1
|
Como CN, O'Rourke R, Winkler C, Mitchell D, Tran L, Lorberbaum D, Sussel L, Franco S, Siegenthaler J. Meningeal-derived retinoic acid regulates neurogenesis via suppression of Notch and Sox2. Cell Rep 2025; 44:115637. [PMID: 40310723 DOI: 10.1016/j.celrep.2025.115637] [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: 04/05/2024] [Revised: 02/03/2025] [Accepted: 04/10/2025] [Indexed: 05/03/2025] Open
Abstract
The meninges act as a regulator of brain development by secreting ligands that act on neural cells to regulate neurogenesis and neuronal migration. Meningeal-derived retinoic acid (RA) promotes neocortical neural progenitor cell cycle exit; however, the underlying molecular mechanism is unknown. Here, we used spatial transcriptomics and profiling of retinoic acid receptor α (RARα) DNA binding in Foxc1-mutant embryos that lack meninges-derived signals to identify potential neurogenic transcriptional mechanisms of RA signaling in telencephalic neural progenitors. This identified upregulation of Sox2 and Notch pathway genes, and RARα binds to the Sox2ot promoter, a long noncoding RNA that regulates Sox2 expression. Our experiments using maternal RA treatment and in utero electroporation in Foxc1 mutants support that meningeal-derived RA promotes neurogenesis by suppressing Notch signaling, a progenitor self-renewal pathway. Our findings elucidate a previously unknown mechanism of how meningeal RA coordinates neocortical development and provide insight into how defects in meningeal development may cause neurodevelopmental disorders.
Collapse
Affiliation(s)
- Christina N Como
- University of Colorado Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, Aurora, CO 80045, USA; University of Colorado Anschutz Medical Campus, Neuroscience Graduate Program, Aurora, CO 80045, USA
| | - Rebecca O'Rourke
- University of Colorado Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, Aurora, CO 80045, USA
| | - Caitlin Winkler
- University of Colorado Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, Aurora, CO 80045, USA
| | - Danae Mitchell
- University of Colorado Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, Aurora, CO 80045, USA; University of Colorado Anschutz Medical Campus, Neuroscience Graduate Program, Aurora, CO 80045, USA
| | - Luuli Tran
- University of Colorado Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, Aurora, CO 80045, USA; University of Colorado Anschutz Medical Campus, Molecular Biology Graduate Program, Aurora, CO 80045, USA
| | - David Lorberbaum
- University of Colorado Anschutz Medical Campus, Department of Pediatrics, Barbara Davis Center for Childhood Diabetes, Aurora, CO 80045, USA; University of Michigan Medical School, Department of Pharmacology and Caswell Diabetes Institute, Ann Arbor, MI 48105, USA
| | - Lori Sussel
- University of Colorado Anschutz Medical Campus, Department of Pediatrics, Barbara Davis Center for Childhood Diabetes, Aurora, CO 80045, USA
| | - Santos Franco
- University of Colorado Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, Aurora, CO 80045, USA.
| | - Julie Siegenthaler
- University of Colorado Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, Aurora, CO 80045, USA; University of Colorado Anschutz Medical Campus, Neuroscience Graduate Program, Aurora, CO 80045, USA.
| |
Collapse
|
2
|
Cong Y, Wu Y, Liu Y, Ai Y, Wang X, Wei C, Ding H, Xu G, Sun W. TCDD inhibits the proliferation of C17.2 cells through the activation of the c-Cbl/β-catenin signaling pathway. Toxicol In Vitro 2025; 104:106014. [PMID: 39880321 DOI: 10.1016/j.tiv.2025.106014] [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: 08/22/2024] [Revised: 12/23/2024] [Accepted: 01/26/2025] [Indexed: 01/31/2025]
Abstract
2,3,7,8-tetrachlordibenzo-p-dioxin (TCDD) belongs to the category of persistent environmental pollutants, and gestational exposure to TCDD can lead to cognitive, memory, and motor deficits, as well as altered neuron development in rodents. However, the molecular mechanisms underlying TCDD's neurotoxicity remain unclear. Neural stem cells (NSCs) possess the capacity for self-renewal and can generate various cell types within the brain, playing fundamental roles in brain development and regeneration. This study investigated the impact of TCDD on the proliferation of mouse NSCs, specifically focusing on the C17.2 cell line. The results demonstrated that TCDD inhibited the proliferation of C17.2 cells in a dose-dependent manner. Even low doses of TCDD (5 nM) significantly reduced C17.2 cell proliferation. Regarding the molecular mechanisms, it was found that TCDD induced the degradation of β-catenin, a key regulator of cell proliferation, through the upregulation of the E3 ubiquitin ligase, casitas B-lineage lymphoma (c-Cbl), which was dependent on the aryl-hydrocarbon Receptor (AhR). Furthermore, knockdown of c-Cbl alleviated the TCDD-induced inhibition of C17.2 proliferation and of the reduction of β-catenin expression. Our research provides foundational data to understand the mechanism of TCDD-induced neurotoxicity through the inhibition of NSCs proliferation, and suggests that the c-cbl/β-catenin pathway may serve as a potential therapeutic target for countering the neurotoxicants of TCDD.
Collapse
Affiliation(s)
- Yewen Cong
- School of Public Health, Nantong University, Nantong 226019, Jiangsu, China; Affiliated Matern & Care Hospital, Nantong University, Nantong 226007, Jiangsu, China
| | - Yue Wu
- School of Public Health, Nantong University, Nantong 226019, Jiangsu, China
| | - Yue Liu
- School of Public Health, Nantong University, Nantong 226019, Jiangsu, China
| | - Yongjun Ai
- School of Public Health, Nantong University, Nantong 226019, Jiangsu, China
| | - Xiping Wang
- School of Public Health, Nantong University, Nantong 226019, Jiangsu, China
| | - Chunxi Wei
- School of Public Health, Nantong University, Nantong 226019, Jiangsu, China
| | - Haoyu Ding
- School of Public Health, Nantong University, Nantong 226019, Jiangsu, China
| | - Guangfei Xu
- School of Public Health, Nantong University, Nantong 226019, Jiangsu, China.
| | - Wenxing Sun
- School of Public Health, Nantong University, Nantong 226019, Jiangsu, China.
| |
Collapse
|
3
|
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
|
4
|
Walter NM, Yde Ohki CM, Ruhstaller S, Del Campana L, Salazar Campos JM, Smigielski L, Rubio B, Walitza S, Grünblatt E. Neurodevelopmental effects of omega-3 fatty acids and its combination with Methylphenidate in iPSC models of ADHD. J Psychiatr Res 2025; 184:78-90. [PMID: 40043588 DOI: 10.1016/j.jpsychires.2025.02.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/07/2024] [Accepted: 02/20/2025] [Indexed: 04/09/2025]
Abstract
Attention-Deficit/Hyperactivity Disorder (ADHD) has been linked to altered neurodevelopmental processes, including proliferation and differentiation of neural stem cells (NSC). We aimed to investigate the role of Wnt signaling, a pathway critical for brain development, in ADHD and to determine if modulation of this pathway using ω-3/6 polyunsaturated fatty acids (PUFAs) may provide a beneficial treatment approach. Given the symptom heterogeneity in ADHD and the limited response to conventional therapies for some patients, we examined the effects of ω-3/6 PUFA treatment combined with Methylphenidate (MPH) on neurodevelopmental mechanisms using induced pluripotent stem cell (iPSC)-derived NSCs, comparing controls to ADHD patients. Our results show that ω-3/6 PUFAs differentially regulate Wnt activity in NSCs depending on the patient's condition and the composition of the treatments. These findings highlight the potential of ω-3 PUFA treatment as personalized support for neurodevelopmental processes in ADHD. They also emphasize the importance of investigating ADHD subgroups, including those unresponsive to stimulant treatments, as they may exhibit distinct phenotypes.
Collapse
Affiliation(s)
- Natalie M Walter
- Department of Child and Adolescent Psychiatry and Psychotherapy, Translational Molecular Psychiatry, Psychiatric University Hospital Zurich, University of Zurich, Wagistrasse 12, 8952, Schlieren, Switzerland; ZNZ PhD Program, University of Zurich, Winterthurerstrasse 11, 8057, Zurich, Switzerland
| | - Cristine M Yde Ohki
- Department of Child and Adolescent Psychiatry and Psychotherapy, Translational Molecular Psychiatry, Psychiatric University Hospital Zurich, University of Zurich, Wagistrasse 12, 8952, Schlieren, Switzerland
| | - Sina Ruhstaller
- Department of Child and Adolescent Psychiatry and Psychotherapy, Translational Molecular Psychiatry, Psychiatric University Hospital Zurich, University of Zurich, Wagistrasse 12, 8952, Schlieren, Switzerland
| | - Letizia Del Campana
- Department of Child and Adolescent Psychiatry and Psychotherapy, Translational Molecular Psychiatry, Psychiatric University Hospital Zurich, University of Zurich, Wagistrasse 12, 8952, Schlieren, Switzerland
| | - José Maria Salazar Campos
- Department of Child and Adolescent Psychiatry and Psychotherapy, Translational Molecular Psychiatry, Psychiatric University Hospital Zurich, University of Zurich, Wagistrasse 12, 8952, Schlieren, Switzerland
| | - Lukasz Smigielski
- Department of Child and Adolescent Psychiatry and Psychotherapy, Translational Molecular Psychiatry, Psychiatric University Hospital Zurich, University of Zurich, Wagistrasse 12, 8952, Schlieren, Switzerland
| | - Belén Rubio
- Department of Child and Adolescent Psychiatry and Psychotherapy, Translational Molecular Psychiatry, Psychiatric University Hospital Zurich, University of Zurich, Wagistrasse 12, 8952, Schlieren, Switzerland
| | - Susanne Walitza
- Department of Child and Adolescent Psychiatry and Psychotherapy, Translational Molecular Psychiatry, Psychiatric University Hospital Zurich, University of Zurich, Wagistrasse 12, 8952, Schlieren, Switzerland; Neuroscience Center Zurich, University of Zurich and the ETH Zurich, Winterthurerstrasse 11, 8057, Zurich, Switzerland; Zurich Center for Integrative Human Physiology, University of Zurich, Winterthurerstrasse 11, 8057, Zurich, Switzerland
| | - Edna Grünblatt
- Department of Child and Adolescent Psychiatry and Psychotherapy, Translational Molecular Psychiatry, Psychiatric University Hospital Zurich, University of Zurich, Wagistrasse 12, 8952, Schlieren, Switzerland; Neuroscience Center Zurich, University of Zurich and the ETH Zurich, Winterthurerstrasse 11, 8057, Zurich, Switzerland; Zurich Center for Integrative Human Physiology, University of Zurich, Winterthurerstrasse 11, 8057, Zurich, Switzerland.
| |
Collapse
|
5
|
Harigaya Y, Matoba N, Le BD, Valone JM, Stein JL, Love MI, Valdar W. Probabilistic classification of gene-by-treatment interactions on molecular count phenotypes. PLoS Genet 2025; 21:e1011561. [PMID: 40203278 PMCID: PMC12021428 DOI: 10.1371/journal.pgen.1011561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 04/24/2025] [Accepted: 12/31/2024] [Indexed: 04/11/2025] Open
Abstract
Genetic variation can modulate response to treatment (G×T) or environmental stimuli (G×E), both of which can be highly consequential in biomedicine. An effective approach to identifying G×T signals and gaining insight into molecular mechanisms is mapping quantitative trait loci (QTL) of molecular count phenotypes, such as gene expression and chromatin accessibility, under multiple treatment conditions, which is termed response molecular QTL mapping. Although standard approaches evaluate the interaction between genetics and treatment conditions, they do not distinguish between meaningful interpretations such as whether a genetic effect is observed only in the treated condition or whether a genetic effect is observed always but accentuated in the treated condition. To address this gap, we have developed a downstream method for classifying response molecular QTLs into subclasses with meaningful genetic interpretations. Our method uses Bayesian model selection and assigns posterior probabilities to different types of G×T interactions for a given feature-SNP pair. We compare linear and nonlinear regression of log -scale counts, noting that the latter accounts for an expected biological relationship between the genotype and the molecular count phenotype. Through simulation and application to existing datasets of molecular response QTLs, we show that our method provides an intuitive and well-powered framework to report and interpret G×T interactions. We provide a software package, ClassifyGxT [1].
Collapse
Affiliation(s)
- Yuriko Harigaya
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Nana Matoba
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Brandon D Le
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Jordan M Valone
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Jason L Stein
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Carolina Institute for Developmental Disabilities, Carrboro, North Carolina, United States of America
| | - Michael I Love
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - William Valdar
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| |
Collapse
|
6
|
Huang Z, He C, Wang G, Zhu M, Tong X, Feng Y, Zhang C, Dong S, Harim Y, Liu HK, Zhou W, Lan F, Feng W. Mutation of CHD7 impairs the output of neuroepithelium transition that is reversed by the inhibition of EZH2. Mol Psychiatry 2025:10.1038/s41380-025-02990-6. [PMID: 40164694 DOI: 10.1038/s41380-025-02990-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 03/07/2025] [Accepted: 03/25/2025] [Indexed: 04/02/2025]
Abstract
Haploinsufficiency of CHD7 (Chromo-Helicase-DNA binding protein 7) causes a severe congenital disease CHARGE syndrome. Brain anomaly such as microcephaly and olfactory bulb agenesis seen in CHARGE patients have not been mimicked in previous animal models. Here, we uncover an indispensable function of CHD7 in the neuroepithelium (NE) but not in the neural stem cells (NSCs) after NE transition. Loss of Chd7 in mouse NE resulted in CHARGE-like brain anomalies due to reduced proliferation and differentiation of neural stem and progenitor cells, which were recapitulated in CHD7 KO human forebrain organoids. Mechanistically, we find that CHD7 activates neural transcription factors by removing the repressive histone mark H3K27me3 and promoting chromatin accessibility. Importantly, neurodevelopmental defects caused by CHD7 loss in human brain organoids and mice were ameliorated by the inhibition of H3K27me3 methyltransferase EZH2. Altogether, by implementing appropriate experimental models, we uncover the pathogenesis of CHD7-associated neurodevelopmental diseases, and identify a potential therapeutic opportunity for CHARGE syndrome.
