1
|
Kurahashi H, Kunisawa K, Tanaka KF, Kubota H, Hasegawa M, Miyachi M, Moriya Y, Hasegawa Y, Nagai T, Saito K, Nabeshima T, Mouri A. Autism spectrum disorder-like behaviors induced by hyper-glutamatergic NMDA receptor signaling through hypo-serotonergic 5-HT 1A receptor signaling in the prefrontal cortex in mice exposed to prenatal valproic acid. Neuropsychopharmacology 2024:10.1038/s41386-024-02004-z. [PMID: 39394255 DOI: 10.1038/s41386-024-02004-z] [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: 04/10/2024] [Revised: 08/28/2024] [Accepted: 09/30/2024] [Indexed: 10/13/2024]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by repetitive behaviors, social deficits, and cognitive impairments. Maternal use of valproic acid (VPA) during pregnancy is associated with an increased risk of ASD in offspring. The prevailing pathophysiological hypothesis for ASD involves excitation/inhibition (E/I) imbalances and serotonergic dysfunction. Here, we investigated the association between glutamatergic-serotonergic neuronal interactions and ASD-like behaviors in mice exposed to prenatal VPA. Prenatal VPA exposure induced excessive repetitive self-grooming behavior and impaired social behavior and object recognition memory in young adult period. Prenatal VPA mice showed hyper-glutamatergic function (increase in basal extracellular glutamate levels and CaMKII phosphorylation) and hypo-serotonergic function (decrease in 5-hydroxyindoleacetic acid and stimulation-induced serotonin [5-HT] release, but an increase in 5-HT transporter expression) in the prefrontal cortex. Treatment with a low-affinity NMDA receptor antagonist (memantine), a selective 5-HT reuptake inhibitor (fluoxetine), and a 5-HT1A receptor agonist (tandospirone) attenuated both the increase in CaMKII phosphorylation and ASD-like behavior of prenatal VPA mice. Opto-genetic activation of the serotonergic neuronal system attenuated impairments in social behavior and object recognition memory in prenatal VPA mice. WAY-100635-a 5-HT1A receptor antagonist-antagonized the effect of fluoxetine on impaired social behavior and object recognition memory. These results suggest that E/I imbalance and ASD-like behavior are associated with hypo-serotonergic receptor signaling through 5-HT1A receptors in prenatal VPA mice.
Collapse
Affiliation(s)
- Hitomi Kurahashi
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Health Science, Aichi, Japan
| | - Kazuo Kunisawa
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Health Science, Aichi, Japan
- International Center for Brain Science (ICBS), Fujita Health University, Aichi, Japan
| | - Kenji F Tanaka
- Division of Brain Sciences, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Hisayoshi Kubota
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Health Science, Aichi, Japan
- International Center for Brain Science (ICBS), Fujita Health University, Aichi, Japan
| | - Masaya Hasegawa
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Health Science, Aichi, Japan
| | - Mai Miyachi
- Division of pharmaceutical science, Faculty of pharmacy, Meijo University, Aichi, Japan
| | - Yuka Moriya
- Division of pharmaceutical science, Faculty of pharmacy, Meijo University, Aichi, Japan
| | - Yoichi Hasegawa
- Division of pharmaceutical science, Faculty of pharmacy, Meijo University, Aichi, Japan
| | - Taku Nagai
- International Center for Brain Science (ICBS), Fujita Health University, Aichi, Japan
| | - Kuniaki Saito
- Department of Disease Control and Prevention, Fujita Health University Graduate School of Health Science, Aichi, Japan
- Laboratory of Health and Medical Science Innovation (HMSI), Fujita Health University Graduate School of Health Science, Aichi, Japan
- Japanese Drug Organization of Appropriate Use and Research, Aichi, Japan
| | - Toshitaka Nabeshima
- International Center for Brain Science (ICBS), Fujita Health University, Aichi, Japan
- Laboratory of Health and Medical Science Innovation (HMSI), Fujita Health University Graduate School of Health Science, Aichi, Japan
- Japanese Drug Organization of Appropriate Use and Research, Aichi, Japan
| | - Akihiro Mouri
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Health Science, Aichi, Japan.
- International Center for Brain Science (ICBS), Fujita Health University, Aichi, Japan.
- Japanese Drug Organization of Appropriate Use and Research, Aichi, Japan.
| |
Collapse
|
2
|
Gong M, Li J, Qin Z, Machado Bressan Wilke MV, Liu Y, Li Q, Liu H, Liang C, Morales-Rosado JA, Cohen ASA, Hughes SS, Sullivan BR, Waddell V, van den Boogaard MJH, van Jaarsveld RH, van Binsbergen E, van Gassen KL, Wang T, Hiatt SM, Amaral MD, Kelley WV, Zhao J, Feng W, Ren C, Yu Y, Boczek NJ, Ferber MJ, Lahner C, Elliott S, Ruan Y, Mignot C, Keren B, Xie H, Wang X, Popp B, Zweier C, Piard J, Coubes C, Mau-Them FT, Safraou H, Innes AM, Gauthier J, Michaud JL, Koboldt DC, Sylvie O, Willems M, Tan WH, Cogne B, Rieubland C, Braun D, McLean SD, Platzer K, Zacher P, Oppermann H, Evenepoel L, Blanc P, El Khattabi L, Haque N, Dsouza NR, Zimmermann MT, Urrutia R, Klee EW, Shen Y, Du H, Rappaport L, Liu CM, Chen X. MARK2 variants cause autism spectrum disorder via the downregulation of WNT/β-catenin signaling pathway. Am J Hum Genet 2024:S0002-9297(24)00366-5. [PMID: 39419027 DOI: 10.1016/j.ajhg.2024.09.006] [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: 03/25/2024] [Revised: 09/19/2024] [Accepted: 09/25/2024] [Indexed: 10/19/2024] Open
Abstract
Microtubule affinity-regulating kinase 2 (MARK2) contributes to establishing neuronal polarity and developing dendritic spines. Although large-scale sequencing studies have associated MARK2 variants with autism spectrum disorder (ASD), the clinical features and variant spectrum in affected individuals with MARK2 variants, early developmental phenotypes in mutant human neurons, and the pathogenic mechanism underlying effects on neuronal development have remained unclear. Here, we report 31 individuals with MARK2 variants and presenting with ASD, other neurodevelopmental disorders, and distinctive facial features. Loss-of-function (LoF) variants predominate (81%) in affected individuals, while computational analysis and in vitro expression assay of missense variants supported the effect of MARK2 loss. Using proband-derived and CRISPR-engineered isogenic induced pluripotent stem cells (iPSCs), we show that MARK2 loss leads to early neuronal developmental and functional deficits, including anomalous polarity and dis-organization in neural rosettes, as well as imbalanced proliferation and differentiation in neural progenitor cells (NPCs). Mark2+/- mice showed abnormal cortical formation and partition and ASD-like behavior. Through the use of RNA sequencing (RNA-seq) and lithium treatment, we link MARK2 loss to downregulation of the WNT/β-catenin signaling pathway and identify lithium as a potential drug for treating MARK2-associated ASD.
Collapse
Affiliation(s)
- Maolei Gong
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China; The Ninth Medical Center of PLA General Hospital, Beijing, China
| | - Jiayi Li
- Department of Medical Genetics, Capital Institute of Pediatrics, Beijing, China; Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zailong Qin
- Genetic and Metabolic Central Laboratory, Birth Defect Prevention Research Institute, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | | | - Yijun Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China; Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Qian Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China; Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Haoran Liu
- Department of Medical Genetics, Capital Institute of Pediatrics, Beijing, China
| | - Chen Liang
- Department of Medical Genetics, Capital Institute of Pediatrics, Beijing, China
| | - Joel A Morales-Rosado
- Department of Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ana S A Cohen
- Department of Pathology and Laboratory Medicine, Genomic Medicine Center, Children's Mercy-Kansas City, Kansas City, MO, USA; The University of Missouri-Kansas City, School of Medicine, Kansas City, MO, USA
| | - Susan S Hughes
- The University of Missouri-Kansas City, School of Medicine, Kansas City, MO, USA; Division of Clinical Genetics, Children's Mercy Kansas City, Kansas City, MO, USA
| | - Bonnie R Sullivan
- The University of Missouri-Kansas City, School of Medicine, Kansas City, MO, USA; Division of Clinical Genetics, Children's Mercy Kansas City, Kansas City, MO, USA
| | - Valerie Waddell
- Department of Neurology, Children's Mercy Kansas City, Kansas City, MO, USA
| | | | - Richard H van Jaarsveld
- Department of Genetics, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands
| | - Ellen van Binsbergen
- Department of Genetics, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands
| | - Koen L van Gassen
- Department of Genetics, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands
| | - Tianyun Wang
- Department of Medical Genetics, Center for Medical Genetics, School of Basic Medical Sciences, Autism Research Center, Peking University Health Science Center, Beijing, China; Neuroscience Research Institute, Peking University, Key Laboratory for Neuroscience, Ministry of Education of China & National Health Commission of China, Beijing, China
| | - Susan M Hiatt
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
| | | | | | - Jianbo Zhao
- Department of Neurology Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Weixing Feng
- Department of Neurology Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Changhong Ren
- Department of Neurology Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Yazhen Yu
- Department of Pediatrics, Beijing Tiantan Hospital affiliated with Capital University of Medical Sciences, Beijing, China
| | - Nicole J Boczek
- Department of Laboratory Medicine and Pathology, Genomics Laboratory, Mayo Clinic, Rochester, MN, USA
| | - Matthew J Ferber
- Department of Laboratory Medicine and Pathology, Genomics Laboratory, Mayo Clinic, Rochester, MN, USA
| | - Carrie Lahner
- Department of Laboratory Medicine and Pathology, Genomics Laboratory, Mayo Clinic, Rochester, MN, USA
| | - Sherr Elliott
- Departments of Neurology and Pediatrics, Institute of Human Genetics and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Yiyan Ruan
- Guangxi Clinical Research Center for Pediatric Diseases, The Maternal and Child Health Care Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Cyril Mignot
- APHP Sorbonne Université, Département de Génétique, Hôpital Pitié-Salpêtrière et Hôpital Trousseau, Centre de Référence Déficiences Intellectuelles de Causes Rares, Paris, France
| | - Boris Keren
- APHP Sorbonne Université, Département de Génétique, Hôpital Pitié-Salpêtrière et Hôpital Trousseau, Centre de Référence Déficiences Intellectuelles de Causes Rares, Paris, France
| | - Hua Xie
- Department of Medical Genetics, Capital Institute of Pediatrics, Beijing, China
| | - Xiaoyan Wang
- Department of Children's Nutrition Research Center, Affiliated Children's Hospital of Capital Institute of Pediatrics, Beijing, China
| | - Bernt Popp
- Institute of Human Genetics, University of Leipzig Hospitals and Clinics, Leipzig, Germany; Berlin Institute of Health at Charité-Universitäts medizin Berlin, Center of Functional Genomics, Hessische Straße 4A, Berlin, Germany
| | - Christiane Zweier
- Department of Human Genetics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Institute of Human Genetics, University Hospital Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Juliette Piard
- Centre de Génétique Humaine, Centre Hospitalier Régional Universitaire, Université de Franche-Comté, Besançon, France; UMR 1231 GAD, Inserm, Université de Bourgogne Franche Comté, Dijon, France
| | - Christine Coubes
- Département de Génétique Médicale, Maladies Rares et Médecine Personnalisée Hôpital Arnaud de Villeneuve, 34295 Montpellier Cedex, Dijon, France
| | - Frederic Tran Mau-Them
- UF6254 Innovation en Diagnostic Genomique des Maladies Rares, Dijon, France; Inserm UMR1231 GAD, 21000 Dijon, France
| | - Hana Safraou
- UF6254 Innovation en Diagnostic Genomique des Maladies Rares, Dijon, France; Inserm UMR1231 GAD, 21000 Dijon, France
| | - A Micheil Innes
- Department of Medical Genetics and Pediatrics and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Julie Gauthier
- Molecular Diagnostic Laboratory, Centre Hospitalier Universitaire Sainte-Justine, Montréal, QC, Canada; Department of Pediatrics, Université de Montréal, Montréal, QC, Canada
| | - Jacques L Michaud
- Department of Pediatrics, Université de Montréal, Montréal, QC, Canada; CHU Sainte-Justine Research Center, Montreal, QC, Canada
| | - Daniel C Koboldt
- The Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, USA
| | - Odent Sylvie
- Service de Génétique clinique, CHU Rennes, ERN ITHACA, Rennes, France; University Rennes, CNRS, INSERM, IGDR (Institut de Génétique et développement de Rennes), UMR 6290, ERL U1305, Rennes, France
| | - Marjolaine Willems
- Medical Genetic Department for Rare Diseases and Personalized Medicine, Reference Center AD SOOR, AnDDI-RARE, Inserm U1298, INM, Montpellier University, Centre Hospitalier Universitaire de Montpellier, Montpellier, France
| | - Wen-Hann Tan
- Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Benjamin Cogne
- Nantes Université, CHU Nantes, Service de Génétique Médicale, Nantes, France; Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France
| | - Claudine Rieubland
- Department of Human Genetics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Dominique Braun
- Department of Human Genetics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Scott Douglas McLean
- Division of Clinical Genetics, The Children's Hospital of San Antonio, San Antonio, TX, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Konrad Platzer
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany
| | - Pia Zacher
- Epilepsy Center Kleinwachau, Dresden-Radeberg, Germany
| | - Henry Oppermann
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany
| | - Lucie Evenepoel
- Centre de Génétique Humaine, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Avenue Hippocrate 10-1200, Brussels, Belgium
| | - Pierre Blanc
- Sorbonne Université, Department of Medical Genetics, APHP, Pitié-Salpêtrière hospital, Paris Brain Institute-ICM, Laboratoire SeqOIA-PFMG2025, Paris, France
| | - Laïla El Khattabi
- Department of Medical Genetics, APHP, Armand Trousseau and Pitié-Salpêtrière hospitals, Brain Development team, Paris Brain Institute-ICM, Sorbonne Université, Paris, France; Laboratoire SeqOIA-PFMG2025, Paris, France
| | - Neshatul Haque
- Bioinformatics Research and Development Laboratory, Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Nikita R Dsouza
- Bioinformatics Research and Development Laboratory, Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Michael T Zimmermann
- Bioinformatics Research and Development Laboratory, Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA; Clinical and Translational Sciences Institute, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Raul Urrutia
- Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Eric W Klee
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN, USA; Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Yiping Shen
- Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; SynerGene Education, Hejun College, Huichang Jiangxi, China
| | - Hongzhen Du
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Leonard Rappaport
- Division of Developmental Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Chang-Mei Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China; Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China.
| | - Xiaoli Chen
- Department of Medical Genetics, Capital Institute of Pediatrics, Beijing, China; Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
3
|
De Vincenti AP, Bonafina A, Ledda F, Paratcha G. Lrig1 regulates cell fate specification of glutamatergic neurons via FGF-driven Jak2/Stat3 signaling in cortical progenitors. Development 2024; 151:dev202879. [PMID: 39250533 DOI: 10.1242/dev.202879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 08/07/2024] [Indexed: 09/11/2024]
Abstract
The cell-intrinsic mechanisms underlying the decision of a stem/progenitor cell to either proliferate or differentiate remain incompletely understood. Here, we identify the transmembrane protein Lrig1 as a physiological homeostatic regulator of FGF2-driven proliferation and self-renewal of neural progenitors at early-to-mid embryonic stages of cortical development. We show that Lrig1 is expressed in cortical progenitors (CPs), and its ablation caused expansion and increased proliferation of radial/apical progenitors and of neurogenic transit-amplifying Tbr2+ intermediate progenitors. Notably, our findings identify a previously unreported EGF-independent mechanism through which Lrig1 negatively regulates neural progenitor proliferation by modulating the FGF2-induced IL6/Jak2/Stat3 pathway, a molecular cascade that plays a pivotal role in the generation and maintenance of CPs. Consistently, Lrig1 knockout mice showed a significant increase in the density of pyramidal glutamatergic neurons placed in superficial layers 2 and 3 of the postnatal neocortex. Together, these results support a model in which Lrig1 regulates cortical neurogenesis by influencing the cycling activity of a set of progenitors that are temporally specified to produce upper layer glutamatergic neurons.
