of GBM tumors, indicating that GSCs are a crucial target for treatment. Herein, we summarize the current knowledge regarding GSCs, their related signaling pathways, resistance mechanisms, crosstalk linking mechanisms, and microenvironment or niche. Subsequently, we present a framework of targeted therapy for GSCs based on direct strategies, including blockade of the pathways necessary to overcome resistance or prevent their function, promotion of GSC differentiation, virotherapy, and indirect strategies, including targeting the perivascular, hypoxic, and immune niches of the GSCs. In summary, targeting GSCs provides a tremendous opportunity for revolutionary approaches to improve the prognosis and therapy of GBM, despite a variety of challenges.
Collapse
Affiliation(s)
- Xuejia Tang
- Department of Neurosurgery, University-Town Hospital of Chongqing Medical University, Chongqing, China.,Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Chenghai Zuo
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Pengchao Fang
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Guojing Liu
- Department of Neurosurgery, University-Town Hospital of Chongqing Medical University, Chongqing, China
| | - Yongyi Qiu
- Department of Neurosurgery, University-Town Hospital of Chongqing Medical University, Chongqing, China
| | - Yi Huang
- Department of Neurosurgery, The Ninth People's Hospital of Chongqing, Chongqing, China
| | - Rongrui Tang
- Department of Neurosurgery, University-Town Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
44
|
Hu L, Chen Y, Yang CP, Huang Y, Song NN, Chen JY, Sun YL, Ding YQ, Lang B. Ulk4, a Newly Discovered Susceptibility Gene for Schizophrenia, Regulates Corticogenesis in Mice. Front Cell Dev Biol 2021; 9:645368. [PMID: 34235142 PMCID: PMC8255617 DOI: 10.3389/fcell.2021.645368] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 04/23/2021] [Indexed: 11/13/2022] Open
Abstract
Schizophrenia (SCZ) is a chronic and severe mental disease that affects around 1% of the population. The precise etiology of SCZ still remains largely unknown, and no conclusive mechanisms are firmly established. Recent advances in epidemiological and clinical investigation support an overwhelmingly strong neurodevelopmental origin for SCZ. Here, we demonstrated that Unc-51-like kinase 4 (Ulk4), a novel risk factor for major mental disorders including schizophrenia, is involved in the corticogenesis. Deletion of Ulk4 in mice led to significantly thinner layers of II–III, and V in the cerebral cortex, which was confirmed in conditional Ulk4 deletion mice achieved by Cre-loxp strategy. This abnormality might be caused by decreased intermediate neural progenitors and increased apoptosis. Thus, our data suggest that Ulk4 manipulates the behaviors of neural progenitors during brain development and, when functionally defective, leads to the reduction of specific cortical layers. This anomaly may increase predisposition to a range of neurodevelopmental disorders, including SCZ.
Collapse
Affiliation(s)
- Ling Hu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yi Chen
- Key Laboratory of Arrhythmias, Ministry of Education, East Hospital, and Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai, China
| | - Cui-Ping Yang
- Key Laboratory of Arrhythmias, Ministry of Education, East Hospital, and Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai, China
| | - Ying Huang
- Department of Laboratory Animal Science, Fudan University, Shanghai, China
| | - Ning-Ning Song
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Jia-Yin Chen
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yu-Ling Sun
- Key Laboratory of Arrhythmias, Ministry of Education, East Hospital, and Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai, China
| | - Yu-Qiang Ding
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China.,Key Laboratory of Arrhythmias, Ministry of Education, East Hospital, and Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai, China.,Department of Laboratory Animal Science, Fudan University, Shanghai, China
| | - Bing Lang
- National Clinical Research Centre for Mental Health, Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
45
|
Choi BR, Cave C, Na CH, Sockanathan S. GDE2-Dependent Activation of Canonical Wnt Signaling in Neurons Regulates Oligodendrocyte Maturation. Cell Rep 2021; 31:107540. [PMID: 32375055 PMCID: PMC7254694 DOI: 10.1016/j.celrep.2020.107540] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 03/09/2020] [Accepted: 03/28/2020] [Indexed: 12/30/2022] Open
Abstract
Neurons and oligodendrocytes communicate to regulate oligodendrocyte development and ensure appropriate axonal myelination. Here, we show that Glycerophosphodiester phosphodiesterase 2 (GDE2) signaling underlies a neuronal pathway that promotes oligodendrocyte maturation through the release of soluble neuronally derived factors. Mice lacking global or neuronal GDE2 expression have reduced mature oligodendrocytes and myelin proteins but retain normal numbers of oligodendrocyte precursor cells (OPCs). Wild-type (WT) OPCs cultured in conditioned medium (CM) from Gde2-null (Gde2KO) neurons exhibit delayed maturation, recapitulating in vivo phenotypes. Gde2KO neurons show robust reduction in canonical Wnt signaling, and genetic activation of Wnt signaling in Gde2KO neurons rescues in vivo and in vitro oligodendrocyte maturation. Phosphacan, a known stimulant of oligodendrocyte maturation, is reduced in CM from Gde2KO neurons but is restored when Wnt signaling is activated. These studies identify GDE2 control of Wnt signaling as a neuronal pathway that signals to oligodendroglia to promote oligodendrocyte maturation.