Collapse
Affiliation(s)
- Zhuxi Huang
- 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
| | - Chenxi He
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Liver Cancer Institute, Zhongshan Hospital, Fudan University and Shanghai Key Laboratory of Medical Epigenetics, International Laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Guangfu Wang
- 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
| | - Ming Zhu
- 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
| | - Xiaoyu Tong
- State Key Laboratory of Medical Neurobiology, Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, 200032, China
| | - Yi Feng
- State Key Laboratory of Medical Neurobiology, Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, 200032, China
| | - Chenyang Zhang
- 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
| | - Shuhua Dong
- 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
| | - Yassin Harim
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, 69120, Germany
| | - Hai-Kun Liu
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, 69120, Germany
| | - Wenhao Zhou
- Division of Neonatology and Center for Newborn Care, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Fei Lan
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Liver Cancer Institute, Zhongshan Hospital, Fudan University and Shanghai Key Laboratory of Medical Epigenetics, International Laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, 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.
| |
Collapse
|
7
|
Song JHT, Carter AC, Bushinsky EM, Beck SG, Petrocelli JE, Koreman GT, Babu J, Kingsley DM, Greenberg ME, Walsh CA. Human-chimpanzee tetraploid system defines mechanisms of species-specific neural gene regulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.31.646367. [PMID: 40236112 PMCID: PMC11996389 DOI: 10.1101/2025.03.31.646367] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
A major challenge in human evolutionary biology is to pinpoint genetic differences that underlie human-specific traits, such as increased neuron number and differences in cognitive behaviors. We used human-chimpanzee tetraploid cells to distinguish gene expression changes due to cis -acting sequence variants that change local gene regulation, from trans expression changes due to species differences in the cellular environment. In neural progenitor cells, examination of both cis and trans changes - combined with CRISPR inhibition and transcription factor motif analyses - identified cis -acting, species-specific gene regulatory changes, including to TNIK, FOSL2 , and MAZ , with widespread trans effects on neurogenesis-related gene programs. In excitatory neurons, we identified POU3F2 as a key cis -regulated gene with trans effects on synaptic gene expression and neuronal firing. This study identifies cis -acting genomic changes that cause cascading trans gene regulatory effects to contribute to human neural specializations, and provides a general framework for discovering genetic differences underlying human traits.
Collapse
|
8
|
Fu S, Wynshaw-Boris A. Autism risk genes converge on PBX1 to govern neural cell growth. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.12.642693. [PMID: 40161581 PMCID: PMC11952423 DOI: 10.1101/2025.03.12.642693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The alteration of neural progenitor cell (NPC) proliferation underlies autism spectrum disorders (ASD). It remains unclear whether targeting convergent downstream targets among mutations from different genes and individuals can rescue this alteration. We identified PBX1 as a convergent target of three autism risk genes: CTNNB1, PTEN, and DVL3, using isogenic iPSC-derived 2D NPCs. Overexpression of the PBX1a isoform effectively rescued increased NPC proliferation in all three isogenic ASD-related variants. Dysregulation of PBX1 in NPCs was further confirmed in publicly available datasets from other models of ASD. These findings spotlight PBX1, known to play important roles during olfactory bulb/adult neurogenesis and in multiple cancers, as an unexpected and key downstream target, influencing NPC proliferation in ASD and neurodevelopmental syndromes.
Collapse
Affiliation(s)
- Shuai Fu
- Department of Genetics and Genome Sciences, Case Western Reserve University; Cleveland, OH, USA
- Present address: Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Anthony Wynshaw-Boris
- Department of Genetics and Genome Sciences, Case Western Reserve University; Cleveland, OH, USA
| |
Collapse
|
9
|
Yang Z. The Principle of Cortical Development and Evolution. Neurosci Bull 2025; 41:461-485. [PMID: 39023844 PMCID: PMC11876516 DOI: 10.1007/s12264-024-01259-2] [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: 05/29/2024] [Accepted: 06/21/2024] [Indexed: 07/20/2024] Open
Abstract
Human's robust cognitive abilities, including creativity and language, are made possible, at least in large part, by evolutionary changes made to the cerebral cortex. This paper reviews the biology and evolution of mammalian cortical radial glial cells (primary neural stem cells) and introduces the concept that a genetically step wise process, based on a core molecular pathway already in use, is the evolutionary process that has molded cortical neurogenesis. The core mechanism, which has been identified in our recent studies, is the extracellular signal-regulated kinase (ERK)-bone morphogenic protein 7 (BMP7)-GLI3 repressor form (GLI3R)-sonic hedgehog (SHH) positive feedback loop. Additionally, I propose that the molecular basis for cortical evolutionary dwarfism, exemplified by the lissencephalic mouse which originated from a larger gyrencephalic ancestor, is an increase in SHH signaling in radial glia, that antagonizes ERK-BMP7 signaling. Finally, I propose that: (1) SHH signaling is not a key regulator of primate cortical expansion and folding; (2) human cortical radial glial cells do not generate neocortical interneurons; (3) human-specific genes may not be essential for most cortical expansion. I hope this review assists colleagues in the field, guiding research to address gaps in our understanding of cortical development and evolution.
Collapse
Affiliation(s)
- Zhengang Yang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurology, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
10
|
Matoba N, McAfee JC, Krupa O, Bell J, Le BD, Valone JM, Crawford GE, Won H, Stein JL. Massively parallel assessment of gene regulatory activity at human cortical structure associated variants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.08.635393. [PMID: 39974944 PMCID: PMC11839127 DOI: 10.1101/2025.02.08.635393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Genetic association studies have identified hundreds of largely non-coding loci associated with inter-individual differences in the structure of the human cortex, though the specific genetic variants that impact regulatory activity are unknown. We implemented a Massively Parallel Reporter Assay (MPRA) to measure the regulatory activity of 9,092 cortical structure associated DNA variants in human neural progenitor cells during Wnt stimulation and at baseline. We identified 918 variants with regulatory potential from 150 cortical structure associated loci (76% of loci studied), of which >50% showed allelic effects. Wnt stimulation modified regulatory activity at a subset of loci that functioned as condition-dependent enhancers. Regulatory activity in MPRA was largely induced by Alu elements that were hypothesized to contribute to cortical expansion. The regionally specific impact of genetic variants that disrupt motifs is likely mediated through the levels of transcription factor expression during development, further clarifying the molecular mechanisms altering cortical structure.
Collapse
Affiliation(s)
- Nana Matoba
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Jessica C. McAfee
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Oleh Krupa
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Jess Bell
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Brandon D. Le
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Jordan M. Valone
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | | | - Hyejung Won
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Jason L. Stein
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| |
Collapse
|
11
|
Glass MR, Matoba N, Beltran AA, Patel NK, Farah TM, Eswar K, Bhargava S, Huang K, Curtin I, Ahmed S, Srivastava M, Drake E, Davis LT, Yeturi M, Sun K, Love MI, Simon JM, St John T, Marrus N, Pandey J, Estes A, Dager S, Schultz RT, Botteron K, Evans A, Kim SH, Styner M, McKinstry RC, Collins DL, Volk H, Benke K, Zwaigenbaum L, Hazlett H, Beltran AS, Girault JB, Shen MD, Piven J, Stein JL. Early cell cycle genes in cortical organoid progenitors predict interindividual variability in infant brain growth trajectories. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.07.637106. [PMID: 39974968 PMCID: PMC11839139 DOI: 10.1101/2025.02.07.637106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Human induced pluripotent stem cell (iPSC) derived cortical organoids (hCOs) model neurogenesis on an individual's genetic background. The degree to which hCO phenotypes recapitulate the brain growth of the participants from which they were derived is not well established. We generated up to 3 iPSC clones from each of 18 participants in the Infant Brain Imaging Study, who have undergone longitudinal brain imaging during infancy. We identified consistent hCO morphology and cortical cell types across clones from the same participant. hCO cross-sectional area and production of cortical hem cells were associated with in vivo cortical growth rates. Cell cycle associated genes expression in early progenitors at the crux of fate decision trajectories were correlated with cortical growth rate from 6-12 months of age, and were enriched in microcephaly and neurodevelopmental disorder genes. Our data suggest the hCOs capture inter-individual variation in cortical cell types influencing infant cortical surface area expansion.
Collapse
Affiliation(s)
- Madison R Glass
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- These authors contributed equally
| | - Nana Matoba
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- These authors contributed equally
| | - Alvaro A Beltran
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Niyanta K Patel
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Tala M Farah
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Karthik Eswar
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Shivam Bhargava
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Karen Huang
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Ian Curtin
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Sara Ahmed
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Mary Srivastava
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Emma Drake
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Liam T Davis
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Meghana Yeturi
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Kexin Sun
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Michael I Love
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- Department of Biostatistics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Jeremy M Simon
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- Present address: Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
- Present address: Department of Biostatistics, Harvard T.H. Chan School of Public Health, MA, USA
| | - Tanya St John
- Department of Speech and Hearing Sciences, University of Washington; Seattle, WA, USA
- University of Washington Autism Center, University of Washington; Seattle, WA, USA
| | - Natasha Marrus
- Department of Psychiatry, Washington University School of Medicine; St. Louis, MO, USA
| | - Juhi Pandey
- Department of Psychiatry, University of Pennsylvania; Philadelphia, PA, USA
- Center for Autism Research, Children's Hospital of Philadelphia; Philadelphia, PA, USA
- Department of Pediatrics, University of Pennsylvania; Philadelphia, USA
| | - Annette Estes
- Department of Speech and Hearing Sciences, University of Washington; Seattle, WA, USA
- University of Washington Autism Center, University of Washington; Seattle, WA, USA
| | - Stephen Dager
- Department of Radiology, University of Washington; Seattle, WA, USA
- Department of Bioengineering, University of Washington; Seattle, WA, USA
| | - Robert T Schultz
- Department of Psychiatry, University of Pennsylvania; Philadelphia, PA, USA
- Center for Autism Research, Children's Hospital of Philadelphia; Philadelphia, PA, USA
- Department of Pediatrics, University of Pennsylvania; Philadelphia, USA
| | - Kelly Botteron
- Department of Psychiatry, Washington University School of Medicine; St. Louis, MO, USA
| | - Alan Evans
- Department of Neurology and Neurosurgery, McGill University; Montreal, QC, CA
- Department of Psychiatry, McGill University; Montreal, QC, CA
- Department of Biomedical Engineering, McGill University; Montreal, QC, CA
| | - Sun Hyung Kim
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Martin Styner
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- Department of Psychiatry, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- Department of Computer Sciences, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Robert C McKinstry
- Department of Psychiatry, Washington University School of Medicine; St. Louis, MO, USA
| | - D Louis Collins
- Department of Neurology and Neurosurgery, McGill University; Montreal, QC, CA
- Department of Biomedical Engineering, McGill University; Montreal, QC, CA
| | - Heather Volk
- School of Public Health, Johns Hopkins; Baltimore, MD, USA
| | - Kelly Benke
- School of Public Health, Johns Hopkins; Baltimore, MD, USA
| | - Lonnie Zwaigenbaum
- Department of Developmental Pediatrics, University of Alberta; Edmonton, AB, CA
| | - Heather Hazlett
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- Department of Psychiatry, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Adriana S Beltran
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Jessica B Girault
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- Department of Psychiatry, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- These authors jointly supervised
| | - Mark D Shen
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- Department of Psychiatry, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- These authors jointly supervised
| | - Joseph Piven
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- Department of Psychiatry, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- These authors jointly supervised
| | - Jason L Stein
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- These authors jointly supervised
- Lead contact
| |
Collapse
|
12
|
Chen S, Shcherbina A, Schafer ST, Mattingly ZA, Ramesh J, Narayanan C, Banerjee S, Heath B, Regester M, Chen I, Thakurela S, Hallmayer J, O'Hara R, Solomon M, Nordahl CW, Amaral DG, Chetty S. Cellular mechanisms of early brain overgrowth in autistic children: elevated levels of GPX4 and resistance to ferroptosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.30.635706. [PMID: 39975145 PMCID: PMC11838294 DOI: 10.1101/2025.01.30.635706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Autistic individuals with disproportionate megalencephaly (ASD-DM), characterized by enlarged brains relative to body height, have higher rates of intellectual disability and face more severe cognitive challenges than autistic children with average brain sizes. The cellular and molecular mechanisms underlying this neurophenotype remain poorly understood. To investigate these mechanisms, we generated human induced pluripotent stem cells from non-autistic typically developing children and autistic children with and without disproportionate megalencephaly. We assessed these children longitudinally from ages two to twelve years using magnetic resonance imaging and comprehensive cognitive and medical evaluations. We show that neural progenitor cells (NPCs) derived from ASD-DM children exhibit increased rates of cell survival and suppressed cell death, accompanied by heightened oxidative stress and ferrous iron accumulation. Despite these stressors, ASD-DM NPCs actively suppress apoptosis and ferroptosis by regulating proteins such as caspase-3 (CASP3), poly(ADP-ribose) polymerase 1 (PARP1), and glutathione peroxidase 4 (GPX4). Cellular ferroptotic signatures are further supported by elevated expression of selenocysteine genes, including GPX4 , in the blood of ASD-DM children and their mothers, suggesting potential hereditary or environmental influences. Furthermore, we show that peripheral expression of GPX4 and other selenocysteine genes correlate with cognitive outcomes (IQ). These findings underscore the role of ferroptosis in autism, pointing to potential diagnostic biomarkers and targets for intervention.
