1
|
Tendilla-Beltrán H, Perez-Osornio DL, Apam-Castillejos DJ, Flores G. Atypical antipsychotics improve dendritic spine pathology in temporal lobe cortex neurons in a developmental rodent model of schizophrenia. Behav Brain Res 2025; 478:115341. [PMID: 39549876 DOI: 10.1016/j.bbr.2024.115341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 11/04/2024] [Accepted: 11/13/2024] [Indexed: 11/18/2024]
Abstract
"Dendritic spine pathology" refers to alterations in density and morphology of dendritic spines, crucial in corticolimbic neurons in schizophrenia. These structural neuroplasticity changes contribute to the disease's neurobiological underpinnings, alongside alterations in other brain regions, such as temporal lobe cortices like the auditory cortex (Au1) and the entorhinal cortex (Ent), involved in sensory processing, memory, and learning. The neonatal ventral hippocampus lesion (NVHL) in rats exhibits behavioral abnormalities akin to schizophrenia symptoms and corticolimbic dendritic spine pathology, mitigated by atypical antipsychotic drugs (AADs) like risperidone (RISP) and olanzapine (OLZ). This study investigated NVHL-induced dendritic spine pathology in Au1 and Ent, evaluating RISP and OLZ effects. NVHL induced dendritic spine pathology mainly by reducing the dendritic spine density in Au1 and Ent neurons; both RISP and OLZ mitigated it, increasing dendritic spine density and mushroom spine population, the ones related with synaptic strengthening, while decreasing stubby spine population. These findings underscore the role of impaired neuroplasticity in the temporal lobe cortices in schizophrenia pathophysiology and highlight the relevance of the NVHL model for studying neuroplasticity mechanisms in the disease. They also contribute to the growing understanding of targeting structural and functional neuroplasticity for novel drugs in the pharmacotherapy of the disease.
Collapse
Affiliation(s)
- Hiram Tendilla-Beltrán
- Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico
| | | | | | - Gonzalo Flores
- Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico.
| |
Collapse
|
2
|
Tumdam R, Hussein Y, Garin-Shkolnik T, Stern S. NMDA Receptors in Neurodevelopmental Disorders: Pathophysiology and Disease Models. Int J Mol Sci 2024; 25:12366. [PMID: 39596430 PMCID: PMC11594297 DOI: 10.3390/ijms252212366] [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: 10/07/2024] [Revised: 11/08/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
N-methyl-D-aspartate receptors (NMDARs) are critical components of the mammalian central nervous system, involved in synaptic transmission, plasticity, and neurodevelopment. This review focuses on the structural and functional characteristics of NMDARs, with a particular emphasis on the GRIN2 subunits (GluN2A-D). The diversity of GRIN2 subunits, driven by alternative splicing and genetic variants, significantly impacts receptor function, synaptic localization, and disease manifestation. The temporal and spatial expression of these subunits is essential for typical neural development, with each subunit supporting distinct phases of synaptic formation and plasticity. Disruptions in their developmental regulation are linked to neurodevelopmental disorders, underscoring the importance of understanding these dynamics in NDD pathophysiology. We explore the physiological properties and developmental regulation of these subunits, highlighting their roles in the pathophysiology of various NDDs, including ASD, epilepsy, and schizophrenia. By reviewing current knowledge and experimental models, including mouse models and human-induced pluripotent stem cells (hiPSCs), this article aims to elucidate different approaches through which the intricacies of NMDAR dysfunction in NDDs are currently being explored. The comprehensive understanding of NMDAR subunit composition and their mutations provides a foundation for developing targeted therapeutic strategies to address these complex disorders.