Collapse
Affiliation(s)
- Ana Paula De Vincenti
- Laboratorio de Neurociencia Molecular y Celular, Instituto de Biología Celular y Neurociencias (IBCN)-CONICET-UBA, Facultad de Medicina. Universidad de Buenos Aires (UBA), Buenos Aires CP1121, Argentina
| | - Antonela Bonafina
- Laboratorio de Neurociencia Molecular y Celular, Instituto de Biología Celular y Neurociencias (IBCN)-CONICET-UBA, Facultad de Medicina. Universidad de Buenos Aires (UBA), Buenos Aires CP1121, Argentina
- Fundación Instituto Leloir, Instituto de Investigaciones Bioquímicas de Buenos Aires, Buenos Aires C1405 BWE, Argentina
| | - Fernanda Ledda
- Fundación Instituto Leloir, Instituto de Investigaciones Bioquímicas de Buenos Aires, Buenos Aires C1405 BWE, Argentina
| | - Gustavo Paratcha
- Laboratorio de Neurociencia Molecular y Celular, Instituto de Biología Celular y Neurociencias (IBCN)-CONICET-UBA, Facultad de Medicina. Universidad de Buenos Aires (UBA), Buenos Aires CP1121, Argentina
| |
Collapse
|
4
|
Gasperoni JG, Tran SC, Grommen SVH, De Groef B, Dworkin S. The Role of PLAG1 in Mouse Brain Development and Neurogenesis. Mol Neurobiol 2024; 61:5851-5867. [PMID: 38240991 PMCID: PMC11249490 DOI: 10.1007/s12035-024-03943-w] [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: 01/10/2024] [Indexed: 07/16/2024]
Abstract
The pleomorphic adenoma gene 1 (Plag1) is a transcription factor involved in the regulation of growth and cellular proliferation. Here, we report the spatial distribution and functional implications of PLAG1 expression in the adult mouse brain. We identified Plag1 promoter-dependent β-galactosidase expression in various brain structures, including the hippocampus, cortex, choroid plexus, subcommisural organ, ependymal cells lining the third ventricle, medial and lateral habenulae and amygdala. We noted striking spatial-restriction of PLAG1 within the cornu ammonis (CA1) region of the hippocampus and layer-specific cortical expression, with abundant expression noted in all layers except layer 5. Furthermore, our study delved into the role of PLAG1 in neurodevelopment, focusing on its impact on neural stem/progenitor cell proliferation. Loss of Plag1 resulted in reduced proliferation and decreased production of neocortical progenitors in vivo, although ex vivo neurosphere experiments revealed no cell-intrinsic defects in the proliferative or neurogenic capacity of Plag1-deficient neural progenitors. Lastly, we explored potential target genes of PLAG1 in the cortex, identifying that Neurogenin 2 (Ngn2) was significantly downregulated in Plag1-deficient mice. In summary, our study provides novel insights into the spatial distribution of PLAG1 expression in the adult mouse brain and its potential role in neurodevelopment. These findings expand our understanding of the functional significance of PLAG1 within the brain, with potential implications for neurodevelopmental disorders and therapeutic interventions.
Collapse
Affiliation(s)
- Jemma G Gasperoni
- Department of Microbiology, Anatomy, Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, Victoria, 3086, Australia
| | - Stephanie C Tran
- Department of Microbiology, Anatomy, Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, Victoria, 3086, Australia
| | - Sylvia V H Grommen
- Department of Microbiology, Anatomy, Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, Victoria, 3086, Australia
- Department of Biology, KU Leuven, B3000, Leuven, Belgium
| | - Bert De Groef
- Department of Microbiology, Anatomy, Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, Victoria, 3086, Australia
- Department of Biology, KU Leuven, B3000, Leuven, Belgium
| | - Sebastian Dworkin
- Department of Microbiology, Anatomy, Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, Victoria, 3086, Australia.
| |
Collapse
|
5
|
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
|
6
|
Awad PN, Zerbi V, Johnson-Venkatesh EM, Damiani F, Pagani M, Markicevic M, Nickles S, Gozzi A, Umemori H, Fagiolini M. CDKL5 sculpts functional callosal connectivity to promote cognitive flexibility. Mol Psychiatry 2024; 29:1698-1709. [PMID: 36737483 PMCID: PMC11371650 DOI: 10.1038/s41380-023-01962-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 01/02/2023] [Accepted: 01/13/2023] [Indexed: 02/05/2023]
Abstract
Functional and structural connectivity alterations in short- and long-range projections have been reported across neurodevelopmental disorders (NDD). Interhemispheric callosal projection neurons (CPN) represent one of the major long-range projections in the brain, which are particularly important for higher-order cognitive function and flexibility. However, whether a causal relationship exists between interhemispheric connectivity alterations and cognitive deficits in NDD remains elusive. Here, we focused on CDKL5 Deficiency Disorder (CDD), a severe neurodevelopmental disorder caused by mutations in the X-linked Cyclin-dependent kinase-like 5 (CDKL5) gene. We found an increase in homotopic interhemispheric connectivity and functional hyperconnectivity across higher cognitive areas in adult male and female CDKL5-deficient mice by resting-state functional MRI (rs-fMRI) analysis. This was accompanied by an increase in the number of callosal synaptic inputs but decrease in local synaptic connectivity in the cingulate cortex of juvenile CDKL5-deficient mice, suggesting an impairment in excitatory synapse development and a differential role of CDKL5 across excitatory neuron subtypes. These deficits were associated with significant cognitive impairments in CDKL5 KO mice. Selective deletion of CDKL5 in the largest subtype of CPN likewise resulted in an increase of functional callosal inputs, without however significantly altering intracortical cingulate networks. Notably, such callosal-specific changes were sufficient to cause cognitive deficits. Finally, when CDKL5 was selectively re-expressed only in this CPN subtype, in otherwise CDKL5-deficient mice, it was sufficient to prevent the cognitive impairments of CDKL5 mutants. Together, these results reveal a novel role of CDKL5 by demonstrating that it is both necessary and sufficient for proper CPN connectivity and cognitive function and flexibility, and further validates a causal relationship between CPN dysfunction and cognitive impairment in a model of NDD.
Collapse
Affiliation(s)
- Patricia Nora Awad
- F. M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Valerio Zerbi
- Neural Control of Movement Lab, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- Neuro-X Institute, School of Engineering (STI), École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- CIBM Center for Biomedical Imaging, Lausanne, Switzerland
| | - Erin M Johnson-Venkatesh
- F. M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Francesca Damiani
- F. M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Marco Pagani
- Functional Neuroimaging Laboratory, Center for Neuroscience and Cognitive Systems, Istituto Italiano di Tecnologia, Rovereto, Italy
- Autism Center, Child Mind Institute, New York, NY, USA
| | - Marija Markicevic
- Neural Control of Movement Lab, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Sarah Nickles
- F. M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Alessandro Gozzi
- Functional Neuroimaging Laboratory, Center for Neuroscience and Cognitive Systems, Istituto Italiano di Tecnologia, Rovereto, Italy
| | - Hisashi Umemori
- F. M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Michela Fagiolini
- F. M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
- Hock E. Tan and K. Lisa Yang Center for Autism Research at Harvard University, Boston, MA, USA.
- International Research Center for Neurointelligence (IRCN), University of Tokyo Institutes for Advanced Study, Tokyo, Japan.
| |
Collapse
|
7
|
Zaidel DW, Fabri M. Editorial: The legacy of Dr. Roger W. Sperry: current advances in brain lateralization and interhemispheric transfer. Front Hum Neurosci 2024; 18:1433410. [PMID: 38868543 PMCID: PMC11167068 DOI: 10.3389/fnhum.2024.1433410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 05/20/2024] [Indexed: 06/14/2024] Open
Affiliation(s)
- Dahlia W. Zaidel
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Mara Fabri
- Dipartimento di Scienze della Vita e dell'Ambiente, Università Politecnica delle Marche, Ancona, Italy
| |
Collapse
|
8
|
K. C. R, Tiemroth AS, Thurmon AN, Meadows SM, Galazo MJ. Zmiz1 is a novel regulator of brain development associated with autism and intellectual disability. Front Psychiatry 2024; 15:1375492. [PMID: 38686122 PMCID: PMC11057416 DOI: 10.3389/fpsyt.2024.1375492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 03/26/2024] [Indexed: 05/02/2024] Open
Abstract
Neurodevelopmental disorders (NDDs) are a class of pathologies arising from perturbations in brain circuit formation and maturation with complex etiological triggers often classified as environmental and genetic. Neuropsychiatric conditions such as autism spectrum disorders (ASD), intellectual disability (ID), and attention deficit hyperactivity disorders (ADHD) are common NDDs characterized by their hereditary underpinnings and inherent heterogeneity. Genetic risk factors for NDDs are increasingly being identified in non-coding regions and proteins bound to them, including transcriptional regulators and chromatin remodelers. Importantly, de novo mutations are emerging as important contributors to NDDs and neuropsychiatric disorders. Recently, de novo mutations in transcriptional co-factor Zmiz1 or its regulatory regions have been identified in unrelated patients with syndromic ID and ASD. However, the role of Zmiz1 in brain development is unknown. Here, using publicly available databases and a Zmiz1 mutant mouse model, we reveal that Zmiz1 is highly expressed during embryonic brain development in mice and humans, and though broadly expressed across the brain, Zmiz1 is enriched in areas prominently impacted in ID and ASD such as cortex, hippocampus, and cerebellum. We investigated the relationship between Zmiz1 structure and pathogenicity of protein variants, the epigenetic marks associated with Zmiz1 regulation, and protein interactions and signaling pathways regulated by Zmiz1. Our analysis reveals that Zmiz1 regulates multiple developmental processes, including neurogenesis, neuron connectivity, and synaptic signaling. This work paves the way for future studies on the functions of Zmiz1 and highlights the importance of combining analysis of mouse models and human data.
Collapse
Affiliation(s)
- Rajan K. C.
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, United States
| | - Alina S. Tiemroth
- Tulane Brain Institute, Tulane University, New Orleans, LA, United States
| | - Abbigail N. Thurmon
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, United States
| | - Stryder M. Meadows
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, United States
- Tulane Brain Institute, Tulane University, New Orleans, LA, United States
| | - Maria J. Galazo
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, United States
- Tulane Brain Institute, Tulane University, New Orleans, LA, United States
| |
Collapse
|
9
|
Liu H, Zhong Y, Liu G, Su H, Liu Z, Wei J, Mo L, Tan C, Liu X, Chen L. Corpus callosum and cerebellum participate in semantic dysfunction of Parkinson's disease: a diffusion tensor imaging-based cross-sectional study. Neuroreport 2024; 35:366-373. [PMID: 38526949 DOI: 10.1097/wnr.0000000000002015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
Language dysfunction is common in Parkinson's disease (PD) patients, among which, the decline of semantic fluency is usually observed. This study aims to explore the relationship between white matter (WM) alterations and semantic fluency changes in PD patients. 127 PD patients from the Parkinson's Progression Markers Initiative cohort who received diffusion tensor imaging scanning, clinical assessment and semantic fluency test (SFT) were included. Tract-based special statistics, automated fiber quantification, graph-theoretical and network-based analyses were performed to analyze the correlation between WM structural changes, brain network features and semantic fluency in PD patients. Fractional anisotropy of corpus callosum, anterior thalamic radiation, inferior front-occipital fasciculus, and uncinate fasciculus, were positively correlated with SFT scores, while a negative correlation was identified between radial diffusion of the corpus callosum, inferior longitudinal fasciculus, and SFT scores. Automatic fiber quantification identified similar alterations with more details in these WM tracts. Brain network analysis positively correlated SFT scores with nodal efficiency of cerebellar lobule VIII, and nodal local efficiency of cerebellar lobule X. WM integrity and myelin integrity in the corpus callosum and several other language-related WM tracts may influence the semantic function in PD patients. Damage to the cerebellum lobule VIII and lobule X may also be involved in semantic dysfunction in PD patients.
Collapse
Affiliation(s)
- Hang Liu
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Seas A, Noor MS, Choi KS, Veerakumar A, Obatusin M, Dahill-Fuchel J, Tiruvadi V, Xu E, Riva-Posse P, Rozell CJ, Mayberg HS, McIntyre CC, Waters AC, Howell B. Subcallosal cingulate deep brain stimulation evokes two distinct cortical responses via differential white matter activation. Proc Natl Acad Sci U S A 2024; 121:e2314918121. [PMID: 38527192 PMCID: PMC10998591 DOI: 10.1073/pnas.2314918121] [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: 09/01/2023] [Accepted: 02/20/2024] [Indexed: 03/27/2024] Open
Abstract
Subcallosal cingulate (SCC) deep brain stimulation (DBS) is an emerging therapy for refractory depression. Good clinical outcomes are associated with the activation of white matter adjacent to the SCC. This activation produces a signature cortical evoked potential (EP), but it is unclear which of the many pathways in the vicinity of SCC is responsible for driving this response. Individualized biophysical models were built to achieve selective engagement of two target bundles: either the forceps minor (FM) or cingulum bundle (CB). Unilateral 2 Hz stimulation was performed in seven patients with treatment-resistant depression who responded to SCC DBS, and EPs were recorded using 256-sensor scalp electroencephalography. Two distinct EPs were observed: a 120 ms symmetric response spanning both hemispheres and a 60 ms asymmetrical EP. Activation of FM correlated with the symmetrical EPs, while activation of CB was correlated with the asymmetrical EPs. These results support prior model predictions that these two pathways are predominantly activated by clinical SCC DBS and provide first evidence of a link between cortical EPs and selective fiber bundle activation.
Collapse
Affiliation(s)
- Andreas Seas
- Department of Biomedical Engineering, Duke University, Durham, NC27708
- Department of Neurosurgery, Duke University, Durham, NC27708
| | - M. Sohail Noor
- Department of Biomedical Engineering, Duke University, Durham, NC27708
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH10900
| | - Ki Sueng Choi
- Nash Family Center for Advanced Circuit Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY10029
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA30329
| | - Ashan Veerakumar
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA30329
| | - Mosadoluwa Obatusin
- Nash Family Center for Advanced Circuit Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY10029
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA30329
| | - Jacob Dahill-Fuchel
- Nash Family Center for Advanced Circuit Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY10029
| | - Vineet Tiruvadi
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA30329
| | - Elisa Xu
- Nash Family Center for Advanced Circuit Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY10029
| | - Patricio Riva-Posse
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA30329
| | - Christopher J. Rozell
- School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA30332
| | - Helen S. Mayberg
- Nash Family Center for Advanced Circuit Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY10029
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA30329
| | - Cameron C. McIntyre
- Department of Biomedical Engineering, Duke University, Durham, NC27708
- Department of Neurosurgery, Duke University, Durham, NC27708
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH10900
| | - Allison C. Waters
- Nash Family Center for Advanced Circuit Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY10029
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA30329
| | - Bryan Howell
- Department of Biomedical Engineering, Duke University, Durham, NC27708
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH10900
| |
Collapse
|
11
|
Poulopoulos A, Davis P, Brandenburg C, Itoh Y, Galazo MJ, Greig LC, Romanowski AJ, Budnik B, Macklis JD. Symmetry in levels of axon-axon homophilic adhesion establishes topography in the corpus callosum and development of connectivity between brain hemispheres. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.28.587108. [PMID: 38585721 PMCID: PMC10996634 DOI: 10.1101/2024.03.28.587108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Specific and highly diverse connectivity between functionally specialized regions of the nervous system is controlled at multiple scales, from anatomically organized connectivity following macroscopic axon tracts to individual axon target-finding and synapse formation. Identifying mechanisms that enable entire subpopulations of related neurons to project their axons with regional specificity within stereotyped tracts to form appropriate long-range connectivity is key to understanding brain development, organization, and function. Here, we investigate how axons of the cerebral cortex form precise connections between the two cortical hemispheres via the corpus callosum. We identify topographic principles of the developing trans-hemispheric callosal tract that emerge through intrinsic guidance executed by growing axons in the corpus callosum within the first postnatal week in mice. Using micro-transplantation of regionally distinct neurons, subtype-specific growth cone purification, subcellular proteomics, and in utero gene manipulation, we investigate guidance mechanisms of transhemispheric axons. We find that adhesion molecule levels instruct tract topography and target field guidance. We propose a model in which transcallosal axons in the developing brain perform a "handshake" that is guided through co-fasciculation with symmetric contralateral axons, resulting in the stereotyped homotopic connectivity between the brain's hemispheres.
Collapse
Affiliation(s)
- Alexandros Poulopoulos
- Department of Stem Cell and Regenerative Biology, and Center for Brain Science, Harvard University, Cambridge, MA, USA
- University of Maryland School of Medicine, Department of Pharmacology, Baltimore, MD, USA
| | - Patrick Davis
- Department of Stem Cell and Regenerative Biology, and Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Cheryl Brandenburg
- University of Maryland School of Medicine, Department of Pharmacology, Baltimore, MD, USA
| | - Yasuhiro Itoh
- Department of Stem Cell and Regenerative Biology, and Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Maria J. Galazo
- Department of Stem Cell and Regenerative Biology, and Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Luciano C. Greig
- Department of Stem Cell and Regenerative Biology, and Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Andrea J. Romanowski
- University of Maryland School of Medicine, Department of Pharmacology, Baltimore, MD, USA
| | - Bogdan Budnik
- Harvard Center for Mass Spectrometry, Harvard University, Cambridge, MA, USA
| | - Jeffrey D. Macklis
- Department of Stem Cell and Regenerative Biology, and Center for Brain Science, Harvard University, Cambridge, MA, USA
| |
Collapse
|
12
|
Hirata T. Olfactory information processing viewed through mitral and tufted cell-specific channels. Front Neural Circuits 2024; 18:1382626. [PMID: 38523698 PMCID: PMC10957668 DOI: 10.3389/fncir.2024.1382626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 02/29/2024] [Indexed: 03/26/2024] Open
Abstract
Parallel processing is a fundamental strategy of sensory coding. Through this processing, unique and distinct features of sensations are computed and projected to the central targets. This review proposes that mitral and tufted cells, which are the second-order projection neurons in the olfactory bulb, contribute to parallel processing within the olfactory system. Based on anatomical and functional evidence, I discuss potential features that could be conveyed through the unique channel formed by these neurons.