Collapse
|
13
|
Lainšček D, Forstnerič V, Miroševič Š. CTNNB1 syndrome mouse models. Mamm Genome 2025:10.1007/s00335-025-10105-3. [PMID: 39833474 DOI: 10.1007/s00335-025-10105-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 01/09/2025] [Indexed: 01/22/2025]
Abstract
CTNNB1 syndrome is a rare neurodevelopmental disorder, affecting children worldwide with a prevalence of 2.6-3.2 per 100,000 births and often misdiagnosed as cerebral palsy. De novo loss-of-function mutations in the Ctnnb1 gene result in dysfunction of the β-catenin protein, disrupting the canonical Wnt signaling pathway, which plays a key role in cell proliferation, differentiation, and tissue homeostasis. Additionally, these mutations impair the formation of cell junctions, adversely affecting tissue architecture. Motor and speech deficits, cognitive impairment, cardiovascular and visual problems are just some of the key symptoms that occur in CTNNB1 syndrome patients. There is currently no effective treatment option available for patients with CTNNB1 syndrome, with support largely focused on the management of symptoms and physiotherapy, yet recently some therapeutic approaches are being developed. Animal testing is still crucial in the process of new drug development, and mouse models are particularly important. These models provide researchers with new understanding of the disease mechanisms and are invaluable for testing the efficacy and safety of potential treatments. The development of various mouse models with β-catenin loss- and gain-of-function mutations successfully replicates key features of intellectual disability, autism-like behaviors, motor deficits, and more. These models provide a valuable platform for studying disease mechanisms and offer a powerful tool for testing the therapeutic potential and effectiveness of new drug candidates, paving the way for future clinical trials.
Collapse
Affiliation(s)
- Duško Lainšček
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Ljubljana, 1000, Slovenia.
- Centre for Technologies of Gene and Cell Therapy, Ljubljana, 1000, Slovenia.
- EN-FIST Centre of Excellence, Ljubljana, 1000, Slovenia.
| | - Vida Forstnerič
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Ljubljana, 1000, Slovenia.
| | - Špela Miroševič
- The Gene Therapy Research Institute, CTNNB1 Foundation, Ljubljana, 1000, Slovenia.
- Department of Family Medicine, Faculty of Medicine Ljubljana, University of Ljubljana, Ljubljana, 1000, Slovenia.
| |
Collapse
|
14
|
OuYang Y, Yi J, Chen B, Zeng F, Chen X, Yang H, Xu Y, Liu Z, Ning W, Liu B. Mechanism of Buyang Huanwu Decoction mediating Cav1-regulated Wnt pathway to promote neural regeneration in cerebral ischemic mice. JOURNAL OF ETHNOPHARMACOLOGY 2025; 339:119121. [PMID: 39551283 DOI: 10.1016/j.jep.2024.119121] [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: 10/01/2024] [Revised: 11/14/2024] [Accepted: 11/15/2024] [Indexed: 11/19/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Promoting neural repair after cerebral ischemia (CI) is one of the important intervention strategies. Buyang Huanwu Decoction (BHD) is a traditional Chinese medicine prescription commonly used for the treatment of CI.Previous studies by the research group have shown that BHD can promote neural regeneration after CI. The core cause of motor, sensory, and autonomic dysfunction caused by CI injury is neuronal death. Promoting endogenous neural regeneration is of great significance for neural repair after CI. In this context, the Wnt pathway promotes endogenous neural regeneration worthy of attention. AIM OF THE STUDY This study aims to elucidate the mechanism by which BHD promotes neural regeneration after CI, focusing on how it mediates caveolin-1 (Cav1) to regulate the Wnt signaling pathway. MATERIALS AND METHODS Using the middle cerebral artery occlusion (MCAO) technique, a CI model was created. To establish the neuroprotective properties of BHD and determine the ideal therapeutic dosage for CI, neurobehavioral scores and pathological alterations were found across several groups of mice after varying doses of BHD were administered. Furthermore, Cav1 knockout (Cav1-/-) mice were used to confirm Cav1's function in BHD-mediated neuronal regeneration following CI. RESULTS In CI models, BHD was shown to enhance neural regeneration. In vivo research indicate that its mechanism of action is through Cav1 stimulation of the Wnt signaling pathway, which causes related brain-derived trophic factors to be upregulated. CONCLUSION This study confirmed, using knockout mice, that BHD promotes nerve regeneration following CI. The results indicate that Cav1 control of the Wnt signaling pathway is likely the mechanism by which this impact is mediated, so offering insights into the possible mechanism of action of BHD and reaffirming Cav1's function in brain regeneration following CI.
Collapse
Affiliation(s)
- Yin OuYang
- The First Affiliated Hospital, Hunan University of Chinese Medicine, Changsha, 410000, China; Hunan Academy of Chinese Medicine, Changsha, 410000, China; Hunan University of Chinese Medicine, Changsha, 410000, China
| | - Jian Yi
- The First Affiliated Hospital, Hunan University of Chinese Medicine, Changsha, 410000, China
| | - Bowei Chen
- The First Affiliated Hospital, Hunan University of Chinese Medicine, Changsha, 410000, China
| | - Fanzuo Zeng
- The First Affiliated Hospital, Hunan University of Chinese Medicine, Changsha, 410000, China; Hunan University of Chinese Medicine, Changsha, 410000, China
| | - Xuemei Chen
- Hunan University of Chinese Medicine, Changsha, 410000, China
| | - Hua Yang
- The First Affiliated Hospital, Hunan University of Chinese Medicine, Changsha, 410000, China; Hunan University of Chinese Medicine, Changsha, 410000, China
| | - Yaqian Xu
- The First Affiliated Hospital, Hunan University of Chinese Medicine, Changsha, 410000, China
| | - Zhenkui Liu
- Liaoning University of Traditional Chinese Medicine, Shenyang, 116600, China
| | - Wanling Ning
- Hunan University of Chinese Medicine, Changsha, 410000, China
| | - Baiyan Liu
- Hunan Academy of Chinese Medicine, Changsha, 410000, China; Hunan University of Chinese Medicine, Changsha, 410000, China.
| |
Collapse
|
15
|
Rosenblum J, Meuwissen M, Jansen AC, Oegema R, Reddy N, Mankad K, Sudhakar S. Recognisable Neuroradiological Findings in Five Neurogenetic Disorders. Clin Genet 2025; 107:13-22. [PMID: 39462795 DOI: 10.1111/cge.14637] [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: 07/30/2024] [Revised: 09/26/2024] [Accepted: 10/10/2024] [Indexed: 10/29/2024]
Abstract
The rate of discovery and increased understanding of genetic causes for neurodevelopmental disorders has peaked over the past decade. It is well recognised that some genes show marked variability in neuroradiological phenotypes, and inversely, some radiological phenotypes are associated with several different genetic conditions. However, some readily recognisable brain magnetic resonance imaging (MRI) patterns, especially in the context of corresponding associated clinical findings, should prompt consideration of a pathogenic variant in a specific gene or gene pathway. As these conditions can often prove challenging to diagnose, a clinical suspicion of a specific disorder may be invaluable to guide and interpret genetic testing. This review focuses on five neurogenetic syndromes with recognisable brain findings that radiologists, paediatric neurologists, geneticists, and other specialists involved in neurodevelopmental disorders should be able to recognise in order to pinpoint the gene or gene groups involved and delve into their molecular mechanisms. The comprehensively reviewed conditions include DDX3X-related neurodevelopmental disorder, Van Maldergem syndrome, NMDAR-related disorders, EML1-associated disorder and ARFGEF2-related periventricular nodular heterotopia with microcephaly.
Collapse
Affiliation(s)
- Jessica Rosenblum
- Center of Clinical Genetics, Antwerp University Hospital, University of Antwerp, Edegem, Belgium
- Translational Neurosciences, University of Antwerp, Edegem, Belgium
| | - Marije Meuwissen
- Center of Clinical Genetics, Antwerp University Hospital, University of Antwerp, Edegem, Belgium
| | - Anna C Jansen
- Translational Neurosciences, University of Antwerp, Edegem, Belgium
- Department of Pediatric Neurology, Antwerp University Hospital, University of Antwerp, Edegem, Belgium
| | - Renske Oegema
- Department of Genetics, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Nihaal Reddy
- Rainbow Children's Hospital and Tenet Diagnostics, Hyderabad, India
| | - Kshitij Mankad
- Department of Radiology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Sniya Sudhakar
- Department of Radiology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| |
Collapse
|
16
|
He L, Bhat K, Ioannidis A, Pajonk F. Effects of dopamine receptor antagonists and radiation on mouse neural stem/progenitor cells. Radiother Oncol 2024; 201:110562. [PMID: 39341503 PMCID: PMC11987595 DOI: 10.1016/j.radonc.2024.110562] [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: 02/21/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND Dopamine receptor antagonists have recently been identified as potential anti-cancer agents in combination with radiation, and a first drug of this class is in clinical trials against pediatric glioma. Radiotherapy causes cognitive impairment primarily by eliminating neural stem/progenitor cells and subsequent loss of neurogenesis, along with inducing inflammation, vascular damage, and synaptic alterations. Here, we tested the combined effects of dopamine receptor antagonists and radiation on neural stem/progenitor cells. METHODS Using transgenic mice that report the presence of neural stem/progenitor cells through Nestin promoter-driven expression of EGFP, the effects of dopamine receptor antagonists alone or in combination with radiation on neural stem/progenitor cells were assessed in sphere-formation assays, extreme limiting dilution assays, flow cytometry and real-time PCR in vitro and in vivo in both sexes. RESULTS We report that hydroxyzine and trifluoperazine exhibited sex-dependent effects on murine newborn neural stem/progenitor cells in vitro. In contrast, amisulpride, nemonapride, and quetiapine, when combined with radiation, significantly increased the number of neural stem/progenitor cells in both sexes. In vivo, trifluoperazine showed sex-dependent effects on adult neural stem/progenitor cells, while amisulpride demonstrated significant effects in both sexes. Further, amisulpride increased sphere forming capacity and stem cell frequency in both sexes when compared to controls. CONCLUSION We conclude that a therapeutic window for dopamine receptor antagonists in combination with radiation potentially exists, making it a novel combination therapy against glioblastoma. Normal tissue toxicity following this treatment scheme likely differs depending on age and sex and should be taken into consideration when designing clinical trials.
Collapse
Affiliation(s)
- Ling He
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, United States.
| | - Kruttika Bhat
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, United States
| | - Angeliki Ioannidis
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, United States
| | - Frank Pajonk
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, United States; Jonsson Comprehensive Cancer Center at UCLA, United States; Department of Neurosurgery, David Geffen School of Medicine at UCLA, United States
| |
Collapse
|
17
|
Matoba N, Le BD, Valone JM, Wolter JM, Mory JT, Liang D, Aygün N, Broadaway KA, Bond ML, Mohlke KL, Zylka MJ, Love MI, Stein JL. Stimulating Wnt signaling reveals context-dependent genetic effects on gene regulation in primary human neural progenitors. Nat Neurosci 2024; 27:2430-2442. [PMID: 39349663 PMCID: PMC11633645 DOI: 10.1038/s41593-024-01773-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 08/28/2024] [Indexed: 10/09/2024]
Abstract
Gene regulatory effects have been difficult to detect at many non-coding loci associated with brain-related traits, likely because some genetic variants have distinct functions in specific contexts. To explore context-dependent gene regulation, we measured chromatin accessibility and gene expression after activation of the canonical Wnt pathway in primary human neural progenitors (n = 82 donors). We found that TCF/LEF motifs and brain-structure-associated and neuropsychiatric-disorder-associated variants were enriched within Wnt-responsive regulatory elements. Genetically influenced regulatory elements were enriched in genomic regions under positive selection along the human lineage. Wnt pathway stimulation increased detection of genetically influenced regulatory elements/genes by 66%/53% and enabled identification of 397 regulatory elements primed to regulate gene expression. Stimulation also increased identification of shared genetic effects on molecular and complex brain traits by up to 70%, suggesting that genetic variant function during neurodevelopmental patterning can lead to differences in adult brain and behavioral traits.
Collapse
Affiliation(s)
- Nana Matoba
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Brandon D Le
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jordan M Valone
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Justin M Wolter
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Carolina Institute for Developmental Disabilities, Carrboro, NC, USA
| | - Jessica T Mory
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Dan Liang
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Nil Aygün
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - K Alaine Broadaway
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Marielle L Bond
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Mark J Zylka
- UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Carolina Institute for Developmental Disabilities, Carrboro, NC, USA
| | - Michael I Love
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jason L Stein
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Carolina Institute for Developmental Disabilities, Carrboro, NC, USA.
| |
Collapse
|
18
|
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
|
19
|
Sandran NG, Badawi N, Gecz J, van Eyk CL. Cerebral palsy as a childhood-onset neurological disorder caused by both genetic and environmental factors. Semin Fetal Neonatal Med 2024; 29:101551. [PMID: 39523172 DOI: 10.1016/j.siny.2024.101551] [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] [Indexed: 11/16/2024]
Abstract
Cerebral palsy (CP) is a clinical term used to describe a spectrum of movement and posture disorders resulting from non-progressive disturbances in the developing fetal brain. The clinical diagnosis of CP does not include pathological or aetiological defining features, therefore both genetic and environmental causal pathways are encompassed under the CP diagnostic umbrella. In this review, we explore several genetic causal pathways, including both monogenic and polygenic risks, and present evidence supporting the multifactorial contributions to CP. Historically, CP has been associated with various risk factors such as pre-term birth, multiple gestation, intrauterine growth restriction (IUGR), maternal infection, and perinatal asphyxia. Thus, we also examine genetic predispositions that may contribute to these risk factors. Understanding the specific aetiology of CP enables more tailored treatments, especially with the increasing potential for early genetic testing.