Collapse
Affiliation(s)
- Roshan Tumdam
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3103301, Israel
| | - Yara Hussein
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3103301, Israel
| | | | - Shani Stern
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3103301, Israel
| |
Collapse
|
3
|
Tordai C, Hathy E, Gyergyák H, Vincze K, Baradits M, Koller J, Póti Á, Jezsó B, Homolya L, Molnár MJ, Nagy L, Szüts D, Apáti Á, Réthelyi JM. Probing the biological consequences of a previously undescribed de novo mutation of ZMYND11 in a schizophrenia patient by CRISPR genome editing and induced pluripotent stem cell based in vitro disease-modeling. Schizophr Res 2024; 273:107-120. [PMID: 38290943 DOI: 10.1016/j.schres.2024.01.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 02/01/2024]
Abstract
BACKGROUND Schizophrenia (SCZ) is a severe neuropsychiatric disorder of complex, poorly understood etiology, associated with both genetic and environmental factors. De novo mutations (DNMs) represent a new source of genetic variation in SCZ, however, in most cases their biological significance remains unclear. We sought to investigate molecular disease pathways connected to DNMs in SCZ by combining human induced pluripotent stem cell (hiPSC) based disease modeling and CRISPR-based genome editing. METHODS We selected a SCZ case-parent trio with the case individual carrying a potentially disease causing 1495C > T nonsense DNM in the zinc finger MYND domain-containing protein 11 (ZMYND11), a gene implicated in biological processes relevant for SCZ. In the patient-derived hiPSC line the mutation was corrected using CRISPR, while monoallelic or biallelic frameshift mutations were introduced into a control hiPSC line. Isogenic cell lines were differentiated into hippocampal neuronal progenitor cells (NPCs) and functionally active dentate gyrus granule cells (DGGCs). Immunofluorescence microscopy and RNA sequencing were used to test for morphological and transcriptomic differences at NPC and DGCC stages. Functionality of neurons was investigated using calcium-imaging and multi-electrode array measurements. RESULTS Morphology in the mutant hippocampal NPCs and neurons was preserved, however, we detected significant transcriptomic and functional alterations. RNA sequencing showed massive upregulation of neuronal differentiation genes, and downregulation of cell adhesion genes. Decreased reactivity to glutamate was demonstrated by calcium-imaging. CONCLUSIONS Our findings lend support to the involvement of glutamatergic dysregulation in the pathogenesis of SCZ. This approach represents a powerful model system for precision psychiatry and pharmacological research.
Collapse
Affiliation(s)
- Csongor Tordai
- Institute of Molecular Life Sciences, Research Center for Natural Sciences, 1117 Budapest, Magyar tudósok körútja 2, Budapest, Hungary; Molecular Psychiatry Research Group, Semmelweis University, 1083 Budapest, Balassa utca 6, Budapest, Hungary
| | - Edit Hathy
- Molecular Psychiatry Research Group, Semmelweis University, 1083 Budapest, Balassa utca 6, Budapest, Hungary
| | - Hella Gyergyák
- Institute of Molecular Life Sciences, Research Center for Natural Sciences, 1117 Budapest, Magyar tudósok körútja 2, Budapest, Hungary
| | - Katalin Vincze
- Institute of Molecular Life Sciences, Research Center for Natural Sciences, 1117 Budapest, Magyar tudósok körútja 2, Budapest, Hungary; Molecular Psychiatry Research Group, Semmelweis University, 1083 Budapest, Balassa utca 6, Budapest, Hungary
| | - Máté Baradits
- Molecular Psychiatry Research Group, Semmelweis University, 1083 Budapest, Balassa utca 6, Budapest, Hungary; Department of Psychiatry and Psychotherapy, Semmelweis University, 1083 Budapest, Balassa utca 6, Budapest, Hungary
| | - Júlia Koller
- Molecular Psychiatry Research Group, Semmelweis University, 1083 Budapest, Balassa utca 6, Budapest, Hungary; Institute of Genomic Medicine and Rare Disorders, Semmelweis University, 1083 Budapest, Balassa utca 6, Budapest, Hungary
| | - Ádám Póti
- Institute of Molecular Life Sciences, Research Center for Natural Sciences, 1117 Budapest, Magyar tudósok körútja 2, Budapest, Hungary
| | - Bálint Jezsó
- Institute of Molecular Life Sciences, Research Center for Natural Sciences, 1117 Budapest, Magyar tudósok körútja 2, Budapest, Hungary; Doctoral School of Biology and Institute of Biology, Eötvös Loránd University, 1117 Budapest, Pázmány Péter sétány 1/c, Budapest, Hungary
| | - László Homolya
- Institute of Molecular Life Sciences, Research Center for Natural Sciences, 1117 Budapest, Magyar tudósok körútja 2, Budapest, Hungary
| | - Mária Judit Molnár
- Institute of Genomic Medicine and Rare Disorders, Semmelweis University, 1083 Budapest, Balassa utca 6, Budapest, Hungary
| | - László Nagy
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Egyetem tér 1, Debrecen, Hungary
| | - Dávid Szüts
- Institute of Molecular Life Sciences, Research Center for Natural Sciences, 1117 Budapest, Magyar tudósok körútja 2, Budapest, Hungary.