Collapse
Affiliation(s)
- Tatsumi Hirata
- Brain Function Laboratory, National Institute of Genetics, SOKENDAI, Mishima, Japan
| |
Collapse
|
13
|
Rodríguez-Pérez LM, López-de-San-Sebastián J, de Diego I, Smith A, Roales-Buján R, Jiménez AJ, Paez-Gonzalez P. A selective defect in the glial wedge as part of the neuroepithelium disruption in hydrocephalus development in the mouse hyh model is associated with complete corpus callosum dysgenesis. Front Cell Neurosci 2024; 18:1330412. [PMID: 38450283 PMCID: PMC10915275 DOI: 10.3389/fncel.2024.1330412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 02/08/2024] [Indexed: 03/08/2024] Open
Abstract
Introduction Dysgenesis of the corpus callosum is present in neurodevelopmental disorders and coexists with hydrocephalus in several human congenital syndromes. The mechanisms that underlie the etiology of congenital hydrocephalus and agenesis of the corpus callosum when they coappear during neurodevelopment persist unclear. In this work, the mechanistic relationship between both disorders is investigated in the hyh mouse model for congenital hydrocephalus, which also develops agenesis of the corpus callosum. In this model, hydrocephalus is generated by a defective program in the development of neuroepithelium during its differentiation into radial glial cells. Methods In this work, the populations implicated in the development of the corpus callosum (callosal neurons, pioneering axons, glial wedge cells, subcallosal sling and indusium griseum glial cells) were studied in wild-type and hyh mutant mice. Immunohistochemistry, mRNA in situ hybridization, axonal tracing experiments, and organotypic cultures from normal and hyh mouse embryos were used. Results Our results show that the defective program in the neuroepithelium/radial glial cell development in the hyh mutant mouse selectively affects the glial wedge cells. The glial wedge cells are necessary to guide the pioneering axons as they approach the corticoseptal boundary. Our results show that the pioneering callosal axons arising from neurons in the cingulate cortex can extend projections to the interhemispheric midline in normal and hyh mice. However, pioneering axons in the hyh mutant mouse, when approaching the area corresponding to the damaged glial wedge cell population, turned toward the ipsilateral lateral ventricle. This defect occurred before the appearance of ventriculomegaly. Discussion In conclusion, the abnormal development of the ventricular zone, which appears to be inherent to the etiology of several forms of congenital hydrocephalus, can explain, in some cases, the common association between hydrocephalus and corpus callosum dysgenesis. These results imply that further studies may be needed to understand the corpus callosum dysgenesis etiology when it concurs with hydrocephalus.
Collapse
Affiliation(s)
- Luis-Manuel Rodríguez-Pérez
- Departamento de Fisiología Humana, Histología Humana, Anatomía Patológica y Educación Física y Deportiva, Universidad de Málaga, Malaga, Spain
- Instituto de Investigación Biomédica de Málaga (IBIMA), Malaga, Spain
| | | | - Isabel de Diego
- Departamento de Anatomía y Medicina Legal e Historia de la Ciencia, Universidad de Málaga, Malaga, Spain
| | - Aníbal Smith
- Departamento de Anatomía y Medicina Legal e Historia de la Ciencia, Universidad de Málaga, Malaga, Spain
| | - Ruth Roales-Buján
- Departamento de Biología Celular, Genética y Fisiología, Universidad de Málaga, Malaga, Spain
| | - Antonio J. Jiménez
- Instituto de Investigación Biomédica de Málaga (IBIMA), Malaga, Spain
- Departamento de Biología Celular, Genética y Fisiología, Universidad de Málaga, Malaga, Spain
| | - Patricia Paez-Gonzalez
- Instituto de Investigación Biomédica de Málaga (IBIMA), Malaga, Spain
- Departamento de Biología Celular, Genética y Fisiología, Universidad de Málaga, Malaga, Spain
| |
Collapse
|
14
|
Kitazawa M. Evolution of the nervous system by acquisition of retrovirus-derived genes in mammals. Genes Genet Syst 2024; 98:321-336. [PMID: 38220159 DOI: 10.1266/ggs.23-00197] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2024] Open
Abstract
In the course of evolution, the most highly developed organ is likely the brain, which has become more complex over time and acquired diverse forms and functions in different species. In particular, mammals have developed complex and high-functioning brains, and it has been reported that several genes derived from retroviruses were involved in mammalian brain evolution, that is, generating the complexity of the nervous system. Especially, the sushi-ichi-related retrotransposon homolog (SIRH)/retrotransposon gag-like (RTL) genes have been suggested to play a role in the evolutionary processes shaping brain morphology and function in mammals. Genetic mutation and altered expression of genes are linked to neurological disorders, highlighting how the acquisition of virus-derived genes in mammals has both driven brain evolution and imposed a susceptibility to diseases. This review provides an overview of the functions, diversity, evolution and diseases associated with SIRH/RTL genes in the nervous system. The contribution of retroviruses to brain evolution is an important research topic in evolutionary biology and neuroscience, and further insights are expected to be gained through future studies.
Collapse
Affiliation(s)
- Moe Kitazawa
- School of BioSciences, Faculty of Science, The University of Melbourne
| |
Collapse
|
15
|
Pal S, Lim JWC, Richards LJ. Diverse axonal morphologies of individual callosal projection neurons reveal new insights into brain connectivity. Curr Opin Neurobiol 2024; 84:102837. [PMID: 38271848 PMCID: PMC11265515 DOI: 10.1016/j.conb.2023.102837] [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: 07/27/2023] [Accepted: 12/20/2023] [Indexed: 01/27/2024]
Abstract
In the mature brain, functionally distinct areas connect to specific targets, mediating network activity required for function. New insights are still occurring regarding how specific connectivity occurs in the developing brain. Decades of work have revealed important insights into the molecular and genetic mechanisms regulating cell type specification in the brain. This work classified long-range projection neurons of the cerebral cortex into three major classes based on their primary target (e.g. subcortical, intracortical, and interhemispheric projections). However, painstaking single-cell mapping reveals that long-range projection neurons of the corpus callosum connect to multiple and overlapping ipsilateral and contralateral targets with often highly branched axons. In addition, their scRNA transcriptomes are highly variable, making it difficult to identify meaningful subclasses. This work has prompted us to reexamine how cortical projection neurons that comprise the corpus callosum are currently classified and how this stunning array of variability might be achieved during development.
Collapse
Affiliation(s)
- Suranjana Pal
- Department of Neuroscience, Washington University in St Louis School of Medicine, St Louis, MO 63110, USA. https://twitter.com/PalSuranjana
| | - Jonathan W C Lim
- Department of Neuroscience, Washington University in St Louis School of Medicine, St Louis, MO 63110, USA
| | - Linda J Richards
- Department of Neuroscience, Washington University in St Louis School of Medicine, St Louis, MO 63110, USA.
| |
Collapse
|
16
|
Porcu M, Cocco L, Marrosu F, Cau R, Suri JS, Qi Y, Pineda V, Bosin A, Malloci G, Ruggerone P, Puig J, Saba L. Impact of corpus callosum integrity on functional interhemispheric connectivity and cognition in healthy subjects. Brain Imaging Behav 2024; 18:141-158. [PMID: 37955809 DOI: 10.1007/s11682-023-00814-1] [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] [Accepted: 10/15/2023] [Indexed: 11/14/2023]
Abstract
To examine the corpus callosum's (CC) integrity in terms of fractional anisotropy (FA) and how it affects resting-state hemispheric connectivity (rs-IHC) and cognitive function in healthy individuals. Sixty-eight healthy individuals were recruited for the study. The global FA (gFA) and FA values of each CC tract (forceps minor, body, tapetum, and forceps major) were evaluated using diffusion-weighted imaging (DWI) sequences. The homotopic functional connectivity technique was used to quantify the effects of FA in the CC tracts on bilateral functional connectivity, including the confounding effect of gFA. Brain regions with higher or lower rs-IHC were identified using the threshold-free cluster enhancement family-wise error-corrected p-value of 0.05. The null hypothesis was rejected if the p-value was ≤ 0.05 for the nonparametric partial correlation technique. Several clusters of increased rs-IHC were identified in relation to the FA of individual CC tracts, each with a unique topographic distribution and extension. Only forceps minor FA values correlated with cognitive scores. The integrity of CC influences rs-IHC differently in healthy subjects. Specifically, forceps minor anisotropy impacts rs-IHC and cognition more than other CC tracts do.
Collapse
Affiliation(s)
- Michele Porcu
- Department of Radiology, AOU Cagliari, University of Cagliari, Cagliari, Italy.
- Department of Medical Imaging, Azienda Ospedaliera Universitaria di Cagliari, S.S: 554, Km 4,500 - CAP, Monserrato, 09042, Cagliari, Italy.
| | - Luigi Cocco
- Department of Radiology, AOU Cagliari, University of Cagliari, Cagliari, Italy
| | - Francesco Marrosu
- Department of Radiology, AOU Cagliari, University of Cagliari, Cagliari, Italy
| | - Riccardo Cau
- Department of Radiology, AOU Cagliari, University of Cagliari, Cagliari, Italy
| | - Jasjit S Suri
- Stroke Monitoring and Diagnostic Division, AtheroPoint™, Roseville, CA, USA
| | - Yang Qi
- Xuanwu Hospital, Capital Medical University, No.45 Changchun Street, Xicheng District, Beijing, China
| | - Victor Pineda
- Department of Medical Sciences, Hospital Universitari de Girona Dr Josep Trueta, Girona, Spain
- Department of Radiology (IDI), Hospital Universitari de Girona Dr Josep Trueta, Girona, Spain
| | - Andrea Bosin
- Department of Physics, University of Cagliari, Cagliari, Italy
| | | | - Paolo Ruggerone
- Department of Physics, University of Cagliari, Cagliari, Italy
| | - Josep Puig
- Department of Medical Sciences, Hospital Universitari de Girona Dr Josep Trueta, Girona, Spain
- Department of Radiology (IDI), Hospital Universitari de Girona Dr Josep Trueta, Girona, Spain
| | - Luca Saba
- Department of Radiology, AOU Cagliari, University of Cagliari, Cagliari, Italy
| |
Collapse
|
17
|
Hsieh CCJ, Lo YC, Wang HH, Shen HY, Chen YY, Lee YC. Amelioration of the brain structural connectivity is accompanied with changes of gut microbiota in a tuberous sclerosis complex mouse model. Transl Psychiatry 2024; 14:68. [PMID: 38296969 PMCID: PMC10830571 DOI: 10.1038/s41398-024-02752-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/04/2024] [Accepted: 01/08/2024] [Indexed: 02/02/2024] Open
Abstract
Tuberous sclerosis complex (TSC) is a genetic disease that causes benign tumors and dysfunctions in many organs, including the brain. Aside from the brain malformations, many individuals with TSC exhibit neuropsychiatric symptoms. Among these symptoms, autism spectrum disorder (ASD) is one of the most common co-morbidities, affecting up to 60% of the population. Past neuroimaging studies strongly suggested that the impairments in brain connectivity contribute to ASD, whether or not TSC-related. Specifically, the tract-based diffusion tensor imaging (DTI) analysis provides information on the fiber integrity and has been used to study the neuropathological changes in the white matter of TSC patients with ASD symptoms. In our previous study, curcumin, a diet-derived mTOR inhibitor has been shown to effectively mitigate learning and memory deficits and anxiety-like behavior in Tsc2+/- mice via inhibiting astroglial proliferation. Recently, gut microbiota, which is greatly influenced by the diet, has been considered to play an important role in regulating several components of the central nervous system, including glial functions. In this study, we showed that the abnormal social behavior in the Tsc2+/- mice can be ameliorated by the dietary curcumin treatment. Second, using tract-based DTI analysis, we found that the Tsc2+/- mice exhibited altered fractional anisotropy, axial and radial diffusivities of axonal bundles connecting the prefrontal cortex, nucleus accumbens, hypothalamus, and amygdala, indicating a decreased brain network. Third, the dietary curcumin treatment improved the DTI metrics, in accordance with changes in the gut microbiota composition. At the bacterial phylum level, we showed that the abundances of Actinobacteria, Verrucomicrobia, and Tenericutes were significantly correlated with the DTI metrics FA, AD, and RD, respectively. Finally, we revealed that the expression of myelin-associated proteins, myelin bassic protein (MBP) and proteolipid protein (PLP) was increased after the treatment. Overall, we showed a strong correlation between structural connectivity alterations and social behavioral deficits, as well as the diet-dependent changes in gut microbiota composition.
Collapse
Affiliation(s)
| | - Yu-Chun Lo
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Neuroscience Research Center, Taipei Medical University, Taipei, Taiwan
| | - Hsin-Hui Wang
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Hsin-Ying Shen
- Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan
| | - You-Yin Chen
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| | - Yi-Chao Lee
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.
- Neuroscience Research Center, Taipei Medical University, Taipei, Taiwan.
- International Master Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
18
|
Zhang Y, Jin Y, Li J, Yan Y, Wang T, Wang X, Li Z, Qin X. CXCL14 as a Key Regulator of Neuronal Development: Insights from Its Receptor and Multi-Omics Analysis. Int J Mol Sci 2024; 25:1651. [PMID: 38338930 PMCID: PMC10855946 DOI: 10.3390/ijms25031651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 01/22/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
CXCL14 is not only involved in the immune process but is also closely related to neurodevelopment according to its molecular evolution. However, what role it plays in neurodevelopment remains unclear. In the present research, we found that, by crossbreeding CXCL14+/- and CXCL14-/- mice, the number of CXCL14-/- mice in their offspring was lower than the Mendelian frequency; CXCL14-/- mice had significantly fewer neurons in the external pyramidal layer of cortex than CXCL14+/- mice; and CXCL14 may be involved in synaptic plasticity, neuron projection, and chemical synaptic transmission based on analysis of human clinical transcriptome data. The expression of CXCL14 was highest at day 14.5 in the embryonic phase and after birth in the mRNA and protein levels. Therefore, we hypothesized that CXCL14 promotes the development of neurons in the somatic layer of the pyramidal cells of mice cortex on embryonic day 14.5. In order to further explore its mechanism, CXCR4 and CXCR7 were suggested as receptors by Membrane-Anchored Ligand and Receptor Yeast Two-Hybrid technology. Through metabolomic techniques, we inferred that CXCL14 promotes the development of neurons by regulating fatty acid anabolism and glycerophospholipid anabolism.