Collapse
Affiliation(s)
- Nandini G Sandran
- Neurogenetics Research Program, Adelaide Medical School, University of Adelaide, Adelaide, Australia; Australian Collaborative Cerebral Palsy Research Group, Robinson Research Institute, University of Adelaide, Adelaide, Australia
| | - Nadia Badawi
- Children's Hospital Westmead Clinical School, University of Sydney, Sydney, Australia; Grace Centre for Newborn Intensive Care, The Children's Hospital Westmead, Sydney, Australia; Discipline of Child and Adolescent Health, Cerebral Palsy Alliance Research Institute, University of Sydney, Sydney, Australia
| | - Jozef Gecz
- Neurogenetics Research Program, Adelaide Medical School, University of Adelaide, Adelaide, Australia; Australian Collaborative Cerebral Palsy Research Group, Robinson Research Institute, University of Adelaide, Adelaide, Australia; South Australian Health and Medical Research Institute, Adelaide, Australia.
| | - Clare L van Eyk
- Neurogenetics Research Program, Adelaide Medical School, University of Adelaide, Adelaide, Australia; Australian Collaborative Cerebral Palsy Research Group, Robinson Research Institute, University of Adelaide, Adelaide, Australia
| |
Collapse
|
20
|
Jang S, Abarinov E, Dobkin J, Wichterle H. Independent control of neurogenesis and dorsoventral patterning by NKX2-2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.13.618113. [PMID: 39415990 PMCID: PMC11482905 DOI: 10.1101/2024.10.13.618113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Human neurogenesis is disproportionately protracted, lasting >10 times longer than in mouse, allowing neural progenitors to undergo more rounds of self-renewing cell divisions and generate larger neuronal populations. In the human spinal cord, expansion of the motor neuron lineage is achieved through a newly evolved progenitor domain called vpMN (ventral motor neuron progenitor) that uniquely extends and expands motor neurogenesis. This behavior of vpMNs is controlled by transcription factor NKX2-2, which in vpMNs is co-expressed with classical motor neuron progenitor (pMN) marker OLIG2. In this study, we sought to determine the molecular basis of NKX2-2-mediated extension and expansion of motor neurogenesis. We found that NKX2-2 represses proneural gene NEUROG2 by two distinct, Notch-independent mechanisms that are respectively apparent in rodent and human spinal progenitors: in rodents (and chick), NKX2-2 represses Olig2 and the motor neuron lineage through its tinman domain, leading to loss of Neurog2 expression. In human vpMNs, however, NKX2-2 represses NEUROG2 but not OLIG2, thereby allowing motor neurogenesis to proceed, albeit in a delayed and protracted manner. Interestingly, we found that ectopic expression of tinman-mutant Nkx2-2 in mouse pMNs phenocopies human vpMNs, repressing Neurog2 but not Olig2, and leading to delayed and protracted motor neurogenesis. Our studies identify a Notch- and tinman-independent mode of Nkx2-2-mediated Neurog2 repression that is observed in human spinal progenitors, but is normally masked in rodents and chicks due to Nkx2-2's tinman-dependent repression of Olig2.
Collapse
Affiliation(s)
- Sumin Jang
- Departments of Pathology and Cell Biology, Neuroscience, and Neurology, Center for Motor Neuron Biology and Disease, Columbia Stem Cell Initiative, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Elena Abarinov
- Departments of Pathology and Cell Biology, Neuroscience, and Neurology, Center for Motor Neuron Biology and Disease, Columbia Stem Cell Initiative, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Julie Dobkin
- Departments of Pathology and Cell Biology, Neuroscience, and Neurology, Center for Motor Neuron Biology and Disease, Columbia Stem Cell Initiative, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Hynek Wichterle
- Departments of Pathology and Cell Biology, Neuroscience, and Neurology, Center for Motor Neuron Biology and Disease, Columbia Stem Cell Initiative, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
21
|
Alonso-Olivares H, Marques MM, Prieto-Colomina A, López-Ferreras L, Martínez-García N, Vázquez-Jiménez A, Borrell V, Marin MC, Fernandez-Alonso R. Mouse cortical organoids reveal key functions of p73 isoforms: TAp73 governs the establishment of the archetypical ventricular-like zones while DNp73 is central in the regulation of neural cell fate. Front Cell Dev Biol 2024; 12:1464932. [PMID: 39376628 PMCID: PMC11456701 DOI: 10.3389/fcell.2024.1464932] [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: 07/15/2024] [Accepted: 09/04/2024] [Indexed: 10/09/2024] Open
Abstract
Introduction Neurogenesis is tightly regulated in space and time, ensuring the correct development and organization of the central nervous system. Critical regulators of brain development and morphogenesis in mice include two members of the p53 family: p53 and p73. However, dissecting the in vivo functions of these factors and their various isoforms in brain development is challenging due to their pleiotropic effects. Understanding their role, particularly in neurogenesis and brain morphogenesis, requires innovative experimental approaches. Methods To address these challenges, we developed an efficient and highly reproducible protocol to generate mouse brain organoids from pluripotent stem cells. These organoids contain neural progenitors and neurons that self-organize into rosette-like structures resembling the ventricular zone of the embryonic forebrain. Using this model, we generated organoids from p73-deficient mouse cells to investigate the roles of p73 and its isoforms (TA and DNp73) during brain development. Results and Discussion Organoids derived from p73-deficient cells exhibited increased neuronal apoptosis and reduced neural progenitor proliferation, linked to compensatory activation of p53. This closely mirrors previous in vivo observations, confirming that p73 plays a pivotal role in brain development. Further dissection of p73 isoforms function revealed a dual role of p73 in regulating brain morphogenesis, whereby TAp73 controls transcriptional programs essential for the establishment of the neurogenic niche structure, while DNp73 is responsible for the precise and timely regulation of neural cell fate. These findings highlight the distinct roles of p73 isoforms in maintaining the balance of neural progenitor cell biology, providing a new understanding of how p73 regulates brain morphogenesis.
Collapse
Affiliation(s)
- Hugo Alonso-Olivares
- Instituto de Biomedicina and Departamento de Biología Molecular, Universidad de León, León, Spain
| | - Margarita M. Marques
- Instituto de Desarrollo Ganadero y Sanidad Animal and Departamento de Producción Animal, Universidad de León, León, Spain
| | - Anna Prieto-Colomina
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernández, Alicante, Spain
| | - Lorena López-Ferreras
- Instituto de Biomedicina and Departamento de Biología Molecular, Universidad de León, León, Spain
| | - Nicole Martínez-García
- Instituto de Biomedicina and Departamento de Producción Animal, Universidad de León, León, Spain
| | - Alberto Vázquez-Jiménez
- Instituto de Biomedicina and Departamento de Biología Molecular, Universidad de León, León, Spain
| | - Victor Borrell
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernández, Alicante, Spain
| | - Maria C. Marin
- Instituto de Biomedicina and Departamento de Biología Molecular, Universidad de León, León, Spain
| | - Rosalia Fernandez-Alonso
- Instituto de Biomedicina and Departamento de Biología Molecular, Universidad de León, León, Spain
| |
Collapse
|
22
|
Zhu Y, Huang Y, Tang T, Xie Y. HDAC1 and HDAC2 orchestrate Wnt signaling to regulate neural progenitor transition during brain development. iScience 2024; 27:110600. [PMID: 39224519 PMCID: PMC11367519 DOI: 10.1016/j.isci.2024.110600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 06/19/2024] [Accepted: 07/24/2024] [Indexed: 09/04/2024] Open
Abstract
Tightly controlled neurogenesis is crucial for generating the precise number of neurons and establishing the intricate architecture of the cortex, with deficiencies often leading to neurodevelopmental disorders. Neuroepithelial progenitors (NPs) transit into radial glial progenitors (RGPs) to initiate neural differentiation, yet the governing mechanisms remain elusive. Here, we found that histone deacetylases 1 and 2 (HDAC1/2) mediated suppression of Wnt signaling is essential for the NP-to-RGP transition. Conditional depletion of HDAC1/2 from NPs upregulated Wnt signaling genes, impairing the transition to RGPs and resulting in rosette structures within the neocortex. Multi-omics analysis revealed that HDAC1/2 are critical for downregulating Wnt signaling, identifying Wnt9a as a key target. Overexpression of Wnt9a led to an increased population of NPs and the disruption of cortical organization. Notably, Wnt inhibitor administration partially rescued the disrupted cortical architecture. Our findings reveal the significance of tightly controlled Wnt signaling through epigenetic mechanisms in neocortical development.
Collapse
Affiliation(s)
- Yue Zhu
- Department of Anesthesia, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yunyun Huang
- Department of Anesthesia, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Tianxiang Tang
- Department of Anesthesia, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yunli Xie
- Department of Anesthesia, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
23
|
Nieto-Estevez V, Varma P, Mirsadeghi S, Caballero J, Gamero-Alameda S, Hosseini A, Silvosa MJ, Thodeson DM, Lybrand ZR, Giugliano M, Navara C, Hsieh J. Dual effects of ARX poly-alanine mutations in human cortical and interneuron development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.25.577271. [PMID: 38328230 PMCID: PMC10849640 DOI: 10.1101/2024.01.25.577271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Infantile spasms, with an incidence of 1.6 to 4.5 per 10,000 live births, are a relentless and devastating childhood epilepsy marked by severe seizures but also leads to lifelong intellectual disability. Alarmingly, up to 5% of males with this condition carry a mutation in the Aristaless-related homeobox ( ARX ) gene. Our current lack of human-specific models for developmental epilepsy, coupled with discrepancies between animal studies and human data, underscores the gap in knowledge and urgent need for innovative human models, organoids being one of the best available. Here, we used human neural organoid models, cortical organoids (CO) and ganglionic eminences organoids (GEO) which mimic cortical and interneuron development respectively, to study the consequences of PAE mutations, one of the most prevalent mutation in ARX . ARX PAE produces a decrease expression of ARX in GEOs, and an enhancement in interneuron migration. That accelerated migration is cell autonomously driven, and it can be rescued by inhibiting CXCR4. We also found that PAE mutations result in an early increase in radial glia cells and intermediate progenitor cells, followed by a subsequent loss of cortical neurons at later timepoints. Moreover, ARX expression is upregulated in COs derived from patients at 30 DIV and is associated with alterations in the expression of CDKN1C . Furthermore, ARX PAE assembloids had hyperactivity which were evident at early stages of development. With effective treatments for infantile spasms and developmental epilepsies still elusive, delving into the role of ARX PAE mutations in human brain organoids represents a pivotal step toward uncovering groundbreaking therapeutic strategies.
Collapse
|
24
|
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
|
25
|
Sanchez-Ruiz JA, Treviño-Alvarez AM, Zambrano-Lucio M, Lozano Díaz ST, Wang N, Biernacka JM, Tye SJ, Cuellar-Barboza AB. The Wnt signaling pathway in major depressive disorder: A systematic review of human studies. Psychiatry Res 2024; 339:115983. [PMID: 38870775 DOI: 10.1016/j.psychres.2024.115983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 05/20/2024] [Accepted: 05/26/2024] [Indexed: 06/15/2024]
Abstract
Despite uncertainty about the specific molecular mechanisms driving major depressive disorder (MDD), the Wnt signaling pathway stands out as a potentially influential factor in the pathogenesis of MDD. Known for its role in intercellular communication, cell proliferation, and fate, Wnt signaling has been implicated in diverse biological phenomena associated with MDD, spanning neurodevelopmental to neurodegenerative processes. In this systematic review, we summarize the functional differences in protein and gene expression of the Wnt signaling pathway, and targeted genetic association studies, to provide an integrated synthesis of available human data examining Wnt signaling in MDD. Thirty-three studies evaluating protein expression (n = 15), gene expression (n = 9), or genetic associations (n = 9) were included. Only fifteen demonstrated a consistently low overall risk of bias in selection, comparability, and exposure. We found conflicting observations of limited and distinct Wnt signaling components across diverse tissue sources. These data do not demonstrate involvement of Wnt signaling dysregulation in MDD. Given the well-established role of Wnt signaling in antidepressant response, we propose that a more targeted and functional assessment of Wnt signaling is needed to understand its role in depression pathophysiology. Future studies should include more components, assess multiple tissues concurrently, and follow a standardized approach.
Collapse
Affiliation(s)
- Jorge A Sanchez-Ruiz
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, MN, USA; Department of Psychiatry, Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | | | | | - Sofía T Lozano Díaz
- Vicerrectoría de Ciencias de la Salud, Universidad de Monterrey, San Pedro Garza Garcia, Nuevo Leon, Mexico
| | - Ning Wang
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Joanna M Biernacka
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, MN, USA; Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Susannah J Tye
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, MN, USA; Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia; Department of Psychiatry & Behavioral Sciences, Emory University, Atlanta, GA, USA; Department of Psychiatry, University of Minnesota, Minneapolis, MN, USA
| | - Alfredo B Cuellar-Barboza
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, MN, USA; Department of Psychiatry, Universidad Autónoma de Nuevo León, Monterrey, Mexico.
| |
Collapse
|
26
|
Zhu F, He P, Jiang W, Afridi SK, Xu H, Alahmad M, Alvin Huang YW, Qiu W, Wang G, Tang C. Astrocyte-secreted C3 signaling impairs neuronal development and cognition in autoimmune diseases. Prog Neurobiol 2024; 240:102654. [PMID: 38945516 DOI: 10.1016/j.pneurobio.2024.102654] [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: 12/09/2023] [Revised: 05/05/2024] [Accepted: 06/24/2024] [Indexed: 07/02/2024]
Abstract
Neuromyelitis optica (NMO) arises from primary astrocytopathy induced by autoantibodies targeting the astroglial protein aquaporin 4 (AQP4), leading to severe neurological sequelae such as vision loss, motor deficits, and cognitive decline. Mounting evidence has shown that dysregulated activation of complement components contributes to NMO pathogenesis. Complement C3 deficiency has been shown to protect against hippocampal neurodegeneration and cognitive decline in neurodegenerative disorders (e.g., Alzheimer's disease, AD) and autoimmune diseases (e.g., multiple sclerosis, MS). However, whether inhibiting the C3 signaling can ameliorate cognitive dysfunctions in NMO remains unclear. In this study, we found that the levels of C3a, a split product of C3, significantly correlate with cognitive impairment in our patient cohort. In response to the stimulation of AQP4 autoantibodies, astrocytes were activated to secrete complement C3, which inhibited the development of cultured neuronal dendritic arborization. NMO mouse models exhibited reduced adult hippocampal newborn neuronal dendritic and spine development, as well as impaired learning and memory functions, which could be rescued by decreasing C3 levels in astrocytes. Mechanistically, we found that C3a engaged with C3aR to impair neuronal development by dampening β-catenin signalling. Additionally, inhibition of the C3-C3aR-GSK3β/β-catenin cascade restored neuronal development and ameliorated cognitive impairments. Collectively, our results suggest a pivotal role of the activation of the C3-C3aR network in neuronal development and cognition through mediating astrocyte and adult-born neuron communication, which represents a potential therapeutic target for autoimmune-related cognitive impairment diseases.