| | - Ágota Apáti
- Institute of Molecular Life Sciences, Research Center for Natural Sciences, 1117 Budapest, Magyar tudósok körútja 2, Budapest, Hungary.
| | - János M Réthelyi
- Molecular Psychiatry Research Group, Semmelweis University, 1083 Budapest, Balassa utca 6, Budapest, Hungary; Department of Psychiatry and Psychotherapy, Semmelweis University, 1083 Budapest, Balassa utca 6, Budapest, Hungary.
| |
Collapse
|
4
|
Stern S, Zhang L, Wang M, Wright R, Rosh I, Hussein Y, Stern T, Choudhary A, Tripathi U, Reed P, Sadis H, Nayak R, Shemen A, Agarwal K, Cordeiro D, Peles D, Hang Y, Mendes APD, Baul TD, Roth JG, Coorapati S, Boks MP, McCombie WR, Hulshoff Pol H, Brennand KJ, Réthelyi JM, Kahn RS, Marchetto MC, Gage FH. Monozygotic twins discordant for schizophrenia differ in maturation and synaptic transmission. Mol Psychiatry 2024; 29:3208-3222. [PMID: 38704507 PMCID: PMC11449799 DOI: 10.1038/s41380-024-02561-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 04/01/2024] [Accepted: 04/12/2024] [Indexed: 05/06/2024]
Abstract
Schizophrenia affects approximately 1% of the world population. Genetics, epigenetics, and environmental factors are known to play a role in this psychiatric disorder. While there is a high concordance in monozygotic twins, about half of twin pairs are discordant for schizophrenia. To address the question of how and when concordance in monozygotic twins occur, we have obtained fibroblasts from two pairs of schizophrenia discordant twins (one sibling with schizophrenia while the second one is unaffected by schizophrenia) and three pairs of healthy twins (both of the siblings are healthy). We have prepared iPSC models for these 3 groups of patients with schizophrenia, unaffected co-twins, and the healthy twins. When the study started the co-twins were considered healthy and unaffected but both the co-twins were later diagnosed with a depressive disorder. The reprogrammed iPSCs were differentiated into hippocampal neurons to measure the neurophysiological abnormalities in the patients. We found that the neurons derived from the schizophrenia patients were less arborized, were hypoexcitable with immature spike features, and exhibited a significant reduction in synaptic activity with dysregulation in synapse-related genes. Interestingly, the neurons derived from the co-twin siblings who did not have schizophrenia formed another distinct group that was different from the neurons in the group of the affected twin siblings but also different from the neurons in the group of the control twins. Importantly, their synaptic activity was not affected. Our measurements that were obtained from schizophrenia patients and their monozygotic twin and compared also to control healthy twins point to hippocampal synaptic deficits as a central mechanism in schizophrenia.