Collapse
Affiliation(s)
- Yinjie Zhang
- Modern Research Center for Traditional Chinese Medicine, The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Road, Taiyuan 030006, China (T.W.)
| | - Yue Jin
- Modern Research Center for Traditional Chinese Medicine, The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Road, Taiyuan 030006, China (T.W.)
| | - Jingjing Li
- Engineering Research Center of Cell & Therapeutic Antibody, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yan Yan
- Modern Research Center for Traditional Chinese Medicine, The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Road, Taiyuan 030006, China (T.W.)
| | - Ting Wang
- Modern Research Center for Traditional Chinese Medicine, The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Road, Taiyuan 030006, China (T.W.)
| | - Xuanlin Wang
- Modern Research Center for Traditional Chinese Medicine, The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Road, Taiyuan 030006, China (T.W.)
| | - Zhenyu Li
- Modern Research Center for Traditional Chinese Medicine, The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Road, Taiyuan 030006, China (T.W.)
| | - Xuemei Qin
- Modern Research Center for Traditional Chinese Medicine, The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Road, Taiyuan 030006, China (T.W.)
| |
Collapse
|
19
|
Yang S, Niou ZX, Enriquez A, LaMar J, Huang JY, Ling K, Jafar-Nejad P, Gilley J, Coleman MP, Tennessen JM, Rangaraju V, Lu HC. NMNAT2 supports vesicular glycolysis via NAD homeostasis to fuel fast axonal transport. Mol Neurodegener 2024; 19:13. [PMID: 38282024 PMCID: PMC10823734 DOI: 10.1186/s13024-023-00690-9] [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: 05/09/2023] [Accepted: 11/28/2023] [Indexed: 01/30/2024] Open
Abstract
BACKGROUND Bioenergetic maladaptations and axonopathy are often found in the early stages of neurodegeneration. Nicotinamide adenine dinucleotide (NAD), an essential cofactor for energy metabolism, is mainly synthesized by Nicotinamide mononucleotide adenylyl transferase 2 (NMNAT2) in CNS neurons. NMNAT2 mRNA levels are reduced in the brains of Alzheimer's, Parkinson's, and Huntington's disease. Here we addressed whether NMNAT2 is required for axonal health of cortical glutamatergic neurons, whose long-projecting axons are often vulnerable in neurodegenerative conditions. We also tested if NMNAT2 maintains axonal health by ensuring axonal ATP levels for axonal transport, critical for axonal function. METHODS We generated mouse and cultured neuron models to determine the impact of NMNAT2 loss from cortical glutamatergic neurons on axonal transport, energetic metabolism, and morphological integrity. In addition, we determined if exogenous NAD supplementation or inhibiting a NAD hydrolase, sterile alpha and TIR motif-containing protein 1 (SARM1), prevented axonal deficits caused by NMNAT2 loss. This study used a combination of techniques, including genetics, molecular biology, immunohistochemistry, biochemistry, fluorescent time-lapse imaging, live imaging with optical sensors, and anti-sense oligos. RESULTS We provide in vivo evidence that NMNAT2 in glutamatergic neurons is required for axonal survival. Using in vivo and in vitro studies, we demonstrate that NMNAT2 maintains the NAD-redox potential to provide "on-board" ATP via glycolysis to vesicular cargos in distal axons. Exogenous NAD+ supplementation to NMNAT2 KO neurons restores glycolysis and resumes fast axonal transport. Finally, we demonstrate both in vitro and in vivo that reducing the activity of SARM1, an NAD degradation enzyme, can reduce axonal transport deficits and suppress axon degeneration in NMNAT2 KO neurons. CONCLUSION NMNAT2 ensures axonal health by maintaining NAD redox potential in distal axons to ensure efficient vesicular glycolysis required for fast axonal transport.
Collapse
Affiliation(s)
- Sen Yang
- The Linda and Jack Gill Center for Biomolecular Sciences, Indiana University, Bloomington, IN, 47405, USA
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, USA
- Program in Neuroscience, Indiana University, Bloomington, IN, 47405, USA
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, 33458, USA
| | - Zhen-Xian Niou
- The Linda and Jack Gill Center for Biomolecular Sciences, Indiana University, Bloomington, IN, 47405, USA
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, USA
- Program in Neuroscience, Indiana University, Bloomington, IN, 47405, USA
| | - Andrea Enriquez
- The Linda and Jack Gill Center for Biomolecular Sciences, Indiana University, Bloomington, IN, 47405, USA
- Program in Neuroscience, Indiana University, Bloomington, IN, 47405, USA
| | - Jacob LaMar
- The Linda and Jack Gill Center for Biomolecular Sciences, Indiana University, Bloomington, IN, 47405, USA
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, USA
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, 33458, USA
- Present address: Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, 33458, USA
| | - Jui-Yen Huang
- The Linda and Jack Gill Center for Biomolecular Sciences, Indiana University, Bloomington, IN, 47405, USA
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, USA
| | - Karen Ling
- Neuroscience Drug Discovery, Ionis Pharmaceuticals, Inc., 2855, Gazelle Court, Carlsbad, CA, 92010, USA
| | - Paymaan Jafar-Nejad
- Neuroscience Drug Discovery, Ionis Pharmaceuticals, Inc., 2855, Gazelle Court, Carlsbad, CA, 92010, USA
| | - Jonathan Gilley
- Department of Clinical Neuroscience, Cambridge University, Cambridge, UK
| | - Michael P Coleman
- Department of Clinical Neuroscience, Cambridge University, Cambridge, UK
| | - Jason M Tennessen
- Department of Biology, Indiana University, Bloomington, IN, 47405, USA
| | - Vidhya Rangaraju
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, 33458, USA
| | - Hui-Chen Lu
- The Linda and Jack Gill Center for Biomolecular Sciences, Indiana University, Bloomington, IN, 47405, USA.
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, USA.
- Program in Neuroscience, Indiana University, Bloomington, IN, 47405, USA.
| |
Collapse
|
20
|
Veeraraghavan P, Engmann AK, Hatch JJ, Itoh Y, Nguyen D, Addison T, Macklis JD. Dynamic subtype- and context-specific subcellular RNA regulation in growth cones of developing neurons of the cerebral cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.24.559186. [PMID: 38328182 PMCID: PMC10849483 DOI: 10.1101/2023.09.24.559186] [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
Molecular mechanisms that cells employ to compartmentalize function via localization of function-specific RNA and translation are only partially elucidated. We investigate long-range projection neurons of the cerebral cortex as highly polarized exemplars to elucidate dynamic regulation of RNA localization, stability, and translation within growth cones (GCs), leading tips of growing axons. Comparison of GC-localized transcriptomes between two distinct subtypes of projection neurons- interhemispheric-callosal and corticothalamic- across developmental stages identifies both distinct and shared subcellular machinery, and intriguingly highlights enrichment of genes associated with neurodevelopmental and neuropsychiatric disorders. Developmental context-specific components of GC-localized transcriptomes identify known and novel potential regulators of distinct phases of circuit formation: long-distance growth, target area innervation, and synapse formation. Further, we investigate mechanisms by which transcripts are enriched and dynamically regulated in GCs, and identify GC-enriched motifs in 3' untranslated regions. As one example, we identify cytoplasmic adenylation element binding protein 4 (CPEB4), an RNA binding protein regulating localization and translation of mRNAs encoding molecular machinery important for axonal branching and complexity. We also identify RNA binding motif single stranded interacting protein 1 (RBMS1) as a dynamically expressed regulator of RNA stabilization that enables successful callosal circuit formation. Subtly aberrant associative and integrative cortical circuitry can profoundly affect cortical function, often causing neurodevelopmental and neuropsychiatric disorders. Elucidation of context-specific subcellular RNA regulation for GC- and soma-localized molecular controls over precise circuit development, maintenance, and function offers generalizable insights for other polarized cells, and might contribute substantially to understanding neurodevelopmental and behavioral-cognitive disorders and toward targeted therapeutics.
Collapse
Affiliation(s)
- Priya Veeraraghavan
- Department of Stem Cell and Regenerative Biology, and Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Anne K. Engmann
- Department of Stem Cell and Regenerative Biology, and Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - John J. Hatch
- Department of Stem Cell and Regenerative Biology, and Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Yasuhiro Itoh
- Department of Stem Cell and Regenerative Biology, and Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Duane Nguyen
- Department of Stem Cell and Regenerative Biology, and Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Thomas Addison
- Department of Stem Cell and Regenerative Biology, and Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Jeffrey D. Macklis
- Department of Stem Cell and Regenerative Biology, and Center for Brain Science, Harvard University, Cambridge, MA, USA
| |
Collapse
|
21
|
Gavrish M, Kustova A, Celis Suescún JC, Bessa P, Mitina N, Tarabykin V. Molecular mechanisms of corpus callosum development: a four-step journey. Front Neuroanat 2024; 17:1276325. [PMID: 38298831 PMCID: PMC10827913 DOI: 10.3389/fnana.2023.1276325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 12/18/2023] [Indexed: 02/02/2024] Open
Abstract
The Corpus Callosum (CC) is a bundle of axons connecting the cerebral hemispheres. It is the most recent structure to have appeared during evolution of placental mammals. Its development is controlled by a very complex interplay of many molecules. In humans it contains almost 80% of all commissural axons in the brain. The formation of the CC can be divided into four main stages, each controlled by numerous intracellular and extracellular molecular factors. First, a newborn neuron has to specify an axon, leave proliferative compartments, the Ventricular Zone (VZ) and Subventricular Zone (SVZ), migrate through the Intermediate Zone (IZ), and then settle at the Cortical Plate (CP). During the second stage, callosal axons navigate toward the midline within a compact bundle. Next stage is the midline crossing into contralateral hemisphere. The last step is targeting a defined area and synapse formation. This review provides an insight into these four phases of callosal axons development, as well as a description of the main molecular players involved.
Collapse
Affiliation(s)
- Maria Gavrish
- Laboratory of Genetics of Brain Development, Research Institute of Neurosciences, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Angelina Kustova
- Laboratory of Genetics of Brain Development, Research Institute of Neurosciences, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Juan C. Celis Suescún
- Laboratory of Genetics of Brain Development, Research Institute of Neurosciences, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Paraskevi Bessa
- Charité Hospital, Institute of Cell Biology and Neurobiology, Berlin, Germany
| | - Natalia Mitina
- Laboratory of Genetics of Brain Development, Research Institute of Neurosciences, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Victor Tarabykin
- Charité Hospital, Institute of Cell Biology and Neurobiology, Berlin, Germany
| |
Collapse
|
22
|
Shen Z, Yang J, Zhang Q, Wang K, Lv X, Hu X, Ma J, Shi SH. How variable progenitor clones construct a largely invariant neocortex. Natl Sci Rev 2024; 11:nwad247. [PMID: 38274004 PMCID: PMC10810685 DOI: 10.1093/nsr/nwad247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/31/2023] [Accepted: 09/04/2023] [Indexed: 01/27/2024] Open
Abstract
The neocortex contains a vast collection of diverse neurons organized into distinct layers. While nearly all neocortical neurons are generated by radial glial progenitors (RGPs), it remains largely unclear how a complex yet organized neocortex is constructed reliably and robustly. Here, we show that the division behavior and neuronal output of RGPs are highly constrained with patterned variabilities to support the reliable and robust construction of the mouse neocortex. The neurogenic process of RGPs can be well-approximated by a consistent Poisson-like process unfolding over time, producing deep to superficial layer neurons progressively. The exact neuronal outputs regarding layer occupation are variable; yet, this variability is constrained systematically to support all layer formation, largely reflecting the variable intermediate progenitor generation and RGP neurogenic entry and exit timing differences. Together, these results define the fundamental features of neocortical neurogenesis with a balanced reliability and variability for the construction of the complex neocortex.
Collapse
Affiliation(s)
- Zhongfu Shen
- New Cornerstone Science Laboratory, IDG/McGovern Institute for Brain Research, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Jiajun Yang
- New Cornerstone Science Laboratory, IDG/McGovern Institute for Brain Research, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Qiangqiang Zhang
- New Cornerstone Science Laboratory, IDG/McGovern Institute for Brain Research, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Kuiyu Wang
- Department of Computer Sciences, Tsinghua University, Beijing 100084, China
| | - Xiaohui Lv
- New Cornerstone Science Laboratory, IDG/McGovern Institute for Brain Research, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing 100084, China
- College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Xiaolin Hu
- Department of Computer Sciences, Tsinghua University, Beijing 100084, China
| | - Jian Ma
- New Cornerstone Science Laboratory, IDG/McGovern Institute for Brain Research, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Song-Hai Shi
- New Cornerstone Science Laboratory, IDG/McGovern Institute for Brain Research, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing 100084, China
- Chinese Institute for Brain Research, Beijing 102206, China
| |
Collapse
|
23
|
Zhou YT, An DD, Xu YX, Zhou Y, Li QQ, Dai HB, Zhang XN, Wang Y, Lou M, Chen Z, Hu WW. Activation of glutamatergic neurons in the somatosensory cortex promotes remyelination in ischemic vascular dementia. FUNDAMENTAL RESEARCH 2024; 4:188-198. [PMID: 38933843 PMCID: PMC11197523 DOI: 10.1016/j.fmre.2022.08.007] [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: 03/05/2022] [Revised: 07/28/2022] [Accepted: 08/03/2022] [Indexed: 11/29/2022] Open
Abstract
Chronic cerebral hypoperfusion can cause progressive demyelination as well as ischemic vascular dementia, however no effective treatments are available. Here, based on magnetic resonance imaging studies of patients with white matter damage, we found that this damage is associated with disorganized cortical structure. In a mouse model, optogenetic activation of glutamatergic neurons in the somatosensory cortex significantly promoted oligodendrocyte progenitor cell (OPC) proliferation, remyelination in the corpus callosum, and recovery of cognitive ability after cerebral hypoperfusion. The therapeutic effect of such stimulation was restricted to the upper layers of the cortex, but also spanned a wide time window after ischemia. Mechanistically, enhancement of glutamatergic neuron-OPC functional synaptic connections is required to achieve the protection effect of activating cortical glutamatergic neurons. Additionally, skin stroking, an easier method to translate into clinical practice, activated the somatosensory cortex, thereby promoting OPC proliferation, remyelination and cognitive recovery following cerebral hypoperfusion. In summary, we demonstrated that activating glutamatergic neurons in the somatosensory cortex promotes the proliferation of OPCs and remyelination to recover cognitive function after chronic cerebral hypoperfusion. It should be noted that this activation may provide new approaches for treating ischemic vascular dementia via the precise regulation of glutamatergic neuron-OPC circuits.
Collapse
Affiliation(s)
- Yi-Ting Zhou
- Department of Pharmacology and Department of Pharmacy of the Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, College of Pharmaceutical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China
- Department of Pharmacy, Sir Run Run Shaw Hospital, Hangzhou 310012, China
| | - Da-Dao An
- Department of Pharmacology and Department of Pharmacy of the Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, College of Pharmaceutical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yi-Xin Xu
- Department of Pharmacology and Department of Pharmacy of the Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, College of Pharmaceutical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Ying Zhou
- Department of Neurology, the Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou 310003, China
| | - Qing-Qing Li
- Department of Neurology, the Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou 310003, China
| | - Hai-Bin Dai
- Department of Pharmacology and Department of Pharmacy of the Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, College of Pharmaceutical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xiang-Nan Zhang
- Department of Pharmacology and Department of Pharmacy of the Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, College of Pharmaceutical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yi Wang
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Min Lou
- Department of Neurology, the Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou 310003, China
| | - Zhong Chen
- Department of Pharmacology and Department of Pharmacy of the Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, College of Pharmaceutical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Wei-Wei Hu
- Department of Pharmacology and Department of Pharmacy of the Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, College of Pharmaceutical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China
| |
Collapse
|
24
|
Li XH, Shi W, Chen QY, Hao S, Miao HH, Miao Z, Xu F, Bi GQ, Zhuo M. Activation of the glutamatergic cingulate cortical-cortical connection facilitates pain in adult mice. Commun Biol 2023; 6:1247. [PMID: 38071375 PMCID: PMC10710420 DOI: 10.1038/s42003-023-05589-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 11/14/2023] [Indexed: 12/18/2023] Open
Abstract
The brain consists of the left and right cerebral hemispheres and both are connected by callosal projections. Less is known about the basic mechanism of this cortical-cortical connection and its functional importance. Here we investigate the cortical-cortical connection between the bilateral anterior cingulate cortex (ACC) by using the classic electrophysiological and optogenetic approach. We find that there is a direct synaptic projection from one side ACC to the contralateral ACC. Glutamate is the major excitatory transmitter for bilateral ACC connection, including projections to pyramidal cells in superficial (II/III) and deep (V/VI) layers of the ACC. Both AMPA and kainate receptors contribute to synaptic transmission. Repetitive stimulation of the projection also evoked postsynaptic Ca2+ influx in contralateral ACC pyramidal neurons. Behaviorally, light activation of the ACC-ACC connection facilitated behavioral withdrawal responses to mechanical stimuli and noxious heat. In an animal model of neuropathic pain, light inhibitory of ACC-ACC connection reduces both primary and secondary hyperalgesia. Our findings provide strong direct evidence for the excitatory or facilitatory contribution of ACC-ACC connection to pain perception, and this mechanism may provide therapeutic targets for future treatment of chronic pain and related emotional disorders.
Collapse
Affiliation(s)
- Xu-Hui Li
- Center for Neuron and Disease, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, China
- Department of Physiology, Faculty of Medicine, University of Toronto, Medical Science Building, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
- Institute of Brain Research, Qingdao International Academician Park, Qingdao, Shandong, 266000, China
| | - Wantong Shi
- Center for Neuron and Disease, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, China
- Institute of Brain Research, Qingdao International Academician Park, Qingdao, Shandong, 266000, China
| | - Qi-Yu Chen
- Institute of Brain Research, Qingdao International Academician Park, Qingdao, Shandong, 266000, China
- CAS Key Laboratory of Brain Connectome and Manipulation, Interdisciplinary Center for Brain Information, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science Shenzhen Fundamental Research Institutions, Shenzhen, Guangdong, 518055, China
| | - Shun Hao
- Institute of Brain Research, Qingdao International Academician Park, Qingdao, Shandong, 266000, China
| | - Hui-Hui Miao
- Department of Physiology, Faculty of Medicine, University of Toronto, Medical Science Building, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
- Department of Anesthesiology, Beijing Shijitan Hospital, Capital Medical University, 10th Tieyi Road, Haidian District, Beijing, 100038, China
| | - Zhuang Miao
- Department of Physiology, Faculty of Medicine, University of Toronto, Medical Science Building, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
| | - Fang Xu
- CAS Key Laboratory of Brain Connectome and Manipulation, Interdisciplinary Center for Brain Information, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science Shenzhen Fundamental Research Institutions, Shenzhen, Guangdong, 518055, China
| | - Guo-Qiang Bi
- CAS Key Laboratory of Brain Connectome and Manipulation, Interdisciplinary Center for Brain Information, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science Shenzhen Fundamental Research Institutions, Shenzhen, Guangdong, 518055, China
| | - Min Zhuo
- Center for Neuron and Disease, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, China.