Collapse
Affiliation(s)
- Fan Zhu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, Guangdong Province 510630, China
| | - Pengyan He
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, Guangdong Province 510630, China
| | - Wei Jiang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, Guangdong Province 510630, China
| | - Shabbir Khan Afridi
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; China Graduate School, University of Chinese Academy of Sciences, Beijing, China
| | - Huiming Xu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, Guangdong Province 510630, China
| | - Maali Alahmad
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Yu-Wen Alvin Huang
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, 70 Ship Street, Providence, RI 02903, United States
| | - Wei Qiu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, Guangdong Province 510630, China
| | - Guangyou Wang
- Department of Neurology, First Affiliated Clinical Hospital of Harbin Medical University, and Department of Neurobiology, Harbin Medical University, Harbin 150081, China.
| | - Changyong Tang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, Guangdong Province 510630, China.
| |
Collapse
|
27
|
Geng Z, Tai YT, Wang Q, Gao Z. AUTS2 disruption causes neuronal differentiation defects in human cerebral organoids through hyperactivation of the WNT/β-catenin pathway. Sci Rep 2024; 14:19522. [PMID: 39174599 PMCID: PMC11341827 DOI: 10.1038/s41598-024-69912-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 08/09/2024] [Indexed: 08/24/2024] Open
Abstract
Individuals with the Autism Susceptibility Candidate 2 (AUTS2) gene disruptions exhibit symptoms such as intellectual disability, microcephaly, growth retardation, and distinct skeletal and facial differences. The role of AUTS2 in neurodevelopment has been investigated using animal and embryonic stem cell models. However, the precise molecular mechanisms of how AUTS2 influences neurodevelopment, particularly in humans, are not thoroughly understood. Our study employed a 3D human cerebral organoid culture system, in combination with genetic, genomic, cellular, and molecular approaches, to investigate how AUTS2 impacts neurodevelopment through cellular signaling pathways. We used CRISPR/Cas9 technology to create AUTS2-deficient human embryonic stem cells and then generated cerebral organoids with these cells. Our transcriptomic analyses revealed that the absence of AUTS2 in cerebral organoids reduces the populations of cells committed to the neuronal lineage, resulting in an overabundance of cells with a transcription profile resembling that of choroid plexus (ChP) cells. Intriguingly, we found that AUTS2 negatively regulates the WNT/β-catenin signaling pathway, evidenced by its overactivation in AUTS2-deficient cerebral organoids and in luciferase reporter cells lacking AUTS2. Importantly, treating the AUTS2-deficient cerebral organoids with a WNT inhibitor reversed the overexpression of ChP genes and increased the downregulated neuronal gene expression. This study offers new insights into the role of AUTS2 in neurodevelopment and suggests potential targeted therapies for neurodevelopmental disorders.
Collapse
Affiliation(s)
- Zhuangzhuang Geng
- Department of Biochemistry and Molecular Biology, Penn State Hershey Cancer Institute, The Stem Cell and Regenerative Biology Program, Penn State College of Medicine, Hershey, USA
| | - Yen Teng Tai
- Department of Biochemistry and Molecular Biology, Penn State Hershey Cancer Institute, The Stem Cell and Regenerative Biology Program, Penn State College of Medicine, Hershey, USA
| | - Qiang Wang
- Department of Biochemistry and Molecular Biology, Penn State Hershey Cancer Institute, The Stem Cell and Regenerative Biology Program, Penn State College of Medicine, Hershey, USA
| | - Zhonghua Gao
- Department of Biochemistry and Molecular Biology, Penn State Hershey Cancer Institute, The Stem Cell and Regenerative Biology Program, Penn State College of Medicine, Hershey, USA.
| |
Collapse
|
28
|
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
|
29
|
Li K, Chen Z, Chang X, Xue R, Wang H, Guo W. Wnt signaling pathway in spinal cord injury: from mechanisms to potential applications. Front Mol Neurosci 2024; 17:1427054. [PMID: 39114641 PMCID: PMC11303303 DOI: 10.3389/fnmol.2024.1427054] [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: 05/02/2024] [Accepted: 07/15/2024] [Indexed: 08/10/2024] Open
Abstract
Spinal cord injury (SCI) denotes damage to both the structure and function of the spinal cord, primarily manifesting as sensory and motor deficits caused by disruptions in neural transmission pathways, potentially culminating in irreversible paralysis. Its pathophysiological processes are complex, with numerous molecules and signaling pathways intricately involved. Notably, the pronounced upregulation of the Wnt signaling pathway post-SCI holds promise for neural regeneration and repair. Activation of the Wnt pathway plays a crucial role in neuronal differentiation, axonal regeneration, local neuroinflammatory responses, and cell apoptosis, highlighting its potential as a therapeutic target for treating SCI. However, excessive activation of the Wnt pathway can also lead to negative effects, highlighting the need for further investigation into its applicability and significance in SCI. This paper provides an overview of the latest research advancements in the Wnt signaling pathway in SCI, summarizing the recent progress in treatment strategies associated with the Wnt pathway and analyzing their advantages and disadvantages. Additionally, we offer insights into the clinical application of the Wnt signaling pathway in SCI, along with prospective avenues for future research direction.
Collapse
Affiliation(s)
| | | | | | | | - Huaibo Wang
- Department of Spine Surgery, The Second Hospital Affiliated to Guangdong Medical University, Zhanjiang, China
| | | |
Collapse
|
30
|
Mahon S. Variation and convergence in the morpho-functional properties of the mammalian neocortex. Front Syst Neurosci 2024; 18:1413780. [PMID: 38966330 PMCID: PMC11222651 DOI: 10.3389/fnsys.2024.1413780] [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: 04/07/2024] [Accepted: 06/03/2024] [Indexed: 07/06/2024] Open
Abstract
Man's natural inclination to classify and hierarchize the living world has prompted neurophysiologists to explore possible differences in brain organisation between mammals, with the aim of understanding the diversity of their behavioural repertoires. But what really distinguishes the human brain from that of a platypus, an opossum or a rodent? In this review, we compare the structural and electrical properties of neocortical neurons in the main mammalian radiations and examine their impact on the functioning of the networks they form. We discuss variations in overall brain size, number of neurons, length of their dendritic trees and density of spines, acknowledging their increase in humans as in most large-brained species. Our comparative analysis also highlights a remarkable consistency, particularly pronounced in marsupial and placental mammals, in the cell typology, intrinsic and synaptic electrical properties of pyramidal neuron subtypes, and in their organisation into functional circuits. These shared cellular and network characteristics contribute to the emergence of strikingly similar large-scale physiological and pathological brain dynamics across a wide range of species. These findings support the existence of a core set of neural principles and processes conserved throughout mammalian evolution, from which a number of species-specific adaptations appear, likely allowing distinct functional needs to be met in a variety of environmental contexts.
Collapse
Affiliation(s)
- Séverine Mahon
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, Paris, France
| |
Collapse
|
31
|
Sung CYW, Li M, Jonjic S, Sanchez V, Britt WJ. Cytomegalovirus infection lengthens the cell cycle of granule cell precursors during postnatal cerebellar development. JCI Insight 2024; 9:e175525. [PMID: 38855871 PMCID: PMC11382886 DOI: 10.1172/jci.insight.175525] [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: 09/07/2023] [Accepted: 04/23/2024] [Indexed: 06/11/2024] Open
Abstract
Human cytomegalovirus (HCMV) infection in infants infected in utero can lead to a variety of neurodevelopmental disorders. However, mechanisms underlying altered neurodevelopment in infected infants remain poorly understood. We have previously described a murine model of congenital HCMV infection in which murine CMV (MCMV) spreads hematogenously and establishes a focal infection in all regions of the brain of newborn mice, including the cerebellum. Infection resulted in disruption of cerebellar cortical development characterized by reduced cerebellar size and foliation. This disruption was associated with altered cell cycle progression of the granule cell precursors (GCPs), which are the progenitors that give rise to granule cells (GCs), the most abundant neurons in the cerebellum. In the current study, we have demonstrated that MCMV infection leads to prolonged GCP cell cycle, premature exit from the cell cycle, and reduced numbers of GCs resulting in cerebellar hypoplasia. Treatment with TNF-α neutralizing antibody partially normalized the cell cycle alterations of GCPs and altered cerebellar morphogenesis induced by MCMV infection. Collectively, our results argue that virus-induced inflammation altered the cell cycle of GCPs resulting in a reduced numbers of GCs and cerebellar cortical hypoplasia, thus providing a potential mechanism for altered neurodevelopment in fetuses infected with HCMV.
Collapse
Affiliation(s)
- Cathy Yea Won Sung
- Department of Microbiology, University of Alabama at Birmingham, School of Medicine, Birmingham, Alabama, USA
- Laboratory of Hearing Biology and Therapeutics, National Institute on Deafness and Other Communication Disorders (NIDCD), NIH, Bethesda, Maryland, USA
| | - Mao Li
- Department of Pediatrics, University of Alabama at Birmingham, School of Medicine, Birmingham, Alabama, USA
| | - Stipan Jonjic
- Department of Histology and Embryology and
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Veronica Sanchez
- Department of Pediatrics, University of Alabama at Birmingham, School of Medicine, Birmingham, Alabama, USA
| | - William J Britt
- Department of Microbiology, University of Alabama at Birmingham, School of Medicine, Birmingham, Alabama, USA
- Department of Pediatrics, University of Alabama at Birmingham, School of Medicine, Birmingham, Alabama, USA
- Department of Neurobiology, University of Alabama at Birmingham, School of Medicine, Birmingham, Alabama, USA
| |
Collapse
|
32
|
Bahar N, Cler GJ, Krishnan S, Asaridou SS, Smith HJ, Willis HE, Healy MP, Watkins KE. Differences in Cortical Surface Area in Developmental Language Disorder. NEUROBIOLOGY OF LANGUAGE (CAMBRIDGE, MASS.) 2024; 5:288-314. [PMID: 38832358 PMCID: PMC11093399 DOI: 10.1162/nol_a_00127] [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: 05/14/2023] [Accepted: 11/08/2023] [Indexed: 06/05/2024]
Abstract
Approximately 7% of children have developmental language disorder (DLD), a neurodevelopmental condition associated with persistent language learning difficulties without a known cause. Our understanding of the neurobiological basis of DLD is limited. Here, we used FreeSurfer to investigate cortical surface area and thickness in a large cohort of 156 children and adolescents aged 10-16 years with a range of language abilities, including 54 with DLD, 28 with a history of speech-language difficulties who did not meet criteria for DLD, and 74 age-matched controls with typical language development (TD). We also examined cortical asymmetries in DLD using an automated surface-based technique. Relative to the TD group, those with DLD showed smaller surface area bilaterally in the inferior frontal gyrus extending to the anterior insula, in the posterior temporal and ventral occipito-temporal cortex, and in portions of the anterior cingulate and superior frontal cortex. Analysis of the whole cohort using a language proficiency factor revealed that language ability correlated positively with surface area in similar regions. There were no differences in cortical thickness, nor in asymmetry of these cortical metrics between TD and DLD. This study highlights the importance of distinguishing between surface area and cortical thickness in investigating the brain basis of neurodevelopmental disorders and suggests the development of cortical surface area to be of importance to DLD. Future longitudinal studies are required to understand the developmental trajectory of these cortical differences in DLD and how they relate to language maturation.
Collapse
Affiliation(s)
- Nilgoun Bahar
- Department of Experimental Psychology & Wellcome Trust Centre for Integrative Neuroimaging, University of Oxford, Oxford, UK
| | - Gabriel J. Cler
- Department of Experimental Psychology & Wellcome Trust Centre for Integrative Neuroimaging, University of Oxford, Oxford, UK
- Department of Speech & Hearing Sciences, University of Washington, Seattle, WA, USA
| | - Saloni Krishnan
- Department of Experimental Psychology & Wellcome Trust Centre for Integrative Neuroimaging, University of Oxford, Oxford, UK
- Department of Psychology, Royal Holloway, University of London, Egham Hill, Surrey, UK
| | - Salomi S. Asaridou
- Department of Experimental Psychology & Wellcome Trust Centre for Integrative Neuroimaging, University of Oxford, Oxford, UK
| | - Harriet J. Smith
- Department of Experimental Psychology & Wellcome Trust Centre for Integrative Neuroimaging, University of Oxford, Oxford, UK
- MRC Cognition & Brain Sciences Unit, University of Cambridge, Cambridge, UK
| | - Hanna E. Willis
- Department of Experimental Psychology & Wellcome Trust Centre for Integrative Neuroimaging, University of Oxford, Oxford, UK
- Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford, UK
| | - Máiréad P. Healy
- Department of Experimental Psychology & Wellcome Trust Centre for Integrative Neuroimaging, University of Oxford, Oxford, UK
- Department of Psychology, University of Cambridge, Cambridge, UK
| | - Kate E. Watkins
- Department of Experimental Psychology & Wellcome Trust Centre for Integrative Neuroimaging, University of Oxford, Oxford, UK
| |
Collapse
|
33
|
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
|
34
|
Liu J, Supekar K, El-Said D, de los Angeles C, Zhang Y, Chang H, Menon V. Neuroanatomical, transcriptomic, and molecular correlates of math ability and their prognostic value for predicting learning outcomes. SCIENCE ADVANCES 2024; 10:eadk7220. [PMID: 38820151 PMCID: PMC11141625 DOI: 10.1126/sciadv.adk7220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 04/29/2024] [Indexed: 06/02/2024]
Abstract
Foundational mathematical abilities, acquired in early childhood, are essential for success in our technology-driven society. Yet, the neurobiological mechanisms underlying individual differences in children's mathematical abilities and learning outcomes remain largely unexplored. Leveraging one of the largest multicohort datasets from children at a pivotal stage of knowledge acquisition, we first establish a replicable mathematical ability-related imaging phenotype (MAIP). We then show that brain gene expression profiles enriched for candidate math ability-related genes, neuronal signaling, synaptic transmission, and voltage-gated potassium channel activity contributed to the MAIP. Furthermore, the similarity between MAIP gene expression signatures and brain structure, acquired before intervention, predicted learning outcomes in two independent math tutoring cohorts. These findings advance our knowledge of the interplay between neuroanatomical, transcriptomic, and molecular mechanisms underlying mathematical ability and reveal predictive biomarkers of learning. Our findings have implications for the development of personalized education and interventions.