Collapse
Affiliation(s)
- Bo-Ran Choi
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, 725 N. Wolfe Street, PCTB 1004, Baltimore, MD 21205, USA
| | - Clinton Cave
- Neuroscience Program, Middlebury College, 276 Bicentennial Way, MBH 351, Middlebury, VT 05753, USA
| | - Chan Hyun Na
- Department of Neurology, Institute for Cell Engineering, Johns Hopkins University School of Medicine, 733 N. Broadway, MRB 753, Baltimore, MD 21205, USA
| | - Shanthini Sockanathan
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, 725 N. Wolfe Street, PCTB 1004, Baltimore, MD 21205, USA.
| |
Collapse
|
46
|
Ou MY, Xiao Q, Ju XC, Zeng PM, Huang J, Sheng AL, Luo ZG. The CTNNBIP1-CLSTN1 fusion transcript regulates human neocortical development. Cell Rep 2021; 35:109290. [PMID: 34192541 DOI: 10.1016/j.celrep.2021.109290] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 02/17/2021] [Accepted: 06/02/2021] [Indexed: 12/21/2022] Open
Abstract
Fusion transcripts or RNAs have been found in both disordered and healthy human tissues and cells; however, their physiological functions in the brain development remain unknown. In the analysis of deposited RNA-sequence libraries covering early to middle embryonic stages, we identify 1,055 fusion transcripts present in the developing neocortex. Interestingly, 98 fusion transcripts exhibit distinct expression patterns in various neural progenitors (NPs) or neurons. We focus on CTNNBIP1-CLSTN1 (CTCL), which is enriched in outer radial glial cells that contribute to cortex expansion during human evolution. Intriguingly, downregulation of CTCL in cultured human cerebral organoids causes marked reduction in NPs and precocious neuronal differentiation, leading to impairment of organoid growth. Furthermore, the expression of CTCL fine-tunes Wnt/β-catenin signaling that controls cortex patterning. Together, this work provides evidence indicating important roles of fusion transcript in human brain development and evolution.
Collapse
Affiliation(s)
- Min-Yi Ou
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institute for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qi Xiao
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institute for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiang-Chun Ju
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institute for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Peng-Ming Zeng
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Jing Huang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Ai-Li Sheng
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institute for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Zhen-Ge Luo
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.
| |
Collapse
|
47
|
Yoon J, Mao Y. Dissecting Molecular Genetic Mechanisms of 1q21.1 CNV in Neuropsychiatric Disorders. Int J Mol Sci 2021; 22:5811. [PMID: 34071723 PMCID: PMC8197994 DOI: 10.3390/ijms22115811] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/23/2021] [Accepted: 05/25/2021] [Indexed: 11/17/2022] Open
Abstract
Pathogenic copy number variations (CNVs) contribute to the etiology of neurodevelopmental/neuropsychiatric disorders (NDs). Increased CNV burden has been found to be critically involved in NDs compared with controls in clinical studies. The 1q21.1 CNVs, rare and large chromosomal microduplications and microdeletions, are detected in many patients with NDs. Phenotypes of duplication and deletion appear at the two ends of the spectrum. Microdeletions are predominant in individuals with schizophrenia (SCZ) and microcephaly, whereas microduplications are predominant in individuals with autism spectrum disorder (ASD) and macrocephaly. However, its complexity hinders the discovery of molecular pathways and phenotypic networks. In this review, we summarize the recent genome-wide association studies (GWASs) that have identified candidate genes positively correlated with 1q21.1 CNVs, which are likely to contribute to abnormal phenotypes in carriers. We discuss the clinical data implicated in the 1q21.1 genetic structure that is strongly associated with neurodevelopmental dysfunctions like cognitive impairment and reduced synaptic plasticity. We further present variations reported in the phenotypic severity, genomic penetrance and inheritance.