- Department of Physiology, Faculty of Medicine, University of Toronto, Medical Science Building, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada.
- Institute of Brain Research, Qingdao International Academician Park, Qingdao, Shandong, 266000, China.
- Department of Neurology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510130, China.
| |
Collapse
|
25
|
Baker MR, Lee AS, Rajadhyaksha AM. L-type calcium channels and neuropsychiatric diseases: Insights into genetic risk variant-associated genomic regulation and impact on brain development. Channels (Austin) 2023; 17:2176984. [PMID: 36803254 PMCID: PMC9980663 DOI: 10.1080/19336950.2023.2176984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 02/01/2023] [Indexed: 02/21/2023] Open
Abstract
Recent human genetic studies have linked a variety of genetic variants in the CACNA1C and CACNA1D genes to neuropsychiatric and neurodevelopmental disorders. This is not surprising given the work from multiple laboratories using cell and animal models that have established that Cav1.2 and Cav1.3 L-type calcium channels (LTCCs), encoded by CACNA1C and CACNA1D, respectively, play a key role in various neuronal processes that are essential for normal brain development, connectivity, and experience-dependent plasticity. Of the multiple genetic aberrations reported, genome-wide association studies (GWASs) have identified multiple single nucleotide polymorphisms (SNPs) in CACNA1C and CACNA1D that are present within introns, in accordance with the growing body of literature establishing that large numbers of SNPs associated with complex diseases, including neuropsychiatric disorders, are present within non-coding regions. How these intronic SNPs affect gene expression has remained a question. Here, we review recent studies that are beginning to shed light on how neuropsychiatric-linked non-coding genetic variants can impact gene expression via regulation at the genomic and chromatin levels. We additionally review recent studies that are uncovering how altered calcium signaling through LTCCs impact some of the neuronal developmental processes, such as neurogenesis, neuron migration, and neuron differentiation. Together, the described changes in genomic regulation and disruptions in neurodevelopment provide possible mechanisms by which genetic variants of LTCC genes contribute to neuropsychiatric and neurodevelopmental disorders.
Collapse
Affiliation(s)
- Madelyn R. Baker
- Neuroscience Program, Weill Cornell Graduate School of Medical Sciences, New York, USA
- Department of Pharmacology, Weill Cornell Medicine, New York, USA
| | - Andrew S. Lee
- Neuroscience Program, Weill Cornell Graduate School of Medical Sciences, New York, USA
- Developmental Biology Program, Sloan Kettering Institute, New York, USA
| | - Anjali M. Rajadhyaksha
- Neuroscience Program, Weill Cornell Graduate School of Medical Sciences, New York, USA
- Pediatric Neurology, Department of Pediatrics, Weill Cornell Medicine, New York, USA
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, USA
- Weill Cornell Autism Research Program, Weill Cornell Medicine, New York, USA
| |
Collapse
|
26
|
Gusain P, Taketoshi M, Tominaga Y, Tominaga T. Functional Dissection of Ipsilateral and Contralateral Neural Activity Propagation Using Voltage-Sensitive Dye Imaging in Mouse Prefrontal Cortex. eNeuro 2023; 10:ENEURO.0161-23.2023. [PMID: 37977827 DOI: 10.1523/eneuro.0161-23.2023] [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: 05/15/2023] [Revised: 11/03/2023] [Accepted: 11/10/2023] [Indexed: 11/19/2023] Open
Abstract
Prefrontal cortex (PFC) intrahemispheric activity and the interhemispheric connection have a significant impact on neuropsychiatric disorder pathology. This study aimed to generate a functional map of FC intrahemispheric and interhemispheric connections. Functional dissection of mouse PFCs was performed using the voltage-sensitive dye (VSD) imaging method with high speed (1 ms/frame), high resolution (256 × 256 pixels), and a large field of view (∼10 mm). Acute serial 350 μm slices were prepared from the bregma covering the PFC and numbered 1-5 based on their distance from the bregma (i.e., 1.70, 1.34, 0.98, 0.62, and 0.26 mm) with reference to the Mouse Brain Atlas (Paxinos and Franklin, 2008). The neural response to electrical stimulation was measured at nine sites and then averaged, and a functional map of the propagation patterns was created. Intracortical propagation was observed in slices 3-5, encompassing the anterior cingulate cortex (ACC) and corpus callosum (CC). The activity reached area 33 of the ACC. Direct white matter stimulation activated area 33 in both hemispheres. Similar findings were obtained via DiI staining of the CC. Imaging analysis revealed directional biases in neural signals traveling within the ACC, whereby the signal transmission speed and probability varied based on the signal direction. Specifically, the spread of neural signals from cg2 to cg1 was stronger than that from cingulate cortex area 1(cg1) to cingulate cortex area 2(cg2), which has implications for interhemispheric functional connections. These findings highlight the importance of understanding the PFC functional anatomy in evaluating neuromodulators like serotonin and dopamine, as well as other factors related to neuropsychiatric diseases.
Collapse
Affiliation(s)
- Pooja Gusain
- Institute of Neuroscience, Tokushima Bunri University, Sanuki 769-2193, Japan
| | - Makiko Taketoshi
- Institute of Neuroscience, Tokushima Bunri University, Sanuki 769-2193, Japan
| | - Yoko Tominaga
- Institute of Neuroscience, Tokushima Bunri University, Sanuki 769-2193, Japan
| | - Takashi Tominaga
- Institute of Neuroscience, Tokushima Bunri University, Sanuki 769-2193, Japan
- Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Sanuki 769-2193, Japan
| |
Collapse
|
27
|
Gonzalez-Ferrer J, Lehrer J, O’Farrell A, Paten B, Teodorescu M, Haussler D, Jonsson VD, Mostajo-Radji MA. Unraveling Neuronal Identities Using SIMS: A Deep Learning Label Transfer Tool for Single-Cell RNA Sequencing Analysis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.28.529615. [PMID: 36909548 PMCID: PMC10002667 DOI: 10.1101/2023.02.28.529615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
Abstract
Large single-cell RNA datasets have contributed to unprecedented biological insight. Often, these take the form of cell atlases and serve as a reference for automating cell labeling of newly sequenced samples. Yet, classification algorithms have lacked the capacity to accurately annotate cells, particularly in complex datasets. Here we present SIMS (Scalable, Interpretable Machine Learning for Single-Cell), an end-to-end data-efficient machine learning pipeline for discrete classification of single-cell data that can be applied to new datasets with minimal coding. We benchmarked SIMS against common single-cell label transfer tools and demonstrated that it performs as well or better than state of the art algorithms. We then use SIMS to classify cells in one of the most complex tissues: the brain. We show that SIMS classifies cells of the adult cerebral cortex and hippocampus at a remarkably high accuracy. This accuracy is maintained in trans-sample label transfers of the adult human cerebral cortex. We then apply SIMS to classify cells in the developing brain and demonstrate a high level of accuracy at predicting neuronal subtypes, even in periods of fate refinement, shedding light on genetic changes affecting specific cell types across development. Finally, we apply SIMS to single cell datasets of cortical organoids to predict cell identities and unveil genetic variations between cell lines. SIMS identifies cell-line differences and misannotated cell lineages in human cortical organoids derived from different pluripotent stem cell lines. When cell types are obscured by stress signals, label transfer from primary tissue improves the accuracy of cortical organoid annotations, serving as a reliable ground truth. Altogether, we show that SIMS is a versatile and robust tool for cell-type classification from single-cell datasets.
Collapse
Affiliation(s)
- Jesus Gonzalez-Ferrer
- These authors contributed equally to this work
- Genomics Institute, University of California Santa Cruz, Santa Cruz, 95060, CA, USA
- Live Cell Biotechnology Discovery Lab, University of California Santa Cruz, Santa Cruz, 95060, CA, USA
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, 95060, CA, USA
| | - Julian Lehrer
- These authors contributed equally to this work
- Genomics Institute, University of California Santa Cruz, Santa Cruz, 95060, CA, USA
- Live Cell Biotechnology Discovery Lab, University of California Santa Cruz, Santa Cruz, 95060, CA, USA
- Department of Applied Mathematics, University of California Santa Cruz, Santa Cruz, 95060, CA, USA
| | - Ash O’Farrell
- Genomics Institute, University of California Santa Cruz, Santa Cruz, 95060, CA, USA
| | - Benedict Paten
- Genomics Institute, University of California Santa Cruz, Santa Cruz, 95060, CA, USA
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, 95060, CA, USA
| | - Mircea Teodorescu
- Genomics Institute, University of California Santa Cruz, Santa Cruz, 95060, CA, USA
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, 95060, CA, USA
- Department of Electrical and Computer Engineering, University of California Santa Cruz, Santa Cruz, 95060, CA, USA
| | - David Haussler
- Genomics Institute, University of California Santa Cruz, Santa Cruz, 95060, CA, USA
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, 95060, CA, USA
| | - Vanessa D. Jonsson
- Department of Applied Mathematics, University of California Santa Cruz, Santa Cruz, 95060, CA, USA
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, 95060, CA, USA
- Co-senior authors
| | - Mohammed A. Mostajo-Radji
- Genomics Institute, University of California Santa Cruz, Santa Cruz, 95060, CA, USA
- Live Cell Biotechnology Discovery Lab, University of California Santa Cruz, Santa Cruz, 95060, CA, USA
- Co-senior authors
| |
Collapse
|
28
|
Molotkov D, Ferrarese L, Boissonnet T, Asari H. Topographic axonal projection at single-cell precision supports local retinotopy in the mouse superior colliculus. Nat Commun 2023; 14:7418. [PMID: 37973798 PMCID: PMC10654506 DOI: 10.1038/s41467-023-43218-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 11/03/2023] [Indexed: 11/19/2023] Open
Abstract
Retinotopy, like all long-range projections, can arise from the axons themselves or their targets. The underlying connectivity pattern, however, remains elusive at the fine scale in the mammalian brain. To address this question, we functionally mapped the spatial organization of the input axons and target neurons in the female mouse retinocollicular pathway at single-cell resolution using in vivo two-photon calcium imaging. We found a near-perfect retinotopic tiling of retinal ganglion cell axon terminals, with an average error below 30 μm or 2° of visual angle. The precision of retinotopy was relatively lower for local neurons in the superior colliculus. Subsequent data-driven modeling ascribed it to a low input convergence, on average 5.5 retinal ganglion cell inputs per postsynaptic cell in the superior colliculus. These results indicate that retinotopy arises largely from topographically precise input from presynaptic cells, rather than elaborating local circuitry to reconstruct the topography by postsynaptic cells.
Collapse
Affiliation(s)
- Dmitry Molotkov
- Epigenetics and Neurobiology Unit, EMBL Rome, European Molecular Biology Laboratory, Monterotondo, 00015, Italy
| | - Leiron Ferrarese
- Epigenetics and Neurobiology Unit, EMBL Rome, European Molecular Biology Laboratory, Monterotondo, 00015, Italy
| | - Tom Boissonnet
- Epigenetics and Neurobiology Unit, EMBL Rome, European Molecular Biology Laboratory, Monterotondo, 00015, Italy
- Collaboration for joint PhD degree between EMBL and Université Grenoble Alpes, Grenoble Institut des Neurosciences, La Tronche, 38700, France
- Center for Advanced Imaging, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, 40225, Germany
| | - Hiroki Asari
- Epigenetics and Neurobiology Unit, EMBL Rome, European Molecular Biology Laboratory, Monterotondo, 00015, Italy.
| |
Collapse
|
29
|
Lynton Z, Suárez R, Fenlon LR. Brain plasticity following corpus callosum agenesis or loss: a review of the Probst bundles. Front Neuroanat 2023; 17:1296779. [PMID: 38020213 PMCID: PMC10657877 DOI: 10.3389/fnana.2023.1296779] [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: 09/24/2023] [Accepted: 10/11/2023] [Indexed: 12/01/2023] Open
Abstract
The corpus callosum is the largest axonal tract in the human brain, connecting the left and right cortical hemipheres. This structure is affected in myriad human neurodevelopmental disorders, and can be entirely absent as a result of congenital or surgical causes. The age when callosal loss occurs, for example via surgical section in cases of refractory epilepsy, correlates with resulting brain morphology and neuropsychological outcomes, whereby an earlier loss generally produces relatively improved interhemispheric connectivity compared to a loss in adulthood (known as the "Sperry's paradox"). However, the mechanisms behind these age-dependent differences remain unclear. Perhaps the best documented and most striking of the plastic changes that occur due to developmental, but not adult, callosal loss is the formation of large, bilateral, longitudinal ectopic tracts termed Probst bundles. Despite over 100 years of research into these ectopic tracts, which are the largest and best described stereotypical ectopic brain tracts in humans, much remains unclear about them. Here, we review the anatomy of the Probst bundles, along with evidence for their faciliatory or detrimental function, the required conditions for their formation, patterns of etiology, and mechanisms of development. We provide hypotheses for many of the remaining mysteries of the Probst bundles, including their possible relationship to preserved interhemispheric communication following corpus callosum absence. Future research into naturally occurring plastic tracts such as Probst bundles will help to inform the general rules governing axon plasticity and disorders of brain miswiring.
Collapse
Affiliation(s)
- Zorana Lynton
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, QLD, Australia
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia
| | - Rodrigo Suárez
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, QLD, Australia
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia
| | - Laura R. Fenlon
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, QLD, Australia
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia
| |
Collapse
|
30
|
Shiura H, Kitazawa M, Ishino F, Kaneko-Ishino T. Roles of retrovirus-derived PEG10 and PEG11/RTL1 in mammalian development and evolution and their involvement in human disease. Front Cell Dev Biol 2023; 11:1273638. [PMID: 37842090 PMCID: PMC10570562 DOI: 10.3389/fcell.2023.1273638] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 09/14/2023] [Indexed: 10/17/2023] Open
Abstract
PEG10 and PEG11/RTL1 are paternally expressed, imprinted genes that play essential roles in the current eutherian developmental system and are therefore associated with developmental abnormalities caused by aberrant genomic imprinting. They are also presumed to be retrovirus-derived genes with homology to the sushi-ichi retrotransposon GAG and POL, further expanding our comprehension of mammalian evolution via the domestication (exaptation) of retrovirus-derived acquired genes. In this manuscript, we review the importance of PEG10 and PEG11/RTL1 in genomic imprinting research via their functional roles in development and human disease, including neurodevelopmental disorders of genomic imprinting, Angelman, Kagami-Ogata and Temple syndromes, and the impact of newly inserted DNA on the emergence of newly imprinted regions. We also discuss their possible roles as ancestors of other retrovirus-derived RTL/SIRH genes that likewise play important roles in the current mammalian developmental system, such as in the placenta, brain and innate immune system.
Collapse
Affiliation(s)
- Hirosuke Shiura
- Faculty of Life and Environmental Sciences, University of Yamanashi, Yamanashi, Japan
| | - Moe Kitazawa
- School of BioSciences, Faculty of Science, The University of Melbourne, Melbourne, VIC, Australia
| | - Fumitoshi Ishino
- Institute of Research, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Tomoko Kaneko-Ishino
- Faculty of Nursing, School of Medicine, Tokai University, Isehara, Kanagawa, Japan
| |
Collapse
|
31
|
Young TR, Yamamoto M, Kikuchi SS, Yoshida AC, Abe T, Inoue K, Johansen JP, Benucci A, Yoshimura Y, Shimogori T. Thalamocortical control of cell-type specificity drives circuits for processing whisker-related information in mouse barrel cortex. Nat Commun 2023; 14:6077. [PMID: 37770450 PMCID: PMC10539368 DOI: 10.1038/s41467-023-41749-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 09/15/2023] [Indexed: 09/30/2023] Open
Abstract
Excitatory spiny stellate neurons are prominently featured in the cortical circuits of sensory modalities that provide high salience and high acuity representations of the environment. These specialized neurons are considered developmentally linked to bottom-up inputs from the thalamus, however, the molecular mechanisms underlying their diversification and function are unknown. Here, we investigated this in mouse somatosensory cortex, where spiny stellate neurons and pyramidal neurons have distinct roles in processing whisker-evoked signals. Utilizing spatial transcriptomics, we identified reciprocal patterns of gene expression which correlated with these cell-types and were linked to innervation by specific thalamic inputs during development. Genetic manipulation that prevents the acquisition of spiny stellate fate highlighted an important role for these neurons in processing distinct whisker signals within functional cortical columns, and as a key driver in the formation of specific whisker-related circuits in the cortex.