Collapse
Affiliation(s)
- Jin Liu
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kaustubh Supekar
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Dawlat El-Said
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Carlo de los Angeles
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Yuan Zhang
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Hyesang Chang
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Vinod Menon
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
35
|
Song Y, Jiang W, Afridi SK, Wang T, Zhu F, Xu H, Nazir FH, Liu C, Wang Y, Long Y, Huang YWA, Qiu W, Tang C. Astrocyte-derived CHI3L1 signaling impairs neurogenesis and cognition in the demyelinated hippocampus. Cell Rep 2024; 43:114226. [PMID: 38733586 DOI: 10.1016/j.celrep.2024.114226] [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: 12/14/2023] [Revised: 03/15/2024] [Accepted: 04/26/2024] [Indexed: 05/13/2024] Open
Abstract
Cognitive dysfunction is a feature in multiple sclerosis (MS), a chronic inflammatory demyelinating disorder. A notable aspect of MS brains is hippocampal demyelination, which is closely associated with cognitive decline. However, the mechanisms underlying this phenomenon remain unclear. Chitinase-3-like (CHI3L1), secreted by activated astrocytes, has been identified as a biomarker for MS progression. Our study investigates CHI3L1's function within the demyelinating hippocampus and demonstrates a correlation between CHI3L1 expression and cognitive impairment in patients with MS. Activated astrocytes release CHI3L1 in reaction to induced demyelination, which adversely affects the proliferation and differentiation of neural stem cells and impairs dendritic growth, complexity, and spine formation in neurons. Our findings indicate that the astrocytic deletion of CHI3L1 can mitigate neurogenic deficits and cognitive dysfunction. We showed that CHI3L1 interacts with CRTH2/receptor for advanced glycation end (RAGE) by attenuating β-catenin signaling. The reactivation of β-catenin signaling can revitalize neurogenesis, which holds promise for therapy of inflammatory demyelination.
Collapse
Affiliation(s)
- Yanna Song
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University; 600 Tianhe Road, Guangzhou 510630, Guangdong Province, China
| | - Wei Jiang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University; 600 Tianhe Road, Guangzhou 510630, Guangdong Province, China
| | - Shabbir Khan Afridi
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Tongtong Wang
- Department of Clinical Immunology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Fan Zhu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University; 600 Tianhe Road, Guangzhou 510630, Guangdong Province, China
| | - Huiming Xu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University; 600 Tianhe Road, Guangzhou 510630, Guangdong Province, China
| | - Faisal Hayat Nazir
- Nash Family Department of Neuroscience, Ronald M. Loeb Center for Alzheimer's Disease, and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chunxin Liu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University; 600 Tianhe Road, Guangzhou 510630, Guangdong Province, China
| | - Yuge Wang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University; 600 Tianhe Road, Guangzhou 510630, Guangdong Province, China
| | - Youming Long
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, 250 Changgang East Road, Guangzhou 510260, Guangdong Province, China; Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Collaborative Innovation Center for Neurogenetics and Channelopathies, 250 Changgang East Road, Guangzhou 510260, Guangdong Province, China
| | - Yu-Wen Alvin Huang
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, 70 Ship Street, Providence, RI 02903, USA
| | - Wei Qiu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University; 600 Tianhe Road, Guangzhou 510630, Guangdong Province, China.
| | - Changyong Tang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University; 600 Tianhe Road, Guangzhou 510630, Guangdong Province, China; Guangdong Provincial Key Laboratory of Diabetology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, China.
| |
Collapse
|
36
|
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
|
37
|
Lou S, Gong D, Yang M, Qiu Q, Luo J, Chen T. Curcumin Improves Neurogenesis in Alzheimer's Disease Mice via the Upregulation of Wnt/β-Catenin and BDNF. Int J Mol Sci 2024; 25:5123. [PMID: 38791161 PMCID: PMC11120842 DOI: 10.3390/ijms25105123] [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: 04/03/2024] [Revised: 04/30/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024] Open
Abstract
Adult neurogenesis in the dentate gyrus (DG) is impaired during Alzheimer's disease (AD) progression. Curcumin has been reported to reduce cell apoptosis and stimulate neurogenesis. This study aimed to investigate the influence of curcumin on adult neurogenesis in AD mice and its potential mechanism. Two-month-old male C57BL/6J mice were injected with soluble β-amyloid (Aβ1-42) using lateral ventricle stereolocalization to establish AD models. An immunofluorescence assay, including bromodeoxyuridine (BrdU), doublecortin (DCX), and neuron-specific nuclear antigen (NeuN), was used to detect hippocampal neurogenesis. Western blot and an enzyme-linked immunosorbent assay (ELISA) were used to test the expression of related proteins and the secretion of brain-derived neurotrophic factor (BDNF). A Morris water maze was used to detect the cognitive function of the mice. Our results showed that curcumin administration (100 mg/kg) rescued the impaired neurogenesis of Aβ1-42 mice, shown as enhanced BrdU+/DCX+ and BrdU+/NeuN+ cells in DG. In addition, curcumin regulated the phosphatidylinositol 3 kinase (PI3K)/protein kinase B (Akt) -mediated glycogen synthase kinase-3β (GSK3β) /Wingless/Integrated (Wnt)/β-catenin pathway and cyclic adenosine monophosphate response element-binding protein (CREB)/BDNF in Aβ1-42 mice. Inhibiting Wnt/β-catenin and depriving BDNF could reverse both the upregulated neurogenesis and cognitive function of curcumin-treated Aβ1-42 mice. In conclusion, our study indicates that curcumin, through targeting PI3K/Akt, regulates GSK3β/Wnt/β-catenin and CREB/BDNF pathways, improving the adult neurogenesis of AD mice.
Collapse
Affiliation(s)
| | | | | | | | - Jialie Luo
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, China; (S.L.); (D.G.); (M.Y.); (Q.Q.)
| | - Tingting Chen
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, China; (S.L.); (D.G.); (M.Y.); (Q.Q.)
| |
Collapse
|
38
|
Liu J, Mosti F, Zhao HT, Sotelo-Fonseca JE, Escobar-Tomlienovich CF, Lollis D, Musso CM, Mao Y, Massri AJ, Doll HM, Sousa AM, Wray GA, Schmidt E, Silver DL. A human-specific enhancer fine-tunes radial glia potency and corticogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.10.588953. [PMID: 38645099 PMCID: PMC11030412 DOI: 10.1101/2024.04.10.588953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Humans evolved an extraordinarily expanded and complex cerebral cortex, associated with developmental and gene regulatory modifications 1-3 . Human accelerated regions (HARs) are highly conserved genomic sequences with human-specific nucleotide substitutions. Although there are thousands of annotated HARs, their functional contribution to human-specific cortical development is largely unknown 4,5 . HARE5 is a HAR transcriptional enhancer of the WNT signaling receptor Frizzled8 (FZD8) active during brain development 6 . Here, using genome-edited mouse and primate models, we demonstrate that human (Hs) HARE5 fine-tunes cortical development and connectivity by controlling the proliferative and neurogenic capacity of neural progenitor cells (NPCs). Hs-HARE5 knock-in mice have significantly enlarged neocortices containing more neurons. By measuring neural dynamics in vivo we show these anatomical features correlate with increased functional independence between cortical regions. To understand the underlying developmental mechanisms, we assess progenitor fate using live imaging, lineage analysis, and single-cell RNA sequencing. This reveals Hs-HARE5 modifies radial glial progenitor behavior, with increased self-renewal at early developmental stages followed by expanded neurogenic potential. We use genome-edited human and chimpanzee (Pt) NPCs and cortical organoids to assess the relative enhancer activity and function of Hs-HARE5 and Pt-HARE5. Using these orthogonal strategies we show four human-specific variants in HARE5 drive increased enhancer activity which promotes progenitor proliferation. These findings illustrate how small changes in regulatory DNA can directly impact critical signaling pathways and brain development. Our study uncovers new functions for HARs as key regulatory elements crucial for the expansion and complexity of the human cerebral cortex.
Collapse
|
39
|
Borzou A, Miller SN, Hommel JD, Schwarz JM. Cocaine diminishes functional network robustness and destabilizes the energy landscape of neuronal activity in the medial prefrontal cortex. PNAS NEXUS 2024; 3:pgae092. [PMID: 38476665 PMCID: PMC10929585 DOI: 10.1093/pnasnexus/pgae092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 02/13/2024] [Indexed: 03/14/2024]
Abstract
We present analysis of neuronal activity recordings from a subset of neurons in the medial prefrontal cortex of rats before and after the administration of cocaine. Using an underlying modern Hopfield model as a description for the neuronal network, combined with a machine learning approach, we compute the underlying functional connectivity of the neuronal network. We find that the functional connectivity changes after the administration of cocaine with both functional-excitatory and functional-inhibitory neurons being affected. Using conventional network analysis, we find that the diameter of the graph, or the shortest length between the two most distant nodes, increases with cocaine, suggesting that the neuronal network is less robust. We also find that the betweenness centrality scores for several of the functional-excitatory and functional-inhibitory neurons decrease significantly, while other scores remain essentially unchanged, to also suggest that the neuronal network is less robust. Finally, we study the distribution of neuronal activity and relate it to energy to find that cocaine drives the neuronal network towards destabilization in the energy landscape of neuronal activation. While this destabilization is presumably temporary given one administration of cocaine, perhaps this initial destabilization indicates a transition towards a new stable state with repeated cocaine administration. However, such analyses are useful more generally to understand how neuronal networks respond to perturbations.
Collapse
Affiliation(s)
- Ahmad Borzou
- Department of Physics and BioInspired Institute, Syracuse University, Syracuse, NY 13244, USA
- CompuFlair, Houston, TX 77064, USA
| | - Sierra N Miller
- Department of Pharmacology and Toxicology, Center for Addiction Sciences and Therapeutics, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Jonathan D Hommel
- Department of Pharmacology and Toxicology, Center for Addiction Sciences and Therapeutics, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - J M Schwarz
- Department of Physics and BioInspired Institute, Syracuse University, Syracuse, NY 13244, USA
- Indian Creek Farm, Ithaca, NY 14850, USA
| |
Collapse
|
40
|
Barresi M, Hickmott RA, Bosakhar A, Quezada S, Quigley A, Kawasaki H, Walker D, Tolcos M. Toward a better understanding of how a gyrified brain develops. Cereb Cortex 2024; 34:bhae055. [PMID: 38425213 DOI: 10.1093/cercor/bhae055] [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: 04/24/2023] [Revised: 01/26/2024] [Accepted: 01/28/2024] [Indexed: 03/02/2024] Open
Abstract
The size and shape of the cerebral cortex have changed dramatically across evolution. For some species, the cortex remains smooth (lissencephalic) throughout their lifetime, while for other species, including humans and other primates, the cortex increases substantially in size and becomes folded (gyrencephalic). A folded cortex boasts substantially increased surface area, cortical thickness, and neuronal density, and it is therefore associated with higher-order cognitive abilities. The mechanisms that drive gyrification in some species, while others remain lissencephalic despite many shared neurodevelopmental features, have been a topic of investigation for many decades, giving rise to multiple perspectives of how the gyrified cerebral cortex acquires its unique shape. Recently, a structurally unique germinal layer, known as the outer subventricular zone, and the specialized cell type that populates it, called basal radial glial cells, were identified, and these have been shown to be indispensable for cortical expansion and folding. Transcriptional analyses and gene manipulation models have provided an invaluable insight into many of the key cellular and genetic drivers of gyrification. However, the degree to which certain biomechanical, genetic, and cellular processes drive gyrification remains under investigation. This review considers the key aspects of cerebral expansion and folding that have been identified to date and how theories of gyrification have evolved to incorporate this new knowledge.