Collapse
Affiliation(s)
- Shani Stern
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel.
| | - Lei Zhang
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Meiyan Wang
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Rebecca Wright
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Idan Rosh
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Yara Hussein
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Tchelet Stern
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Ashwani Choudhary
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Utkarsh Tripathi
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Patrick Reed
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Hagit Sadis
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Ritu Nayak
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Aviram Shemen
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Karishma Agarwal
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Diogo Cordeiro
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - David Peles
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Yuqing Hang
- Razavi Newman Integrative Genomics and Bioinformatics Core, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Ana P D Mendes
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Tithi D Baul
- Department of Psychiatry at the Boston Medical Center, Boston, MA, USA
| | - Julien G Roth
- Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Shashank Coorapati
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Marco P Boks
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, Heidelberglaan 100, 3584CX, Utrecht, The Netherlands
| | | | - Hilleke Hulshoff Pol
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, Heidelberglaan 100, 3584CX, Utrecht, The Netherlands
- Department of Experimental Psychology, Utrecht University, Heidelberglaan 1, 3584CS, Utrecht, The Netherlands
| | - Kristen J Brennand
- Nash Family Department of Neuroscience, Friedman Brain Institute, Pamela Sklar Division of Psychiatric Genomics, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Psychiatry, Department of Genetics, Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT, 06511, USA
| | - János M Réthelyi
- Molecular Psychiatry Research Group and Department of Psychiatry and Psychotherapy, Semmelweis University, Budapest, Hungary
| | - René S Kahn
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research, Education and Clinical Center, James J Peters VA Medical Center, New York, NY, USA
| | - Maria C Marchetto
- Department of Anthropology, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Fred H Gage
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA.
| |
Collapse
|
5
|
Kiltschewskij DJ, Reay WR, Cairns MJ. Schizophrenia is associated with altered DNA methylation variance. Mol Psychiatry 2024:10.1038/s41380-024-02749-5. [PMID: 39271751 DOI: 10.1038/s41380-024-02749-5] [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: 02/19/2024] [Revised: 09/02/2024] [Accepted: 09/04/2024] [Indexed: 09/15/2024]
Abstract
Varying combinations of genetic and environmental risk factors are thought to underpin phenotypic heterogeneity between individuals in psychiatric conditions such as schizophrenia. While epigenome-wide association studies in schizophrenia have identified extensive alteration of mean DNA methylation levels, less is known about the location and impact of DNA methylation variance, which could contribute to phenotypic and treatment response heterogeneity. To explore this question, we conducted the largest meta-analysis of blood DNA methylation variance in schizophrenia to date, leveraging three cohorts comprising 1036 individuals with schizophrenia and 954 non-psychiatric controls. Surprisingly, only a small proportion (0.1%) of the 213 variably methylated positions (VMPs) associated with schizophrenia (Benjamini-Hochberg FDR < 0.05) were shared with differentially methylated positions (DMPs; sites with mean changes between cases and controls). These blood-derived VMPs were found to be overrepresented in genes previously associated with schizophrenia and amongst brain-enriched genes, with evidence of concordant changes at VMPs in the cerebellum, hippocampus, prefrontal cortex, or striatum. Epigenetic covariance was also observed with respect to clinically significant metrics including age of onset, cognitive deficits, and symptom severity. We also uncovered a significant VMP in individuals with first-episode psychosis (n = 644) from additional cohorts and a non-psychiatric comparison group (n = 633). Collectively, these findings suggest schizophrenia is associated with significant changes in DNA methylation variance, which may contribute to individual-to-individual heterogeneity.
Collapse
Affiliation(s)
- Dylan J Kiltschewskij
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia
- Precision Medicine Program, Hunter Medical Research Institute, New Lambton, NSW, Australia
| | - William R Reay
- Menzies Institute for Medical Research, Hobart, TAS, Australia
| | - Murray J Cairns
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia.