Collapse
Affiliation(s)
- Timothy R Young
- Laboratory for Molecular Mechanisms of Brain Development, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
| | - Mariko Yamamoto
- Division of Visual Information Processing, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, 444-8585, Japan
| | - Satomi S Kikuchi
- Laboratory for Molecular Mechanisms of Brain Development, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
| | - Aya C Yoshida
- Laboratory for Molecular Mechanisms of Brain Development, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
| | - Takaya Abe
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 6500047, Japan
| | - Kenichi Inoue
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 6500047, Japan
| | - Joshua P Johansen
- Laboratory for Neural Circuitry of Learning and Memory, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
| | - Andrea Benucci
- Laboratory for Neural Circuits and Behavior, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
- School of Biological and Behavioural Sciences, Queen Mary University of London, London, E1 4NS, UK
| | - Yumiko Yoshimura
- Division of Visual Information Processing, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, 444-8585, Japan
| | - Tomomi Shimogori
- Laboratory for Molecular Mechanisms of Brain Development, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan.
| |
Collapse
|
32
|
Froberg JE, Durak O, Macklis JD. Development of nanoRibo-seq enables study of regulated translation by cortical neuron subtypes, showing uORF translation in synaptic-axonal genes. Cell Rep 2023; 42:112995. [PMID: 37624698 PMCID: PMC10591829 DOI: 10.1016/j.celrep.2023.112995] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 05/26/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023] Open
Abstract
Investigation of translation in rare cell types or subcellular contexts is challenging due to large input requirements for standard approaches. Here, we present "nanoRibo-seq" an optimized approach using 102- to 103-fold less input material than bulk approaches. nanoRibo-seq exhibits rigorous quality control features consistent with quantification of ribosome protected fragments with as few as 1,000 cells. We compare translatomes of two closely related cortical neuron subtypes, callosal projection neurons (CPN) and subcerebral projection neurons (SCPN), during their early postnatal development. We find that, while translational efficiency is highly correlated between CPN and SCPN, several dozen mRNAs are differentially translated. We further examine upstream open reading frame (uORF) translation and identify that mRNAs involved in synapse organization and axon development are highly enriched for uORF translation in both subtypes. nanoRibo-seq enables investigation of translational regulation of rare cell types in vivo and offers a flexible approach for globally quantifying translation from limited input material.
Collapse
Affiliation(s)
- John E Froberg
- Department of Stem Cell and Regenerative Biology, and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Omer Durak
- Department of Stem Cell and Regenerative Biology, and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Jeffrey D Macklis
- Department of Stem Cell and Regenerative Biology, and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
33
|
Altounian M, Bellon A, Mann F. Neuronal miR-17-5p contributes to interhemispheric cortical connectivity defects induced by prenatal alcohol exposure. Cell Rep 2023; 42:113020. [PMID: 37610874 DOI: 10.1016/j.celrep.2023.113020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 06/30/2023] [Accepted: 08/08/2023] [Indexed: 08/25/2023] Open
Abstract
Structural and functional deficits in brain connectivity are reported in patients with fetal alcohol spectrum disorders (FASDs), but whether and how prenatal alcohol exposure (PAE) affects axonal development of neurons and disrupts wiring between brain regions is unknown. Here, we develop a mouse model of moderate alcohol exposure during prenatal brain wiring to study the effects of PAE on corpus callosum (CC) development. PAE induces aberrant navigation of interhemispheric CC axons that persists even after exposure ends, leading to ectopic termination in the contralateral cortex. The neuronal miR-17-5p and its target ephrin type A receptor 4 (EphA4) mediate the effect of alcohol on the contralateral targeting of CC axons. Thus, altered microRNA-mediated regulation of axonal guidance may have implications for interhemispheric cortical connectivity and associated behaviors in FASD.
Collapse
Affiliation(s)
| | - Anaïs Bellon
- Aix Marseille University, INSERM, INMED, Marseille, France
| | - Fanny Mann
- Aix Marseille University, CNRS, IBDM, Marseille, France.
| |
Collapse
|
34
|
Kaneko-Ishino T, Ishino F. Retrovirus-Derived RTL/SIRH: Their Diverse Roles in the Current Eutherian Developmental System and Contribution to Eutherian Evolution. Biomolecules 2023; 13:1436. [PMID: 37892118 PMCID: PMC10604271 DOI: 10.3390/biom13101436] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/18/2023] [Accepted: 09/19/2023] [Indexed: 10/29/2023] Open
Abstract
Eutherians have 11 retrotransposon Gag-like (RTL)/sushi-ichi retrotransposon homolog (SIRH) genes presumably derived from a certain retrovirus. Accumulating evidence indicates that the RTL/SIRH genes play a variety of roles in the current mammalian developmental system, such as in the placenta, brain, and innate immune system, in a eutherian-specific manner. It has been shown that the functional role of Paternally Expressed 10 (PEG10) in placental formation is unique to the therian mammals, as are the eutherian-specific roles of PEG10 and PEG11/RTL1 in maintaining the fetal capillary network and the endocrine regulation of RTL7/SIRH7 (aka Leucine Zipper Down-Regulated in Cancer 1 (LDOCK1)) in the placenta. In the brain, PEG11/RTL1 is expressed in the corticospinal tract and hippocampal commissure, mammalian-specific structures, and in the corpus callosum, a eutherian-specific structure. Unexpectedly, at least three RTL/SIRH genes, RTL5/SIRH8, RTL6/SIRH3, and RTL9/SIRH10, play important roles in combating a variety of pathogens, namely viruses, bacteria, and fungi, respectively, suggesting that the innate immunity system of the brain in eutherians has been enhanced by the emergence of these new components. In this review, we will summarize the function of 10 out of the 11 RTL/SIRH genes and discuss their roles in eutherian development and evolution.
Collapse
Affiliation(s)
- Tomoko Kaneko-Ishino
- Faculty of Nursing, School of Medicine, Tokai University, Kanagawa 259-1193, Japan
| | - Fumitoshi Ishino
- Center for Experimental Animals, Institute of Research, Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan
| |
Collapse
|
35
|
Rabeling A, Goolam M. Cerebral organoids as an in vitro model to study autism spectrum disorders. Gene Ther 2023; 30:659-669. [PMID: 35790793 DOI: 10.1038/s41434-022-00356-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 06/01/2022] [Accepted: 06/23/2022] [Indexed: 11/09/2022]
Abstract
Autism spectrum disorders (ASDs) are a set of disorders characterised by social and communication deficits caused by numerous genetic lesions affecting brain development. Progress in ASD research has been hampered by the lack of appropriate models, as both 2D cell culture as well as animal models cannot fully recapitulate the developing human brain or the pathogenesis of ASD. Recently, cerebral organoids have been developed to provide a more accurate, 3D in vitro model of human brain development. Cerebral organoids have been shown to recapitulate the foetal brain gene expression profile, transcriptome, epigenome, as well as disease dynamics of both idiopathic and syndromic ASDs. They are thus an excellent tool to investigate development of foetal stage ASDs, as well as interventions that can reverse or rescue the altered phenotypes observed. In this review, we discuss the development of cerebral organoids, their recent applications in the study of both syndromic and idiopathic ASDs, their use as an ASD drug development platform, as well as limitations of their use in ASD research.
Collapse
Affiliation(s)
- Alexa Rabeling
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, 7925, South Africa
| | - Mubeen Goolam
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, 7925, South Africa.
- UCT Neuroscience Institute, Cape Town, South Africa.
| |
Collapse
|
36
|
Hong W, Gong P, Pan X, Liu Y, Qi G, Qi C, Qin S. Krüppel-like factor 7 deficiency disrupts corpus callosum development and neuronal migration in the developing mouse cerebral cortex. Brain Pathol 2023; 33:e13186. [PMID: 37401095 PMCID: PMC10467035 DOI: 10.1111/bpa.13186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 06/16/2023] [Indexed: 07/05/2023] Open
Abstract
Krüppel-like Factor 7 (KLF7) is a zinc finger transcription factor that has a critical role in cellular differentiation, tumorigenesis, and regeneration. Mutations in Klf7 are associated with autism spectrum disorder, which is characterized by neurodevelopmental delay and intellectual disability. Here we show that KLF7 regulates neurogenesis and neuronal migration during mouse cortical development. Conditional depletion of KLF7 in neural progenitor cells resulted in agenesis of the corpus callosum, defects in neurogenesis, and impaired neuronal migration in the neocortex. Transcriptomic profiling analysis indicated that KLF7 regulates a cohort of genes involved in neuronal differentiation and migration, including p21 and Rac3. These findings provide insights into our understanding of the potential mechanisms underlying neurological defects associated with Klf7 mutations.
Collapse
Affiliation(s)
- Wentong Hong
- Department of Anatomy, Histology and Embryology, School of Basic Medical SciencesFudan UniversityShanghaiChina
| | - Pifang Gong
- Department of Anatomy, Histology and Embryology, School of Basic Medical SciencesFudan UniversityShanghaiChina
| | - Xinjie Pan
- Department of Anatomy, Histology and Embryology, School of Basic Medical SciencesFudan UniversityShanghaiChina
| | - Yitong Liu
- Department of Anatomy, Histology and Embryology, School of Basic Medical SciencesFudan UniversityShanghaiChina
| | - Guibo Qi
- Department of Anatomy, Histology and Embryology, School of Basic Medical SciencesFudan UniversityShanghaiChina
| | - Congcong Qi
- Department of Laboratory Animal ScienceFudan UniversityShanghaiChina
| | - Song Qin
- Department of Anatomy, Histology and Embryology, School of Basic Medical SciencesFudan UniversityShanghaiChina
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghaiChina
| |
Collapse
|
37
|
Wang Y, Chen Z, Ma G, Wang L, Liu Y, Qin M, Fei X, Wu Y, Xu M, Zhang S. A frontal transcallosal inhibition loop mediates interhemispheric balance in visuospatial processing. Nat Commun 2023; 14:5213. [PMID: 37626171 PMCID: PMC10457336 DOI: 10.1038/s41467-023-40985-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
Interhemispheric communication through the corpus callosum is required for both sensory and cognitive processes. Impaired transcallosal inhibition causing interhemispheric imbalance is believed to underlie visuospatial bias after frontoparietal cortical damage, but the synaptic circuits involved remain largely unknown. Here, we show that lesions in the mouse anterior cingulate area (ACA) cause severe visuospatial bias mediated by a transcallosal inhibition loop. In a visual-change-detection task, ACA callosal-projection neurons (CPNs) were more active with contralateral visual field changes than with ipsilateral changes. Unilateral CPN inactivation impaired contralateral change detection but improved ipsilateral detection by altering interhemispheric interaction through callosal projections. CPNs strongly activated contralateral parvalbumin-positive (PV+) neurons, and callosal-input-driven PV+ neurons preferentially inhibited ipsilateral CPNs, thus mediating transcallosal inhibition. Unilateral PV+ neuron activation caused a similar behavioral bias to contralateral CPN activation and ipsilateral CPN inactivation, and bilateral PV+ neuron activation eliminated this bias. Notably, restoring interhemispheric balance by activating contralesional PV+ neurons significantly improved contralesional detection in ACA-lesioned animals. Thus, a frontal transcallosal inhibition loop comprising CPNs and callosal-input-driven PV+ neurons mediates interhemispheric balance in visuospatial processing, and enhancing contralesional transcallosal inhibition restores interhemispheric balance while also reversing lesion-induced bias.
Collapse
Affiliation(s)
- Yanjie Wang
- Songjiang Research Institute, Shanghai Songjiang District Central Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200025, China
- Center for Brain Science of Shanghai Children's Medical Center, Department of Anatomy and Physiology, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Zhaonan Chen
- Songjiang Research Institute, Shanghai Songjiang District Central Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200025, China
- Center for Brain Science of Shanghai Children's Medical Center, Department of Anatomy and Physiology, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Guofen Ma
- Songjiang Research Institute, Shanghai Songjiang District Central Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200025, China
- Center for Brain Science of Shanghai Children's Medical Center, Department of Anatomy and Physiology, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Lizhao Wang
- Songjiang Research Institute, Shanghai Songjiang District Central Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200025, China
- Center for Brain Science of Shanghai Children's Medical Center, Department of Anatomy and Physiology, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yanmei Liu
- Songjiang Research Institute, Shanghai Songjiang District Central Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200025, China
- Center for Brain Science of Shanghai Children's Medical Center, Department of Anatomy and Physiology, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Meiling Qin
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xiang Fei
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yifan Wu
- Songjiang Research Institute, Shanghai Songjiang District Central Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200025, China
- Center for Brain Science of Shanghai Children's Medical Center, Department of Anatomy and Physiology, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Min Xu
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Siyu Zhang
- Songjiang Research Institute, Shanghai Songjiang District Central Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200025, China.
- Center for Brain Science of Shanghai Children's Medical Center, Department of Anatomy and Physiology, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| |
Collapse
|
38
|
Huang-Hobbs E, Cheng YT, Ko Y, Luna-Figueroa E, Lozzi B, Taylor KR, McDonald M, He P, Chen HC, Yang Y, Maleki E, Lee ZF, Murali S, Williamson MR, Choi D, Curry R, Bayley J, Woo J, Jalali A, Monje M, Noebels JL, Harmanci AS, Rao G, Deneen B. Remote neuronal activity drives glioma progression through SEMA4F. Nature 2023; 619:844-850. [PMID: 37380778 PMCID: PMC10840127 DOI: 10.1038/s41586-023-06267-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 05/26/2023] [Indexed: 06/30/2023]
Abstract
The tumour microenvironment plays an essential role in malignancy, and neurons have emerged as a key component of the tumour microenvironment that promotes tumourigenesis across a host of cancers1,2. Recent studies on glioblastoma (GBM) highlight bidirectional signalling between tumours and neurons that propagates a vicious cycle of proliferation, synaptic integration and brain hyperactivity3-8; however, the identity of neuronal subtypes and tumour subpopulations driving this phenomenon is incompletely understood. Here we show that callosal projection neurons located in the hemisphere contralateral to primary GBM tumours promote progression and widespread infiltration. Using this platform to examine GBM infiltration, we identified an activity-dependent infiltrating population present at the leading edge of mouse and human tumours that is enriched for axon guidance genes. High-throughput, in vivo screening of these genes identified SEMA4F as a key regulator of tumourigenesis and activity-dependent progression. Furthermore, SEMA4F promotes the activity-dependent infiltrating population and propagates bidirectional signalling with neurons by remodelling tumour-adjacent synapses towards brain network hyperactivity. Collectively our studies demonstrate that subsets of neurons in locations remote to primary GBM promote malignant progression, and also show new mechanisms of glioma progression that are regulated by neuronal activity.
Collapse
Affiliation(s)
- Emmet Huang-Hobbs
- The Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Yi-Ting Cheng
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX, USA
| | - Yeunjung Ko
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Program in Immunology and Microbiology, Baylor College of Medicine, Houston, TX, USA
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX, USA
| | - Estefania Luna-Figueroa
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Brittney Lozzi
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX, USA
- Program in Genetics and Genomics, Baylor College of Medicine, Houston, TX, USA
| | - Kathryn R Taylor
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Malcolm McDonald
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Program in Development, Disease, Models and Therapeutics, Baylor College of Medicine, Houston, TX, USA
| | - Peihao He
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Program in Cancer Cell Biology, Baylor College of Medicine, Houston, TX, USA
| | - Hsiao-Chi Chen
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Program in Cancer Cell Biology, Baylor College of Medicine, Houston, TX, USA
| | - Yuhui Yang
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX, USA
| | - Ehson Maleki
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Zhung-Fu Lee
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Program in Development, Disease, Models and Therapeutics, Baylor College of Medicine, Houston, TX, USA
| | - Sanjana Murali
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Program in Cancer Cell Biology, Baylor College of Medicine, Houston, TX, USA
| | - Michael R Williamson
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Dongjoo Choi
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Rachel Curry
- The Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
| | - James Bayley
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX, USA
| | - Junsung Woo
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Ali Jalali
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX, USA
| | - Michelle Monje
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
- Department of Neurosurgery, Stanford University, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Jeffrey L Noebels
- Department of Human and Molecular Genetics, Baylor College of Medicine, Houston, TX, USA
- Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| | - Akdes Serin Harmanci
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX, USA
| | - Ganesh Rao
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX, USA
| | - Benjamin Deneen
- The Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, TX, USA.
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA.
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX, USA.
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX, USA.
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX, USA.
- Program in Development, Disease, Models and Therapeutics, Baylor College of Medicine, Houston, TX, USA.