Collapse
Affiliation(s)
- Mikaela Barresi
- School of Health and Biomedical Sciences, RMIT University, Plenty Road, Bundoora, VIC 3083, Australia
- ACMD, St Vincent's Hospital Melbourne, Regent Street, Fitzroy, VIC 3065, Australia
| | - Ryan Alexander Hickmott
- School of Health and Biomedical Sciences, RMIT University, Plenty Road, Bundoora, VIC 3083, Australia
- ACMD, St Vincent's Hospital Melbourne, Regent Street, Fitzroy, VIC 3065, Australia
| | - Abdulhameed Bosakhar
- School of Health and Biomedical Sciences, RMIT University, Plenty Road, Bundoora, VIC 3083, Australia
| | - Sebastian Quezada
- School of Health and Biomedical Sciences, RMIT University, Plenty Road, Bundoora, VIC 3083, Australia
| | - Anita Quigley
- School of Health and Biomedical Sciences, RMIT University, Plenty Road, Bundoora, VIC 3083, Australia
- ACMD, St Vincent's Hospital Melbourne, Regent Street, Fitzroy, VIC 3065, Australia
- School of Engineering, RMIT University, La Trobe Street, Melbourne, VIC 3000, Australia
- Department of Medicine, University of Melbourne, St Vincent's Hospital, Regent Street, Fitzroy, VIC 3065, Australia
| | - Hiroshi Kawasaki
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Takara-machi 13-1, Kanazawa, Ishikawa 920-8640, Japan
| | - David Walker
- School of Health and Biomedical Sciences, RMIT University, Plenty Road, Bundoora, VIC 3083, Australia
| | - Mary Tolcos
- School of Health and Biomedical Sciences, RMIT University, Plenty Road, Bundoora, VIC 3083, Australia
| |
Collapse
|
41
|
Huang LC, McKeown CR, He HY, Ta AC, Cline HT. BRCA1 and ELK-1 regulate neural progenitor cell fate in the optic tectum in response to visual experience in Xenopus laevis tadpoles. Proc Natl Acad Sci U S A 2024; 121:e2316542121. [PMID: 38198524 PMCID: PMC10801852 DOI: 10.1073/pnas.2316542121] [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: 09/27/2023] [Accepted: 12/05/2023] [Indexed: 01/12/2024] Open
Abstract
In developing Xenopus tadpoles, the optic tectum begins to receive patterned visual input while visuomotor circuits are still undergoing neurogenesis and circuit assembly. This visual input regulates neural progenitor cell fate decisions such that maintaining tadpoles in the dark increases proliferation, expanding the progenitor pool, while visual stimulation promotes neuronal differentiation. To identify regulators of activity-dependent neural progenitor cell fate, we profiled the transcriptomes of proliferating neural progenitor cells and newly differentiated neurons using RNA-Seq. We used advanced bioinformatic analysis of 1,130 differentially expressed transcripts to identify six differentially regulated transcriptional regulators, including Breast Cancer 1 (BRCA1) and the ETS-family transcription factor, ELK-1, which are predicted to regulate the majority of the other differentially expressed transcripts. BRCA1 is known for its role in cancers, but relatively little is known about its potential role in regulating neural progenitor cell fate. ELK-1 is a multifunctional transcription factor which regulates immediate early gene expression. We investigated the potential functions of BRCA1 and ELK-1 in activity-regulated neurogenesis in the tadpole visual system using in vivo time-lapse imaging to monitor the fate of GFP-expressing SOX2+ neural progenitor cells in the optic tectum. Our longitudinal in vivo imaging analysis showed that knockdown of either BRCA1 or ELK-1 altered the fates of neural progenitor cells and furthermore that the effects of visual experience on neurogenesis depend on BRCA1 and ELK-1 expression. These studies provide insight into the potential mechanisms by which neural activity affects neural progenitor cell fate.
Collapse
Affiliation(s)
- Lin-Chien Huang
- Department of Neuroscience, Dorris Neuroscience Center, Scripps Research Institute, La Jolla, CA92037
| | - Caroline R. McKeown
- Department of Neuroscience, Dorris Neuroscience Center, Scripps Research Institute, La Jolla, CA92037
| | - Hai-Yan He
- Department of Neuroscience, Dorris Neuroscience Center, Scripps Research Institute, La Jolla, CA92037
| | - Aaron C. Ta
- Department of Neuroscience, Dorris Neuroscience Center, Scripps Research Institute, La Jolla, CA92037
| | - Hollis T. Cline
- Department of Neuroscience, Dorris Neuroscience Center, Scripps Research Institute, La Jolla, CA92037
| |
Collapse
|
42
|
Abaci Turk E, Yun HJ, Feldman HA, Lee JY, Lee HJ, Bibbo C, Zhou C, Tamen R, Grant PE, Im K. Association between placental oxygen transport and fetal brain cortical development: a study in monochorionic diamniotic twins. Cereb Cortex 2024; 34:bhad383. [PMID: 37885155 PMCID: PMC11032198 DOI: 10.1093/cercor/bhad383] [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: 06/28/2023] [Revised: 09/25/2023] [Accepted: 09/26/2023] [Indexed: 10/28/2023] Open
Abstract
Normal cortical growth and the resulting folding patterns are crucial for normal brain function. Although cortical development is largely influenced by genetic factors, environmental factors in fetal life can modify the gene expression associated with brain development. As the placenta plays a vital role in shaping the fetal environment, affecting fetal growth through the exchange of oxygen and nutrients, placental oxygen transport might be one of the environmental factors that also affect early human cortical growth. In this study, we aimed to assess the placental oxygen transport during maternal hyperoxia and its impact on fetal brain development using MRI in identical twins to control for genetic and maternal factors. We enrolled 9 pregnant subjects with monochorionic diamniotic twins (30.03 ± 2.39 gestational weeks [mean ± SD]). We observed that the fetuses with slower placental oxygen delivery had reduced volumetric and surface growth of the cerebral cortex. Moreover, when the difference between placenta oxygen delivery increased between the twin pairs, sulcal folding patterns were more divergent. Thus, there is a significant relationship between placental oxygen transport and fetal brain cortical growth and folding in monochorionic twins.
Collapse
Affiliation(s)
- Esra Abaci Turk
- Department of Pediatrics, Harvard Medical School, 300 Longwood Ave, Boston, MA 02115, United States
- Division of Newborn Medicine, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, United States
- Fetal Neonatal Neuroimaging and Developmental Science Center, Boston Children’s Hospital, 401 Park Dr, Boston, MA 02115, United States
| | - Hyuk Jin Yun
- Department of Pediatrics, Harvard Medical School, 300 Longwood Ave, Boston, MA 02115, United States
- Division of Newborn Medicine, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, United States
- Fetal Neonatal Neuroimaging and Developmental Science Center, Boston Children’s Hospital, 401 Park Dr, Boston, MA 02115, United States
| | - Henry A Feldman
- Department of Pediatrics, Harvard Medical School, 300 Longwood Ave, Boston, MA 02115, United States
- Division of Newborn Medicine, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, United States
- Institutional Centers for Clinical and Translational Research, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, United States
| | - Joo Young Lee
- Department of Pediatrics, Hanyang University College of Medicine, 222, Wangsimni-ro, Seongdong-gu, Seoul, 04763, South Korea
| | - Hyun Ju Lee
- Department of Pediatrics, Hanyang University College of Medicine, 222, Wangsimni-ro, Seongdong-gu, Seoul, 04763, South Korea
| | - Carolina Bibbo
- Department of Obstetrics and Gynecology, Brigham and Women's Hospital, 75 Francis St, Boston, MA 02115, United States
| | - Cindy Zhou
- Division of Newborn Medicine, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, United States
- Fetal Neonatal Neuroimaging and Developmental Science Center, Boston Children’s Hospital, 401 Park Dr, Boston, MA 02115, United States
| | - Rubii Tamen
- Division of Newborn Medicine, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, United States
- Fetal Neonatal Neuroimaging and Developmental Science Center, Boston Children’s Hospital, 401 Park Dr, Boston, MA 02115, United States
| | - Patricia Ellen Grant
- Department of Pediatrics, Harvard Medical School, 300 Longwood Ave, Boston, MA 02115, United States
- Division of Newborn Medicine, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, United States
- Fetal Neonatal Neuroimaging and Developmental Science Center, Boston Children’s Hospital, 401 Park Dr, Boston, MA 02115, United States
- Department of Radiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, United States
| | - Kiho Im
- Department of Pediatrics, Harvard Medical School, 300 Longwood Ave, Boston, MA 02115, United States
- Division of Newborn Medicine, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, United States
- Fetal Neonatal Neuroimaging and Developmental Science Center, Boston Children’s Hospital, 401 Park Dr, Boston, MA 02115, United States
| |
Collapse
|
43
|
Akula SK, Exposito-Alonso D, Walsh CA. Shaping the brain: The emergence of cortical structure and folding. Dev Cell 2023; 58:2836-2849. [PMID: 38113850 PMCID: PMC10793202 DOI: 10.1016/j.devcel.2023.11.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 04/08/2023] [Accepted: 11/10/2023] [Indexed: 12/21/2023]
Abstract
The cerebral cortex-the brain's covering and largest region-has increased in size and complexity in humans and supports higher cognitive functions such as language and abstract thinking. There is a growing understanding of the human cerebral cortex, including the diversity and number of cell types that it contains, as well as of the developmental mechanisms that shape cortical structure and organization. In this review, we discuss recent progress in our understanding of molecular and cellular processes, as well as mechanical forces, that regulate the folding of the cerebral cortex. Advances in human genetics, coupled with experimental modeling in gyrencephalic species, have provided insights into the central role of cortical progenitors in the gyrification and evolutionary expansion of the cerebral cortex. These studies are essential for understanding the emergence of structural and functional organization during cortical development and the pathogenesis of neurodevelopmental disorders associated with cortical malformations.
Collapse
Affiliation(s)
- Shyam K Akula
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA; Departments of Pediatrics and Neurology, Harvard Medical School, Boston, MA, USA; Allen Discovery Center for Human Brain Evolution, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA; Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| | - David Exposito-Alonso
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA; Departments of Pediatrics and Neurology, Harvard Medical School, Boston, MA, USA; Allen Discovery Center for Human Brain Evolution, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA; Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| | - Christopher A Walsh
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA; Departments of Pediatrics and Neurology, Harvard Medical School, Boston, MA, USA; Allen Discovery Center for Human Brain Evolution, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA; Howard Hughes Medical Institute, Chevy Chase, Maryland, USA.
| |
Collapse
|
44
|
Ye J, Yan X, Zhang W, Lu J, Xu S, Li X, Qin P, Gong X, Liu Y, Ling Y, Li Y, Zhang Y, Fang F. Integrative proteomic and phosphoproteomic analysis in the female goat hypothalamus to study the onset of puberty. BMC Genomics 2023; 24:621. [PMID: 37853328 PMCID: PMC10583467 DOI: 10.1186/s12864-023-09705-7] [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: 03/14/2023] [Accepted: 09/28/2023] [Indexed: 10/20/2023] Open
Abstract
BACKGROUND Puberty marks the end of childhood and achieve sexual maturation and fertility. The role of hypothalamic proteins in regulating puberty onset is unclear. We performed a comprehensive differential proteomics and phosphoproteomics analysis in prepubertal and pubertal goats to determine the roles of hypothalamic proteins and phosphoproteins during the onset of puberty. RESULTS We used peptide and posttranslational modifications peptide quantification and statistical analyses, and identified 69 differentially expressed proteins from 5,057 proteins and 576 differentially expressed phosphopeptides from 1574 phosphorylated proteins. Combined proteomic and phosphoproteomics, 759 correlated proteins were identified, of which 5 were differentially expressed only at the protein level, and 201 were only differentially expressed at the phosphoprotein level. Pathway enrichment analyses revealed that the majority of correlated proteins were associated with glycolysis/gluconeogenesis, Fc gamma R-mediated phagocytosis, focal adhesion, GABAergic synapse, and Rap1 signaling pathway. These pathways are related to cell proliferation, neurocyte migration, and promoting the release of gonadotropin-releasing hormone in the hypothalamus. CTNNB1 occupied important locations in the protein-protein interaction network and is involved in focal adhesion. CONCLUSION The results demonstrate that the proteins differentially expression only at the protein level or only differentially expressed at the phosphoprotein level and their related signalling pathways are crucial in regulating puberty in goats. These differentially expressed proteins and phosphorylated proteins may constitute the proteomic backgrounds between the two different stages.
Collapse
Affiliation(s)
- Jing Ye
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, 230036, Hefei, Anhui, China
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, 230036, Hefei, Anhui, China
| | - Xu Yan
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, 230036, Hefei, Anhui, China
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, 230036, Hefei, Anhui, China
| | - Wei Zhang
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, 230036, Hefei, Anhui, China
| | - Juntai Lu
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, 230036, Hefei, Anhui, China
| | - Shuangshuang Xu
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, 230036, Hefei, Anhui, China
| | - Xiaoqian Li
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, 230036, Hefei, Anhui, China
| | - Ping Qin
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, 230036, Hefei, Anhui, China
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, 230036, Hefei, Anhui, China
| | - Xinbao Gong
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, 230036, Hefei, Anhui, China
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, 230036, Hefei, Anhui, China
| | - Ya Liu
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, 230036, Hefei, Anhui, China
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, 230036, Hefei, Anhui, China
| | - Yinghui Ling
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, 230036, Hefei, Anhui, China
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, 230036, Hefei, Anhui, China
| | - Yunsheng Li
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, 230036, Hefei, Anhui, China
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, 230036, Hefei, Anhui, China
| | - Yunhai Zhang
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, 230036, Hefei, Anhui, China
| | - Fugui Fang
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, 230036, Hefei, Anhui, China.
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, 230036, Hefei, Anhui, China.
| |
Collapse
|
45
|
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
|
46
|
Gaston-Breton R, Maïza Letrou A, Hamoudi R, Stonestreet BS, Mabondzo A. Brain organoids for hypoxic-ischemic studies: from bench to bedside. Cell Mol Life Sci 2023; 80:318. [PMID: 37804439 PMCID: PMC10560197 DOI: 10.1007/s00018-023-04951-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/31/2023] [Accepted: 09/06/2023] [Indexed: 10/09/2023]
Abstract
Our current knowledge regarding the development of the human brain mostly derives from experimental studies on non-human primates, sheep, and rodents. However, these studies may not completely simulate all the features of human brain development as a result of species differences and variations in pre- and postnatal brain maturation. Therefore, it is important to supplement the in vivo animal models to increase the possibility that preclinical studies have appropriate relevance for potential future human trials. Three-dimensional brain organoid culture technology could complement in vivo animal studies to enhance the translatability of the preclinical animal studies and the understanding of brain-related disorders. In this review, we focus on the development of a model of hypoxic-ischemic (HI) brain injury using human brain organoids to complement the translation from animal experiments to human pathophysiology. We also discuss how the development of these tools provides potential opportunities to study fundamental aspects of the pathophysiology of HI-related brain injury including differences in the responses between males and females.