Collapse
Affiliation(s)
| | - Yingwei Mao
- Department of Biology, Eberly College of Science, Pennsylvania State University, University Park, PA 16802, USA;
| |
Collapse
|
48
|
Prieto-Colomina A, Fernández V, Chinnappa K, Borrell V. MiRNAs in early brain development and pediatric cancer: At the intersection between healthy and diseased embryonic development. Bioessays 2021; 43:e2100073. [PMID: 33998002 DOI: 10.1002/bies.202100073] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/12/2021] [Accepted: 04/15/2021] [Indexed: 12/15/2022]
Abstract
The size and organization of the brain are determined by the activity of progenitor cells early in development. Key mechanisms regulating progenitor cell biology involve miRNAs. These small noncoding RNA molecules bind mRNAs with high specificity, controlling their abundance and expression. The role of miRNAs in brain development has been studied extensively, but their involvement at early stages remained unknown until recently. Here, recent findings showing the important role of miRNAs in the earliest phases of brain development are reviewed, and it is discussed how loss of specific miRNAs leads to pathological conditions, particularly adult and pediatric brain tumors. Let-7 miRNA downregulation and the initiation of embryonal tumors with multilayered rosettes (ETMR), a novel link recently discovered by the laboratory, are focused upon. Finally, it is discussed how miRNAs may be used for the diagnosis and therapeutic treatment of pediatric brain tumors, with the hope of improving the prognosis of these devastating diseases.
Collapse
Affiliation(s)
- Anna Prieto-Colomina
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas & Universidad Miguel Hernández, Sant Joan d'Alacant, Spain
| | - Virginia Fernández
- Neurobiology of miRNA, Fondazione Istituto Italiano di Tecnologia (IIT), Genoa, Italy
| | - Kaviya Chinnappa
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas & Universidad Miguel Hernández, Sant Joan d'Alacant, Spain
| | - Víctor Borrell
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas & Universidad Miguel Hernández, Sant Joan d'Alacant, Spain
| |
Collapse
|
49
|
Cederquist GY, Tchieu J, Callahan SJ, Ramnarine K, Ryan S, Zhang C, Rittenhouse C, Zeltner N, Chung SY, Zhou T, Chen S, Betel D, White RM, Tomishima M, Studer L. A Multiplex Human Pluripotent Stem Cell Platform Defines Molecular and Functional Subclasses of Autism-Related Genes. Cell Stem Cell 2021; 27:35-49.e6. [PMID: 32619517 DOI: 10.1016/j.stem.2020.06.004] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/26/2020] [Accepted: 06/05/2020] [Indexed: 01/12/2023]
Abstract
Autism is a clinically heterogeneous neurodevelopmental disorder characterized by impaired social interactions, restricted interests, and repetitive behaviors. Despite significant advances in the genetics of autism, understanding how genetic changes perturb brain development and affect clinical symptoms remains elusive. Here, we present a multiplex human pluripotent stem cell (hPSC) platform, in which 30 isogenic disease lines are pooled in a single dish and differentiated into prefrontal cortex (PFC) lineages to efficiently test early-developmental hypotheses of autism. We define subgroups of autism mutations that perturb PFC neurogenesis and are correlated to abnormal WNT/βcatenin responses. Class 1 mutations (8 of 27) inhibit while class 2 mutations (5 of 27) enhance PFC neurogenesis. Remarkably, autism patient data reveal that individuals carrying subclass-specific mutations differ clinically in their corresponding language acquisition profiles. Our study provides a framework to disentangle genetic heterogeneity associated with autism and points toward converging molecular and developmental pathways of diverse autism-associated mutations.