- Precision Medicine Program, Hunter Medical Research Institute, New Lambton, NSW, Australia.
| |
Collapse
|
6
|
Birnbaum R, Weinberger DR. The Genesis of Schizophrenia: An Origin Story. Am J Psychiatry 2024; 181:482-492. [PMID: 38822584 DOI: 10.1176/appi.ajp.20240305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/03/2024]
Abstract
Schizophrenia is routinely referred to as a neurodevelopmental disorder, but the role of brain development in a disorder typically diagnosed during early adult life is enigmatic. The authors revisit the neurodevelopmental model of schizophrenia with genomic insights from the most recent schizophrenia clinical genetic association studies, transcriptomic and epigenomic analyses from human postmortem brain studies, and analyses from cellular models that recapitulate neurodevelopment. Emerging insights into schizophrenia genetic risk continue to converge on brain development, particularly stages of early brain development, that may be perturbed to deviate from a typical, normative course, resulting in schizophrenia clinical symptomatology. As the authors explicate, schizophrenia genetic risk is likely dynamic and context dependent, with effects of genetic risk varying spatiotemporally, across the neurodevelopmental continuum. Optimizing therapeutic strategies for the heterogeneous collective of individuals with schizophrenia may likely be guided by leveraging markers of genetic risk and derivative functional insights, well before the emergence of psychosis. Ultimately, rather than a focus on therapeutic intervention during adolescence or adulthood, principles of prediction and prophylaxis in the pre- and perinatal and neonatal stages may best comport with the biology of schizophrenia to address the early-stage perturbations that alter the normative neurodevelopmental trajectory.
Collapse
Affiliation(s)
- Rebecca Birnbaum
- Departments of Psychiatry, Genetics, and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York (Birnbaum); Lieber Institute of Brain Development, Maltz Research Laboratory, and Departments of Psychiatry, Neurology, Neuroscience, and Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore (Weinberger)
| | - Daniel R Weinberger
- Departments of Psychiatry, Genetics, and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York (Birnbaum); Lieber Institute of Brain Development, Maltz Research Laboratory, and Departments of Psychiatry, Neurology, Neuroscience, and Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore (Weinberger)
| |
Collapse
|
7
|
Sawada T, Barbosa AR, Araujo B, McCord AE, D’Ignazio L, Benjamin KJM, Sheehan B, Zabolocki M, Feltrin A, Arora R, Brandtjen AC, Kleinman JE, Hyde TM, Bardy C, Weinberger DR, Paquola ACM, Erwin JA. Recapitulation of Perturbed Striatal Gene Expression Dynamics of Donors' Brains With Ventral Forebrain Organoids Derived From the Same Individuals With Schizophrenia. Am J Psychiatry 2024; 181:493-511. [PMID: 37915216 PMCID: PMC11209846 DOI: 10.1176/appi.ajp.20220723] [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] [Indexed: 11/03/2023]
Abstract
OBJECTIVE Schizophrenia is a brain disorder that originates during neurodevelopment and has complex genetic and environmental etiologies. Despite decades of clinical evidence of altered striatal function in affected patients, studies examining its cellular and molecular mechanisms in humans are limited. To explore neurodevelopmental alterations in the striatum associated with schizophrenia, the authors established a method for the differentiation of induced pluripotent stem cells (iPSCs) into ventral forebrain organoids (VFOs). METHODS VFOs were generated from postmortem dural fibroblast-derived iPSCs of four individuals with schizophrenia and four neurotypical control individuals for whom postmortem caudate genotypes and transcriptomic data were profiled in the BrainSeq neurogenomics consortium. Individuals were selected such that the two groups had nonoverlapping schizophrenia polygenic risk scores (PRSs). RESULTS Single-cell RNA sequencing analyses of VFOs revealed differences in developmental trajectory between schizophrenia and control individuals in which inhibitory neuronal cells from the patients exhibited accelerated maturation. Furthermore, upregulated genes in inhibitory neurons in schizophrenia VFOs showed a significant overlap with upregulated genes in postmortem caudate tissue of individuals with schizophrenia compared with control individuals, including the donors of the iPSC cohort. CONCLUSIONS The findings suggest that striatal neurons derived from high-PRS individuals with schizophrenia carry abnormalities that originated during early brain development and that the VFO model can recapitulate disease-relevant cell type-specific neurodevelopmental phenotypes in a dish.