- Program in Cancer Cell Biology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
39
|
Choi S, Chen Y, Zeng H, Biswal B, Yu X. Identifying the distinct spectral dynamics of laminar-specific interhemispheric connectivity with bilateral line-scanning fMRI. J Cereb Blood Flow Metab 2023; 43:1115-1129. [PMID: 36803280 PMCID: PMC10291453 DOI: 10.1177/0271678x231158434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 11/30/2022] [Accepted: 12/12/2022] [Indexed: 02/23/2023]
Abstract
Despite extensive efforts to identify interhemispheric functional connectivity (FC) with resting-state (rs-) fMRI, correlated low-frequency rs-fMRI signal fluctuation across homotopic cortices originates from multiple sources. It remains challenging to differentiate circuit-specific FC from global regulation. Here, we developed a bilateral line-scanning fMRI method to detect laminar-specific rs-fMRI signals from homologous forepaw somatosensory cortices with high spatial and temporal resolution in rat brains. Based on spectral coherence analysis, two distinct bilateral fluctuation spectral features were identified: ultra-slow fluctuation (<0.04 Hz) across all cortical laminae versus Layer (L) 2/3-specific evoked BOLD at 0.05 Hz based on 4 s on/16 s off block design and resting-state fluctuations at 0.08-0.1 Hz. Based on the measurements of evoked BOLD signal at corpus callosum (CC), this L2/3-specific 0.05 Hz signal is likely associated with neuronal circuit-specific activity driven by the callosal projection, which dampened ultra-slow oscillation less than 0.04 Hz. Also, the rs-fMRI power variability clustering analysis showed that the appearance of L2/3-specific 0.08-0.1 Hz signal fluctuation is independent of the ultra-slow oscillation across different trials. Thus, distinct laminar-specific bilateral FC patterns at different frequency ranges can be identified by the bilateral line-scanning fMRI method.
Collapse
Affiliation(s)
- Sangcheon Choi
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, USA
- Max Planck Institute for Biological Cybernetics, Tübingen, Germany
| | - Yi Chen
- Max Planck Institute for Biological Cybernetics, Tübingen, Germany
| | - Hang Zeng
- Max Planck Institute for Biological Cybernetics, Tübingen, Germany
- Graduate Training Centre of Neuroscience, University of Tübingen, Tübingen, Germany
| | - Bharat Biswal
- Department of Biomedical Engineering, NJIT, Newark, NJ, USA
| | - Xin Yu
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, USA
| |
Collapse
|
40
|
Yang S, Niou ZX, Enriquez A, LaMar J, Huang JY, Ling K, Jafar-Nejad P, Gilley J, Coleman MP, Tennessen JM, Rangaraju V, Lu HC. NMNAT2 supports vesicular glycolysis via NAD homeostasis to fuel fast axonal transport. RESEARCH SQUARE 2023:rs.3.rs-2859584. [PMID: 37292715 PMCID: PMC10246254 DOI: 10.21203/rs.3.rs-2859584/v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Background Bioenergetic maladaptations and axonopathy are often found in the early stages of neurodegeneration. Nicotinamide adenine dinucleotide (NAD), an essential cofactor for energy metabolism, is mainly synthesized by Nicotinamide mononucleotide adenylyl transferase 2 (NMNAT2) in CNS neurons. NMNAT2 mRNA levels are reduced in the brains of Alzheimer's, Parkinson's, and Huntington's disease. Here we addressed whether NMNAT2 is required for axonal health of cortical glutamatergic neurons, whose long-projecting axons are often vulnerable in neurodegenerative conditions. We also tested if NMNAT2 maintains axonal health by ensuring axonal ATP levels for axonal transport, critical for axonal function. Methods We generated mouse and cultured neuron models to determine the impact of NMNAT2 loss from cortical glutamatergic neurons on axonal transport, energetic metabolism, and morphological integrity. In addition, we determined if exogenous NAD supplementation or inhibiting a NAD hydrolase, sterile alpha and TIR motif-containing protein 1 (SARM1), prevented axonal deficits caused by NMNAT2 loss. This study used a combination of genetics, molecular biology, immunohistochemistry, biochemistry, fluorescent time-lapse imaging, live imaging with optical sensors, and anti-sense oligos. Results We provide in vivo evidence that NMNAT2 in glutamatergic neurons is required for axonal survival. Using in vivo and in vitro studies, we demonstrate that NMNAT2 maintains the NAD-redox potential to provide "on-board" ATP via glycolysis to vesicular cargos in distal axons. Exogenous NAD+ supplementation to NMNAT2 KO neurons restores glycolysis and resumes fast axonal transport. Finally, we demonstrate both in vitro and in vivo that reducing the activity of SARM1, an NAD degradation enzyme, can reduce axonal transport deficits and suppress axon degeneration in NMNAT2 KO neurons. Conclusion NMNAT2 ensures axonal health by maintaining NAD redox potential in distal axons to ensure efficient vesicular glycolysis required for fast axonal transport.
Collapse
|
41
|
Abstract
Mapping neuronal circuits that generate focal to bilateral tonic-clonic seizures is essential for understanding general principles of seizure propagation and modifying the risk of death and injury due to bilateral motor seizures. We used novel techniques developed over the past decade to study these circuits. We propose the general hypothesis that at the mesoscale, seizures follow anatomical projections of the seizure focus, preferentially activating more excitable neurons.
Collapse
Affiliation(s)
| | - Jaideep Kapur
- Department of Neurology, University of Virginia, Charlottesville, VA, USA
- UVA Brain Institute, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
42
|
Fukuzaki Y, Faustino J, Lecuyer M, Rayasam A, Vexler ZS. Global sphingosine-1-phosphate receptor 2 deficiency attenuates neuroinflammation and ischemic-reperfusion injury after neonatal stroke. iScience 2023; 26:106340. [PMID: 37009213 PMCID: PMC10064246 DOI: 10.1016/j.isci.2023.106340] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 10/31/2022] [Accepted: 03/01/2023] [Indexed: 03/07/2023] Open
Abstract
Arterial ischemic stroke is common in neonates-1 per 2,300-5,000 births-and therapeutic targets remain insufficiently defined. Sphingosine-1-phosphate receptor 2 (S1PR2), a major regulator of the CNS and immune systems, is injurious in adult stroke. Here, we assessed whether S1PR2 contributes to stroke induced by 3 h transient middle cerebral artery occlusion (tMCAO) in S1PR2 heterozygous (HET), knockout (KO), and wild type (WT) postnatal day 9 pups. HET and WT of both sexes displayed functional deficits in Open Field test whereas injured KO at 24 h reperfusion performed similarly to naives. S1PR2 deficiency protected neurons, attenuated infiltration of inflammatory monocytes, and altered vessel-microglia interactions without reducing increased cytokine levels in injured regions at 72 h. Pharmacologic inhibition of S1PR2 after tMCAO by JTE-013 attenuated injury 72 h after tMCAO. Importantly, the lack of S1PR2 alleviated anxiety and brain atrophy during chronic injury. Altogether, we identify S1PR2 as a potential new target for mitigating neonatal stroke.
Collapse
Affiliation(s)
- Yumi Fukuzaki
- Department of Neurology, University California San Francisco, Weill Institute for Neurosciences, San Francisco, CA 94158-0663, USA
| | - Joel Faustino
- Department of Neurology, University California San Francisco, Weill Institute for Neurosciences, San Francisco, CA 94158-0663, USA
| | - Matthieu Lecuyer
- Department of Neurology, University California San Francisco, Weill Institute for Neurosciences, San Francisco, CA 94158-0663, USA
| | - Aditya Rayasam
- Department of Neurology, University California San Francisco, Weill Institute for Neurosciences, San Francisco, CA 94158-0663, USA
| | - Zinaida S. Vexler
- Department of Neurology, University California San Francisco, Weill Institute for Neurosciences, San Francisco, CA 94158-0663, USA
| |
Collapse
|
43
|
Szczupak D, Iack PM, Rayêe D, Liu C, Lent R, Tovar-Moll F, Silva AC. The relevance of heterotopic callosal fibers to interhemispheric connectivity of the mammalian brain. Cereb Cortex 2023; 33:4752-4760. [PMID: 36178137 PMCID: PMC10110439 DOI: 10.1093/cercor/bhac377] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 11/14/2022] Open
Abstract
The corpus callosum (CC) is the largest white matter structure and the primary pathway for interhemispheric brain communication. Investigating callosal connectivity is crucial to unraveling the brain's anatomical and functional organization in health and disease. Classical anatomical studies have characterized the bulk of callosal axonal fibers as connecting primarily homotopic cortical areas. Whenever detected, heterotopic callosal fibers were ascribed to altered sprouting and pruning mechanisms in neurodevelopmental diseases such as CC dysgenesis (CCD). We hypothesized that these heterotopic connections had been grossly underestimated due to their complex nature and methodological limitations. We used the Allen Mouse Brain Connectivity Atlas and high-resolution diffusion-weighted imaging to identify and quantify homotopic and heterotopic callosal connections in mice, marmosets, and humans. In all 3 species, we show that ~75% of interhemispheric callosal connections are heterotopic and comprise the central core of the CC, whereas the homotopic fibers lay along its periphery. We also demonstrate that heterotopic connections have an essential role in determining the global properties of brain networks. These findings reshape our view of the corpus callosum's role as the primary hub for interhemispheric brain communication, directly impacting multiple neuroscience fields investigating cortical connectivity, neurodevelopment, and neurodevelopmental disorders.
Collapse
Affiliation(s)
- Diego Szczupak
- Department of Neurobiology, University of Pittsburgh Brain Institute, University of Pittsburgh, Pittsburgh, PA 15261, United States
| | - Pamela Meneses Iack
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-590, Brazil
| | - Danielle Rayêe
- Institute of Ophtalmology and Visual Sciences, Albert Einstein College of Medicine, NY 10461, United States
| | - Cirong Liu
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Roberto Lent
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-590, Brazil
- D’Or Institute Research and Education (IDOR), Rio de Janeiro 22281-100, Brazil
| | - Fernanda Tovar-Moll
- D’Or Institute Research and Education (IDOR), Rio de Janeiro 22281-100, Brazil
| | - Afonso C Silva
- Department of Neurobiology, University of Pittsburgh Brain Institute, University of Pittsburgh, Pittsburgh, PA 15261, United States
| |
Collapse
|
44
|
Huang-Hobbs E, Cheng YT, Ko Y, Luna-Figueroa E, Lozzi B, Taylor KR, McDonald M, He P, Chen HC, Yang Y, Maleki E, Lee ZF, Murali S, Williamson M, Choi D, Curry R, Bayley J, Woo J, Jalali A, Monje M, Noebels JL, Harmanci AS, Rao G, Deneen B. Remote neuronal activity drives glioma infiltration via Sema4f. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.15.532832. [PMID: 36993539 PMCID: PMC10055154 DOI: 10.1101/2023.03.15.532832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
The tumor microenvironment (TME) plays an essential role in malignancy and neurons have emerged as a key component of the TME that promotes tumorigenesis across a host of cancers. Recent studies on glioblastoma (GBM) highlight bi-directional signaling between tumors and neurons that propagates a vicious cycle of proliferation, synaptic integration, and brain hyperactivity; however, the identity of neuronal subtypes and tumor subpopulations driving this phenomenon are incompletely understood. Here we show that callosal projection neurons located in the hemisphere contralateral to primary GBM tumors promote progression and widespread infiltration. Using this platform to examine GBM infiltration, we identified an activity dependent infiltrating population present at the leading edge of mouse and human tumors that is enriched for axon guidance genes. High-throughput, in vivo screening of these genes identified Sema4F as a key regulator of tumorigenesis and activity-dependent infiltration. Furthermore, Sema4F promotes the activity-dependent infiltrating population and propagates bi-directional signaling with neurons by remodeling tumor adjacent synapses towards brain network hyperactivity. Collectively, our studies demonstrate that subsets of neurons in locations remote to primary GBM promote malignant progression, while revealing new mechanisms of tumor infiltration that are regulated by neuronal activity.
Collapse
Affiliation(s)
- Emmet Huang-Hobbs
- The Integrative Molecular and Biomedical Sciences Graduate Program (IMBS), Baylor College of Medicine, Houston TX 77030
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston TX 77030
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030
| | - Yi-Ting Cheng
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston TX 77030
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Program in Developmental Biology, Baylor College of Medicine, Houston TX 77030
| | - Yeunjung Ko
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston TX 77030
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Program in Immunology and Microbiology, Baylor College of Medicine, Houston, TX 77030
- Department of Neurosurgery, Baylor College of Medicine, Houston TX 77030
| | - Estefania Luna-Figueroa
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston TX 77030
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030
| | - Brittney Lozzi
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston TX 77030
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Department of Neurosurgery, Baylor College of Medicine, Houston TX 77030
- Program in Genetics and Genomics, Baylor College of Medicine, Houston TX 77030
| | - Kathryn R Taylor
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Malcolm McDonald
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston TX 77030
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Program in Development, Disease, Models and Therapeutics, Baylor College of Medicine, Houston TX 77030
| | - Peihao He
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston TX 77030
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Program in Cancer Cell Biology, Baylor College of Medicine, Houston TX 77030
| | - Hsiao-Chi Chen
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston TX 77030
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Program in Cancer Cell Biology, Baylor College of Medicine, Houston TX 77030
| | - Yuhui Yang
- Department of Neurosurgery, Baylor College of Medicine, Houston TX 77030
| | - Ehson Maleki
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston TX 77030
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030
| | - Zhung-Fu Lee
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston TX 77030
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Program in Development, Disease, Models and Therapeutics, Baylor College of Medicine, Houston TX 77030
| | - Sanjana Murali
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston TX 77030
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Program in Cancer Cell Biology, Baylor College of Medicine, Houston TX 77030
| | - Michael Williamson
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston TX 77030
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030
| | - Dongjoo Choi
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston TX 77030
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030
| | - Rachel Curry
- The Integrative Molecular and Biomedical Sciences Graduate Program (IMBS), Baylor College of Medicine, Houston TX 77030
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston TX 77030
| | - James Bayley
- Department of Neurosurgery, Baylor College of Medicine, Houston TX 77030
| | - Junsung Woo
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston TX 77030
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030
| | - Ali Jalali
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Department of Neurosurgery, Baylor College of Medicine, Houston TX 77030
| | - Michelle Monje
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
- Department of Neurosurgery, Stanford University, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Jeffrey L Noebels
- Department of Human and Molecular Genetics, Baylor College of Medicine, Houston, TX, 77030
- Department of Neurology, Baylor College of Medicine, Houston, TX 77030
| | - Akdes Serin Harmanci
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Department of Neurosurgery, Baylor College of Medicine, Houston TX 77030
| | - Ganesh Rao
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Department of Neurosurgery, Baylor College of Medicine, Houston TX 77030
| | - Benjamin Deneen
- The Integrative Molecular and Biomedical Sciences Graduate Program (IMBS), Baylor College of Medicine, Houston TX 77030
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston TX 77030
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Program in Developmental Biology, Baylor College of Medicine, Houston TX 77030
- Department of Neurosurgery, Baylor College of Medicine, Houston TX 77030
- Program in Development, Disease, Models and Therapeutics, Baylor College of Medicine, Houston TX 77030
- Program in Cancer Cell Biology, Baylor College of Medicine, Houston TX 77030
| |
Collapse
|
45
|
Plachez C, Tsytsarev V, Zhao S, Erzurumlu RS. Amyloid Deposition and Dendritic Complexity of Corticocortical Projection Cells in Five Familial Alzheimer's Disease Mouse. Neuroscience 2023; 512:85-98. [PMID: 36549605 PMCID: PMC10112867 DOI: 10.1016/j.neuroscience.2022.12.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
In Alzheimer's disease and related dementias, amyloid beta (Aβ) and amyloid plaques can disrupt long-term synaptic plasticity, learning and memory and cognitive function. Plaque accumulation can disrupt corticocortical circuitry leading to abnormalities in sensory, motor, and cognitive processing. In this study, using 5xFAD (five Familial Alzheimer's Disease - FAD - mutations) mice, we evaluated amyloid plaque formation in different cortical areas, and whether differential amyloid accumulation across cortical fields correlates with changes in dendritic complexity of layer 3 corticocortical projection neurons and functional responses in the primary somatosensory cortex following whisker stimulation. We focused on three cortical areas: the primary somatosensory cortex (S1), the primary motor cortex (M1), and the prefrontal cortex (PFC including the anterior cingulate, prelimbic, and infralimbic subdivisions). We found that Aβ and amyloid plaque accumulation is not uniform across 5xFAD cortical areas, while there is no expression in littermate controls. We also found that there are differential layer 3 pyramidal cell dendritic complexity changes across the three areas in 5xFAD mice, compared to same age controls, with no apparent relation to differential amyloid accumulation. We used voltage-sensitive dye imaging (VSDi) to visualize neural activity in S1, M1 and PFC following whisker activation. Control mice show normal physiological responses in all three cortical areas, whereas 5xFAD mice only display physiological responses in S1. Taken together our results show that 5xFAD mutation affects the overall dendritic morphology of layer 3 pyramidal cells across sensory-motor and association cortex irrespective of the density and distribution of the Aβ amyloid proteins. Corticocortical circuitry between the sensory and motor/association areas is most likely disrupted in 5xFAD mice as cortical responses to whisker stimulation are altered.