Collapse
Affiliation(s)
- Gustav Y Cederquist
- The Center for Stem Cell Biology, Developmental Biology Program, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA; Weill-Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY 10065, USA
| | - Jason Tchieu
- The Center for Stem Cell Biology, Developmental Biology Program, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Scott J Callahan
- The Center for Stem Cell Biology, Developmental Biology Program, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA; Cancer Genetics and Biology Program, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA; Gerstner Graduate School of Biomedical Sciences, Developmental Biology Program, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Kiran Ramnarine
- The Center for Stem Cell Biology, Developmental Biology Program, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Sean Ryan
- The Center for Stem Cell Biology, Developmental Biology Program, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Chao Zhang
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10065, USA; Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Chelsea Rittenhouse
- The Center for Stem Cell Biology, Developmental Biology Program, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Nadja Zeltner
- The Center for Stem Cell Biology, Developmental Biology Program, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA; Center for Molecular Medicine, Department of Cellular Biology, Department of Biochemistry & Molecular Biology, University of Georgia, Athens, GA 30602, USA
| | - Sun Young Chung
- The Center for Stem Cell Biology, Developmental Biology Program, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Ting Zhou
- The Center for Stem Cell Biology, Developmental Biology Program, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA; Department of Surgery, Weill Cornell Medical College, New York, NY 10065, USA
| | - Shuibing Chen
- Department of Surgery, Weill Cornell Medical College, New York, NY 10065, USA
| | - Doron Betel
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10065, USA; Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Richard M White
- Cancer Genetics and Biology Program, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Mark Tomishima
- The Center for Stem Cell Biology, Developmental Biology Program, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Lorenz Studer
- The Center for Stem Cell Biology, Developmental Biology Program, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA.
| |
Collapse
|
50
|
Ruan X, Liu G, Zhou J, Chen P, Sun C, Liu W, Wu C, Hou L, Yin B, Qiang B, Shu P, Peng X. Zbed3 Is Indispensable for Wnt Signaling Regulation of Cortical Layers Formation in Developing Brain. Cereb Cortex 2021; 31:4078-4091. [PMID: 33822906 DOI: 10.1093/cercor/bhab070] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 03/01/2021] [Accepted: 03/03/2021] [Indexed: 11/15/2022] Open
Abstract
Wnt/β-catenin signaling plays multiple important roles during mammalian brain development, and it regulates the proliferation and differentiation of neural progenitors in a context-dependent manner and affects neocortex layer formation. However, the specific role of Wnt/β-catenin in neuronal layer fate determination in the neocortex is still unclear. Here, we report that Zbed3, which is a positive regulator of Wnt/β-catenin signaling, colocalizes with β-catenin at the endfeet of radial glia in the ventricular zone of embryo mouse neocortex. Overexpression and knockdown of Zbed3 increased and decreased the activity of Wnt/β-catenin signaling in the neocortex, respectively. Interestingly, knockdown of Zbed3 in vivo could significantly shift neuronal fates from deep layers to upper layers but is not required for the proliferation and differentiation of neural progenitors. Overexpression of Zbed3 led to increased generation of deep-layer neurons without impairing cell cycle exit of neural progenitors. More importantly, knockdown of Zbed3 could effectively block the effects of the ectopic expression of stabilized β-catenin on neocortex layer formation. Hence, our results demonstrate that Zbed3 is indispensable for Wnt/β-catenin signaling regulating neuronal layer fates in the developing brain.
Collapse
Affiliation(s)
- Xiangbin Ruan
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primate Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Gaoao Liu
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primate Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Jiafeng Zhou
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primate Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Pan Chen
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primate Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Changjie Sun
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primate Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Wei Liu
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primate Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Chao Wu
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primate Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Lin Hou
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primate Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Bin Yin
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primate Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Boqin Qiang
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primate Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Pengcheng Shu
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primate Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China.,Chinese Institute for Brain Research, Beijing 102206, China
| | - Xiaozhong Peng
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primate Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China.,Institute of Medical Biology of the Chinese Academy of Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| |
Collapse
|