Collapse
Affiliation(s)
- Romane Gaston-Breton
- Université Paris Saclay, CEA, INRAE, Médicaments et Technologies pour la Santé (DMTS), Laboratoire d'Etude de l'Unité Neurovasculaire & Innovation Thérapeutique, 91191, Gif-sur-Yvette Cedex, France
| | - Auriane Maïza Letrou
- Université Paris Saclay, CEA, INRAE, Médicaments et Technologies pour la Santé (DMTS), Laboratoire d'Etude de l'Unité Neurovasculaire & Innovation Thérapeutique, 91191, Gif-sur-Yvette Cedex, France
| | - Rifat Hamoudi
- Research Institute for Medical and Health Sciences, University of Sharjah, P. O. 27272, Sharjah, United Arab Emirates
- College of Medicine, University of Sharjah, P. O. 27272, Sharjah, United Arab Emirates
- Division of Surgery and Interventional Science, University College London, London, UK
- ASPIRE Precision Medicine Research Institute Abu Dhabi, University of Sharjah, Sharjah, United Arab Emirates
| | - Barbara S Stonestreet
- Departments of Molecular Biology, Cell Biology and Biochemistry and Department of Pediatrics, Women & Infants Hospital of Rhode Island, The Alpert Medical School of Brown University, 101 Dudley Street, Providence, RI, 02905, USA
| | - Aloïse Mabondzo
- Université Paris Saclay, CEA, INRAE, Médicaments et Technologies pour la Santé (DMTS), Laboratoire d'Etude de l'Unité Neurovasculaire & Innovation Thérapeutique, 91191, Gif-sur-Yvette Cedex, France.
| |
Collapse
|
47
|
Warrier V, Stauffer EM, Huang QQ, Wigdor EM, Slob EAW, Seidlitz J, Ronan L, Valk SL, Mallard TT, Grotzinger AD, Romero-Garcia R, Baron-Cohen S, Geschwind DH, Lancaster MA, Murray GK, Gandal MJ, Alexander-Bloch A, Won H, Martin HC, Bullmore ET, Bethlehem RAI. Genetic insights into human cortical organization and development through genome-wide analyses of 2,347 neuroimaging phenotypes. Nat Genet 2023; 55:1483-1493. [PMID: 37592024 PMCID: PMC10600728 DOI: 10.1038/s41588-023-01475-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 07/13/2023] [Indexed: 08/19/2023]
Abstract
Our understanding of the genetics of the human cerebral cortex is limited both in terms of the diversity and the anatomical granularity of brain structural phenotypes. Here we conducted a genome-wide association meta-analysis of 13 structural and diffusion magnetic resonance imaging-derived cortical phenotypes, measured globally and at 180 bilaterally averaged regions in 36,663 individuals and identified 4,349 experiment-wide significant loci. These phenotypes include cortical thickness, surface area, gray matter volume, measures of folding, neurite density and water diffusion. We identified four genetic latent structures and causal relationships between surface area and some measures of cortical folding. These latent structures partly relate to different underlying gene expression trajectories during development and are enriched for different cell types. We also identified differential enrichment for neurodevelopmental and constrained genes and demonstrate that common genetic variants associated with cortical expansion are associated with cephalic disorders. Finally, we identified complex interphenotype and inter-regional genetic relationships among the 13 phenotypes, reflecting the developmental differences among them. Together, these analyses identify distinct genetic organizational principles of the cortex and their correlates with neurodevelopment.
Collapse
Affiliation(s)
- Varun Warrier
- Department of Psychiatry, University of Cambridge, Cambridge, UK.
- Department of Psychology, University of Cambridge, Cambridge, UK.
| | | | | | | | - Eric A W Slob
- Medical Research Council Biostatistics Unit, University of Cambridge, Cambridge, UK
- Department of Applied Economics, Erasmus School of Economics, Erasmus University Rotterdam, Rotterdam, the Netherlands
- Erasmus University Rotterdam Institute for Behavior and Biology, Erasmus University Rotterdam, Rotterdam, the Netherlands
| | - Jakob Seidlitz
- Department of Child and Adolescent Psychiatry and Behavioral Science, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Lifespan Brain Institute, The Children's Hospital of Philadelphia and Penn Medicine, Philadelphia, PA, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
| | - Lisa Ronan
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Sofie L Valk
- Institute of Neuroscience and Medicine, Brain & Behaviour (INM-7), Research Centre Jülich, FZ Jülich, Jülich, Germany
- Institute of Systems Neuroscience, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Otto Hahn Group Cognitive Neurogenetics, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Travis T Mallard
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Andrew D Grotzinger
- Department of Psychology and Neuroscience, University of Colorado at Boulder, Boulder, CO, USA
- Institute for Behavioral Genetics, University of Colorado at Boulder, Boulder, CO, USA
| | - Rafael Romero-Garcia
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- Instituto de Biomedicina de Sevilla (IBiS) HUVR/CSIC/Universidad de Sevilla/CIBERSAM, ISCIII, Dpto. de Fisiología Médica y Biofísica, Seville, Spain
| | - Simon Baron-Cohen
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- Department of Psychology, University of Cambridge, Cambridge, UK
| | - Daniel H Geschwind
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Program in Neurogenetics, Department of Neurology, University of California, Los Angeles, CA, USA
- Center for Autism Research and Treatment, Jane and TerrySemel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
- Institute of Precision Health, University of California, Los Angeles, CA, USA
| | - Madeline A Lancaster
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge, UK
| | - Graham K Murray
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- Cambridgeshire and Peterborough NHS Trust, Cambridge, UK
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - Michael J Gandal
- Department of Child and Adolescent Psychiatry and Behavioral Science, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Lifespan Brain Institute, The Children's Hospital of Philadelphia and Penn Medicine, Philadelphia, PA, USA
| | - Aaron Alexander-Bloch
- Department of Child and Adolescent Psychiatry and Behavioral Science, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Lifespan Brain Institute, The Children's Hospital of Philadelphia and Penn Medicine, Philadelphia, PA, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
| | - Hyejung Won
- Department of Genetics and the Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Edward T Bullmore
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- Cambridgeshire and Peterborough NHS Trust, Cambridge, UK
| | - Richard A I Bethlehem
- Department of Psychiatry, University of Cambridge, Cambridge, UK.
- Department of Psychology, University of Cambridge, Cambridge, UK.
| |
Collapse
|
48
|
Ewell A, Allard T, Botdorf M, Ji A, Riggins T. Emotion regulation and reactivity are associated with cortical thickness in early to mid-childhood. Dev Psychobiol 2023; 65:e22412. [PMID: 37607888 DOI: 10.1002/dev.22412] [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: 09/09/2022] [Revised: 04/06/2023] [Accepted: 07/05/2023] [Indexed: 08/24/2023]
Abstract
This study explored the neural correlates of emotion regulation and emotional reactivity in early to mid-childhood. A sample of 96 children (70% White, mid-to-high socioeconomic status) aged 3-8 years provided structural neuroimaging data and caregivers reported on emotion regulation and emotional reactivity. The amygdala, insula, inferior frontal gyrus, anterior cingulate cortex, and medial orbitofrontal cortex were explored as a priori regions of interest (ROIs). ROI analyses revealed that emotion regulation was positively associated with cortical thickness in the insula, whereas emotional reactivity was negatively associated with cortical thickness in the inferior frontal gyrus. Exploratory whole-brain analyses suggested positive associations between emotion regulation and both left superior temporal thickness and right inferior temporal thickness, as well as negative associations between emotional reactivity and left superior temporal thickness. There were no significant associations between emotional regulation or reactivity and amygdala volume or cortical surface area. These findings support the notion that surface area and cortical thickness are distinct measures of brain maturation. In sum, these findings suggest that children may rely on a wider set of neural regions for emotion regulation and reactivity than adults, which is consistent with theories of interactive specialization across the life span.
Collapse
Affiliation(s)
- Arcadia Ewell
- Department of Psychological and Brain Sciences, Boston University, Boston, Massachusetts, USA
| | - Tamara Allard
- Department of Psychology, University of Maryland, College Park, Maryland, USA
| | - Morgan Botdorf
- Department of Psychology, University of Maryland, College Park, Maryland, USA
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Angela Ji
- Department of Graduate Psychology, James Madison University, Harrisonburg, Virginia, USA
| | - Tracy Riggins
- Department of Psychology, University of Maryland, College Park, Maryland, USA
| |
Collapse
|
49
|
Watanabe K, Horie M, Hayatsu M, Mikami Y, Sato N. Spatiotemporal expression patterns of R-spondins and their receptors, Lgrs, in the developing mouse telencephalon. Gene Expr Patterns 2023; 49:119333. [PMID: 37651925 DOI: 10.1016/j.gep.2023.119333] [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: 03/27/2023] [Revised: 05/14/2023] [Accepted: 08/24/2023] [Indexed: 09/02/2023]
Abstract
Development of the mammalian telencephalon, which is the most complex region of the central nervous system, is precisely orchestrated by many signaling molecules. Wnt signaling derived from the cortical hem, a signaling center, is crucial for telencephalic development including cortical patterning and the induction of hippocampal development. Secreted protein R-spondin (Rspo) 1-4 and their receptors, leucine-rich repeat-containing G-protein-coupled receptor (Lgr) 4-6, act as activators of Wnt signaling. Although Rspo expression in the hem during the early stages of cortical development has been reported, comparative expression analysis of Rspos and Lgr4-6 has not been performed. In this study, we examined the detailed spatiotemporal expression patterns of Rspo1-4 and Lgr4-6 in the embryonic and postnatal telencephalon to elucidate their functions. In the embryonic day (E) 10.5-14.5 telencephalon, Rspo1-3 were prominently expressed in the cortical hem. Among their receptors, Lgr4 was observed in the ventral telencephalon, and Lgr6 was highly expressed throughout the telencephalon at the same stages. This suggests that Rspo1-3 and Lgr4 initially regulate telencephalic development in restricted regions, whereas Lgr6 functions broadly. From the late embryonic stage, the expression areas of Rspo1-3 and Lgr4-6 dramatically expanded; their expression was found in the neocortex and limbic system, such as the hippocampus, amygdala, and striatum. Increased Rspo and Lgr expression from the late embryonic stages suggests broad roles of Rspo signaling in telencephalic development. Furthermore, the Lgr+ regions were located far from the Rspo+ regions, especially in the E10.5-14.5 ventral telencephalon, suggesting that Lgrs act via a Rspo-independent pathway.
Collapse
Affiliation(s)
- Keisuke Watanabe
- Division of Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan.
| | - Masao Horie
- Department of Nursing, Niigata College of Nursing, Jōetsu, Japan
| | - Manabu Hayatsu
- Division of Microscopic Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Yoshikazu Mikami
- Division of Microscopic Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Noboru Sato
- Division of Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| |
Collapse
|
50
|
Ji F, Feng C, Qin J, Wang C, Zhang D, Su L, Wang W, Zhang M, Li H, Ma L, Lu W, Liu C, Teng Z, Hu B, Jian F, Xie J, Jiao J. Brain-specific Pd1 deficiency leads to cortical neurogenesis defects and depressive-like behaviors in mice. Cell Death Differ 2023; 30:2053-2065. [PMID: 37553426 PMCID: PMC10482844 DOI: 10.1038/s41418-023-01203-3] [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: 11/09/2022] [Revised: 07/18/2023] [Accepted: 08/02/2023] [Indexed: 08/10/2023] Open
Abstract
Embryonic neurogenesis is tightly regulated by multiple factors to ensure the precise development of the cortex. Deficiency in neurogenesis may result in behavioral abnormalities. Pd1 is a well-known inhibitory immune molecule, but its function in brain development remains unknown. Here, we find brain specific deletion of Pd1 results in abnormal cortical neurogenesis, including enhanced proliferation of neural progenitors and reduced neuronal differentiation. In addition, neurons in Pd1 knockout mice exhibit abnormal morphology, both the total length and the number of primary dendrites were reduced. Moreover, Pd1cKO mice exhibit depressive-like behaviors, including immobility, despair, and anhedonia. Mechanistically, Pd1 regulates embryonic neurogenesis by targeting Pax3 through the β-catenin signaling pathway. The constitutive expression of Pax3 partly rescues the deficiency of neurogenesis in the Pd1 deleted embryonic brain. Besides, the administration of β-catenin inhibitor, XAV939, not only rescues abnormal brain development but also ameliorates depressive-like behaviors in Pd1cKO mice. Simultaneously, Pd1 plays a similar role in human neural progenitor cells (hNPCs) proliferation and differentiation. Taken together, our findings reveal the critical role and regulatory mechanism of Pd1 in embryonic neurogenesis and behavioral modulation, which could contribute to understanding immune molecules in brain development.
Collapse
Affiliation(s)
- Fen Ji
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China.
- Innovation Academy for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China.
- University of Chinese Academy of Sciences, 100049, Beijing, China.
| | - Chao Feng
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
- Sino-Danish College at University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Jie Qin
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
- Sino-Danish College at University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Chong Wang
- State Key Laboratory of 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, Anhui, China
| | - Dongming Zhang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Libo Su
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Wenwen Wang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Mengtian Zhang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Hong Li
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- Innovation Academy for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
| | - Longbing Ma
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 100053, Beijing, China
| | - Weicheng Lu
- Department of Anesthesiology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, China
| | - Changmei Liu
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- Innovation Academy for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Zhaoqian Teng
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- Innovation Academy for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Baoyang Hu
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- Innovation Academy for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Fengzeng Jian
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 100053, Beijing, China.
| | - Jingdun Xie
- Department of Anesthesiology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, China.
| | - Jianwei Jiao
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China.
- Innovation Academy for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China.
- University of Chinese Academy of Sciences, 100049, Beijing, China.
| |
Collapse
|