Collapse
Affiliation(s)
- Celine Plachez
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, 20 Penn St, HSF-2, Baltimore, 21201 MD, USA.
| | - Vassiliy Tsytsarev
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, 20 Penn St, HSF-2, Baltimore, 21201 MD, USA.
| | - Shuxin Zhao
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, 20 Penn St, HSF-2, Baltimore, 21201 MD, USA.
| | - Reha S Erzurumlu
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, 20 Penn St, HSF-2, Baltimore, 21201 MD, USA.
| |
Collapse
|
46
|
Babij R, Ferrer C, Donatelle A, Wacks S, Buch AM, Niemeyer JE, Ma H, Duan ZRS, Fetcho RN, Che A, Otsuka T, Schwartz TH, Huang BS, Liston C, De Marco García NV. Gabrb3 is required for the functional integration of pyramidal neuron subtypes in the somatosensory cortex. Neuron 2023; 111:256-274.e10. [PMID: 36446382 PMCID: PMC9852093 DOI: 10.1016/j.neuron.2022.10.037] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 08/30/2022] [Accepted: 10/27/2022] [Indexed: 11/29/2022]
Abstract
Dysfunction of gamma-aminobutyric acid (GABA)ergic circuits is strongly associated with neurodevelopmental disorders. However, it is unclear how genetic predispositions impact circuit assembly. Using in vivo two-photon and widefield calcium imaging in developing mice, we show that Gabrb3, a gene strongly associated with autism spectrum disorder (ASD) and Angelman syndrome (AS), is enriched in contralaterally projecting pyramidal neurons and is required for inhibitory function. We report that Gabrb3 ablation leads to a developmental decrease in GABAergic synapses, increased local network synchrony, and long-lasting enhancement in functional connectivity of contralateral-but not ipsilateral-pyramidal neuron subtypes. In addition, Gabrb3 deletion leads to increased cortical response to tactile stimulation at neonatal stages. Using human transcriptomics and neuroimaging datasets from ASD subjects, we show that the spatial distribution of GABRB3 expression correlates with atypical connectivity in these subjects. Our studies reveal a requirement for Gabrb3 during the emergence of interhemispheric circuits for sensory processing.
Collapse
Affiliation(s)
- Rachel Babij
- Center for Neurogenetics, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA.,Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY 10021, USA
| | - Camilo Ferrer
- Center for Neurogenetics, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Alexander Donatelle
- Center for Neurogenetics, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Sam Wacks
- Center for Neurogenetics, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Amanda M Buch
- Center for Neurogenetics, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - James E Niemeyer
- Department of Neurological Surgery, Weill Cornell Medicine, New-York Presbyterian Hospital, New York, NY 10021, USA
| | - Hongtao Ma
- Department of Neurological Surgery, Weill Cornell Medicine, New-York Presbyterian Hospital, New York, NY 10021, USA
| | - Zhe Ran S Duan
- Center for Neurogenetics, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA.,Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY 10021, USA
| | - Robert N Fetcho
- Center for Neurogenetics, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA.,Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY 10021, USA
| | - Alicia Che
- Center for Neurogenetics, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA.,Current affiliation: Department of Psychiatry, Yale School of Medicine, New Haven, CT 06519, USA
| | - Takumi Otsuka
- Center for Neurogenetics, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Theodore H Schwartz
- Department of Neurological Surgery, Weill Cornell Medicine, New-York Presbyterian Hospital, New York, NY 10021, USA
| | - Ben S Huang
- Center for Neurogenetics, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Conor Liston
- Center for Neurogenetics, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Natalia V De Marco García
- Center for Neurogenetics, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA.,Lead Contact,Correspondence to
| |
Collapse
|
47
|
Fischer J, Fernández Ortuño E, Marsoner F, Artioli A, Peters J, Namba T, Eugster Oegema C, Huttner WB, Ladewig J, Heide M. Human-specific ARHGAP11B ensures human-like basal progenitor levels in hominid cerebral organoids. EMBO Rep 2022; 23:e54728. [PMID: 36098218 PMCID: PMC9646322 DOI: 10.15252/embr.202254728] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 08/18/2022] [Accepted: 08/23/2022] [Indexed: 02/06/2023] Open
Abstract
The human-specific gene ARHGAP11B has been implicated in human neocortex expansion. However, the extent of ARHGAP11B's contribution to this expansion during hominid evolution is unknown. Here we address this issue by genetic manipulation of ARHGAP11B levels and function in chimpanzee and human cerebral organoids. ARHGAP11B expression in chimpanzee cerebral organoids doubles basal progenitor levels, the class of cortical progenitors with a key role in neocortex expansion. Conversely, interference with ARHGAP11B's function in human cerebral organoids decreases basal progenitors down to the chimpanzee level. Moreover, ARHGAP11A or ARHGAP11B rescue experiments in ARHGAP11A plus ARHGAP11B double-knockout human forebrain organoids indicate that lack of ARHGAP11B, but not of ARHGAP11A, decreases the abundance of basal radial glia-the basal progenitor type thought to be of particular relevance for neocortex expansion. Taken together, our findings demonstrate that ARHGAP11B is necessary and sufficient to ensure the elevated basal progenitor levels that characterize the fetal human neocortex, suggesting that this human-specific gene was a major contributor to neocortex expansion during human evolution.
Collapse
Affiliation(s)
- Jan Fischer
- Max Planck Institute of Molecular Cell Biology and GeneticsPfotenhauerstrasse 108DresdenGermany
- Present address:
Institute for Clinical GeneticsUniversity Hospital Carl Gustav CarusDresdenGermany
| | | | - Fabio Marsoner
- Central Institute of Mental HealthUniversity of Heidelberg/Medical Faculty MannheimMannheimGermany
- Hector Institute for Translational Brain Research (HITBR gGmbH)MannheimGermany
- German Cancer Research Center (DKFZ)HeidelbergGermany
| | - Annasara Artioli
- Central Institute of Mental HealthUniversity of Heidelberg/Medical Faculty MannheimMannheimGermany
- Hector Institute for Translational Brain Research (HITBR gGmbH)MannheimGermany
- German Cancer Research Center (DKFZ)HeidelbergGermany
| | - Jula Peters
- Max Planck Institute of Molecular Cell Biology and GeneticsPfotenhauerstrasse 108DresdenGermany
| | - Takashi Namba
- Max Planck Institute of Molecular Cell Biology and GeneticsPfotenhauerstrasse 108DresdenGermany
- Present address:
Neuroscience Center, HiLIFE ‐ Helsinki Institute of Life ScienceUniversity of HelsinkiHelsinkiFinland
| | | | - Wieland B. Huttner
- Max Planck Institute of Molecular Cell Biology and GeneticsPfotenhauerstrasse 108DresdenGermany
| | - Julia Ladewig
- Central Institute of Mental HealthUniversity of Heidelberg/Medical Faculty MannheimMannheimGermany
- Hector Institute for Translational Brain Research (HITBR gGmbH)MannheimGermany
- German Cancer Research Center (DKFZ)HeidelbergGermany
| | - Michael Heide
- Max Planck Institute of Molecular Cell Biology and GeneticsPfotenhauerstrasse 108DresdenGermany
- German Primate CenterLeibniz Institute for Primate ResearchGöttingenGermany
| |
Collapse
|
48
|
Chaudhary S, Roy A, Summers C, Ahles T, Li CSR, Chao HH. Effects of androgen deprivation on white matter integrity and processing speed in prostate cancer patients. Am J Cancer Res 2022; 12:4802-4814. [PMID: 36381311 PMCID: PMC9641391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 09/21/2022] [Indexed: 02/09/2023] Open
Abstract
Studies have associated chemotherapy-elicited changes in cognitive function with impaired white matter integrity in cancer patients. Androgen deprivation therapy (ADT) may lead to cognitive deficits in prostate cancer patients; however, whether ADT influences white matter integrity has never been investigated. In a prospective study, 15 men with non-metastatic prostate cancer receiving ADT and 15 not receiving ADT (controls or CON), comparable in age and years of education, participated in N-back task, flankers' task, and quality-of-life (QoL) assessments. All participants underwent diffusion tensor imaging of the brain at baseline and at 6 months. Imaging data were processed with published routines. The results of a paired t-test of 6-month follow-up vs. baseline were evaluated at a corrected threshold for the whole brain each in ADT and CON. ADT patients showed significantly worse 1-back accuracy during follow-up, but the two groups did not differ in 2-back accuracy, 1- or 2-back reaction time (RT), flankers' task RT or QoL across time points. In ADT, significantly reduced fractional anisotropy (FA) was noted in the corpus callosum, forceps minor/anterior thalamic radiation, superior and posterior corona radiata. The differences in FA correlated significantly with changes in 2-back and flankers' task RT. No significant FA changes were noted during follow-up in CON. Six-month ADT affects white matter integrity, and the deficits were associated with slower processing speed. These findings add to the literature supporting the deleterious effects of androgen deprivation on the brain and cognition in prostate cancer patients.
Collapse
Affiliation(s)
- Shefali Chaudhary
- Department of Psychiatry, Yale University School of MedicineNew Haven, CT, USA
| | - Alicia Roy
- VA Connecticut Healthcare SystemWest Haven, CT, USA
| | | | - Tim Ahles
- Department of Psychiatry and Behavioral Sciences, Memorial Sloan Kettering Cancer CenterNew York, NY, USA
| | - Chiang-Shan R Li
- Department of Psychiatry, Yale University School of MedicineNew Haven, CT, USA,Department of Neuroscience, Yale University School of MedicineNew Haven, CT, USA,Interdepartmental Neuroscience Program, Yale University School of MedicineNew Haven, CT, USA,Wu Tsai Institute, Yale UniversityNew Haven, CT, USA
| | - Herta H Chao
- VA Connecticut Healthcare SystemWest Haven, CT, USA,Department of Medicine & Yale Comprehensive Cancer Center, Yale University School of MedicineNew Haven, CT, USA
| |
Collapse
|
49
|
Wagner NR, Sinha A, Siththanandan V, Kowalchuk AM, MacDonald JL, Tharin S. miR-409-3p represses Cited2 to refine neocortical layer V projection neuron identity. Front Neurosci 2022; 16:931333. [PMID: 36248641 PMCID: PMC9558290 DOI: 10.3389/fnins.2022.931333] [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: 04/28/2022] [Accepted: 09/13/2022] [Indexed: 12/14/2022] Open
Abstract
The evolutionary emergence of the corticospinal tract and corpus callosum are thought to underpin the expansion of complex motor and cognitive abilities in mammals. Molecular mechanisms regulating development of the neurons whose axons comprise these tracts, the corticospinal and callosal projection neurons, remain incompletely understood. Our previous work identified a genomic cluster of microRNAs (miRNAs), Mirg/12qF1, that is unique to placental mammals and specifically expressed by corticospinal neurons, and excluded from callosal projection neurons, during development. We found that one of these, miR-409-3p, can convert layer V callosal into corticospinal projection neurons, acting in part through repression of the transcriptional regulator Lmo4. Here we show that miR-409-3p also directly represses the transcriptional co-regulator Cited2, which is highly expressed by callosal projection neurons from the earliest stages of neurogenesis. Cited2 is highly expressed by intermediate progenitor cells (IPCs) in the embryonic neocortex while Mirg, which encodes miR-409-3p, is excluded from these progenitors. miR-409-3p gain-of-function (GOF) in IPCs results in a phenocopy of established Cited2 loss-of-function (LOF). At later developmental stages, both miR-409-3p GOF and Cited2 LOF promote the expression of corticospinal at the expense of callosal projection neuron markers in layer V. Taken together, this work identifies previously undescribed roles for miR-409-3p in controlling IPC numbers and for Cited2 in controlling callosal fate. Thus, miR-409-3p, possibly in cooperation with other Mirg/12qF1 miRNAs, represses Cited2 as part of the multifaceted regulation of the refinement of neuronal cell fate within layer V, combining molecular regulation at multiple levels in both progenitors and post-mitotic neurons.
Collapse
Affiliation(s)
- Nikolaus R. Wagner
- Department of Biology, Program in Neuroscience, Syracuse University, Syracuse, NY, United States
| | - Ashis Sinha
- Department of Biology, Program in Neuroscience, Syracuse University, Syracuse, NY, United States
| | - Verl Siththanandan
- Department of Neurosurgery, Stanford University Medical Center, Center for Academic Medicine, Palo Alto, CA, United States
| | - Angelica M. Kowalchuk
- Department of Biology, Program in Neuroscience, Syracuse University, Syracuse, NY, United States
| | - Jessica L. MacDonald
- Department of Biology, Program in Neuroscience, Syracuse University, Syracuse, NY, United States,*Correspondence: Jessica L. MacDonald,
| | - Suzanne Tharin
- Department of Neurosurgery, Stanford University Medical Center, Center for Academic Medicine, Palo Alto, CA, United States,Division of Neurosurgery, Palo Alto Veterans Affairs Health Care System, Palo Alto, CA, United States,Suzanne Tharin,
| |
Collapse
|
50
|
Fischer J, Fernández Ortuño E, Marsoner F, Artioli A, Peters J, Namba T, Eugster Oegema C, Huttner WB, Ladewig J, Heide M. Human-specific ARHGAP11B ensures human-like basal progenitor levels in hominid cerebral organoids. EMBO Rep 2022; 23:e54728. [PMID: 36381990 PMCID: PMC9646322 DOI: 10.1101/2020.10.01.322792] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 08/18/2022] [Accepted: 08/23/2022] [Indexed: 06/16/2023] Open
Abstract
The human-specific gene ARHGAP11B has been implicated in human neocortex expansion. However, the extent of ARHGAP11B's contribution to this expansion during hominid evolution is unknown. Here we address this issue by genetic manipulation of ARHGAP11B levels and function in chimpanzee and human cerebral organoids. ARHGAP11B expression in chimpanzee cerebral organoids doubles basal progenitor levels, the class of cortical progenitors with a key role in neocortex expansion. Conversely, interference with ARHGAP11B's function in human cerebral organoids decreases basal progenitors down to the chimpanzee level. Moreover, ARHGAP11A or ARHGAP11B rescue experiments in ARHGAP11A plus ARHGAP11B double-knockout human forebrain organoids indicate that lack of ARHGAP11B, but not of ARHGAP11A, decreases the abundance of basal radial glia - the basal progenitor type thought to be of particular relevance for neocortex expansion. Taken together, our findings demonstrate that ARHGAP11B is necessary and sufficient to ensure the elevated basal progenitor levels that characterize the fetal human neocortex, suggesting that this human-specific gene was a major contributor to neocortex expansion during human evolution.
Collapse
Affiliation(s)
- Jan Fischer
- Max Planck Institute of Molecular Cell Biology and GeneticsPfotenhauerstrasse 108DresdenGermany
- Present address:
Institute for Clinical GeneticsUniversity Hospital Carl Gustav CarusDresdenGermany
| | | | - Fabio Marsoner
- Central Institute of Mental HealthUniversity of Heidelberg/Medical Faculty MannheimMannheimGermany
- Hector Institute for Translational Brain Research (HITBR gGmbH)MannheimGermany
- German Cancer Research Center (DKFZ)HeidelbergGermany
| | - Annasara Artioli
- Central Institute of Mental HealthUniversity of Heidelberg/Medical Faculty MannheimMannheimGermany
- Hector Institute for Translational Brain Research (HITBR gGmbH)MannheimGermany
- German Cancer Research Center (DKFZ)HeidelbergGermany
| | - Jula Peters
- Max Planck Institute of Molecular Cell Biology and GeneticsPfotenhauerstrasse 108DresdenGermany
| | - Takashi Namba
- Max Planck Institute of Molecular Cell Biology and GeneticsPfotenhauerstrasse 108DresdenGermany
- Present address:
Neuroscience Center, HiLIFE ‐ Helsinki Institute of Life ScienceUniversity of HelsinkiHelsinkiFinland
| | | | - Wieland B. Huttner
- Max Planck Institute of Molecular Cell Biology and GeneticsPfotenhauerstrasse 108DresdenGermany
| | - Julia Ladewig
- Central Institute of Mental HealthUniversity of Heidelberg/Medical Faculty MannheimMannheimGermany
- Hector Institute for Translational Brain Research (HITBR gGmbH)MannheimGermany
- German Cancer Research Center (DKFZ)HeidelbergGermany
| | - Michael Heide
- Max Planck Institute of Molecular Cell Biology and GeneticsPfotenhauerstrasse 108DresdenGermany
- German Primate CenterLeibniz Institute for Primate ResearchGöttingenGermany
| |
Collapse
|