Collapse
Affiliation(s)
- Tomoyo Sawada
- Lieber Institute for Brain Development, Baltimore, MD, USA
| | | | - Bruno Araujo
- Lieber Institute for Brain Development, Baltimore, MD, USA
| | | | - Laura D’Ignazio
- Lieber Institute for Brain Development, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Kynon J. M. Benjamin
- Lieber Institute for Brain Development, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Bonna Sheehan
- Lieber Institute for Brain Development, Baltimore, MD, USA
| | - Michael Zabolocki
- South Australian Health and Medical Research Institute (SAHMRI), Laboratory for Human Neurophysiology and Genetics, Adelaide, SA, Australia
- Flinders University, Flinders Health and Medical Research Institute (FHMRI), College of Medicine and Public Health, Adelaide, SA, Australia
| | - Arthur Feltrin
- Lieber Institute for Brain Development, Baltimore, MD, USA
| | - Ria Arora
- Lieber Institute for Brain Development, Baltimore, MD, USA
| | | | - Joel E. Kleinman
- Lieber Institute for Brain Development, Baltimore, MD, USA
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Thomas M. Hyde
- Lieber Institute for Brain Development, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Cedric Bardy
- South Australian Health and Medical Research Institute (SAHMRI), Laboratory for Human Neurophysiology and Genetics, Adelaide, SA, Australia
- Flinders University, Flinders Health and Medical Research Institute (FHMRI), College of Medicine and Public Health, Adelaide, SA, Australia
| | - Daniel R. Weinberger
- Lieber Institute for Brain Development, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Apuā C. M. Paquola
- Lieber Institute for Brain Development, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jennifer A. Erwin
- Lieber Institute for Brain Development, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
8
|
Tandon R, Nasrallah H, Akbarian S, Carpenter WT, DeLisi LE, Gaebel W, Green MF, Gur RE, Heckers S, Kane JM, Malaspina D, Meyer-Lindenberg A, Murray R, Owen M, Smoller JW, Yassin W, Keshavan M. The schizophrenia syndrome, circa 2024: What we know and how that informs its nature. Schizophr Res 2024; 264:1-28. [PMID: 38086109 DOI: 10.1016/j.schres.2023.11.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 11/23/2023] [Accepted: 11/29/2023] [Indexed: 03/01/2024]
Abstract
With new data about different aspects of schizophrenia being continually generated, it becomes necessary to periodically revisit exactly what we know. Along with a need to review what we currently know about schizophrenia, there is an equal imperative to evaluate the construct itself. With these objectives, we undertook an iterative, multi-phase process involving fifty international experts in the field, with each step building on learnings from the prior one. This review assembles currently established findings about schizophrenia (construct, etiology, pathophysiology, clinical expression, treatment) and posits what they reveal about its nature. Schizophrenia is a heritable, complex, multi-dimensional syndrome with varying degrees of psychotic, negative, cognitive, mood, and motor manifestations. The illness exhibits a remitting and relapsing course, with varying degrees of recovery among affected individuals with most experiencing significant social and functional impairment. Genetic risk factors likely include thousands of common genetic variants that each have a small impact on an individual's risk and a plethora of rare gene variants that have a larger individual impact on risk. Their biological effects are concentrated in the brain and many of the same variants also increase the risk of other psychiatric disorders such as bipolar disorder, autism, and other neurodevelopmental conditions. Environmental risk factors include but are not limited to urban residence in childhood, migration, older paternal age at birth, cannabis use, childhood trauma, antenatal maternal infection, and perinatal hypoxia. Structural, functional, and neurochemical brain alterations implicate multiple regions and functional circuits. Dopamine D-2 receptor antagonists and partial agonists improve psychotic symptoms and reduce risk of relapse. Certain psychological and psychosocial interventions are beneficial. Early intervention can reduce treatment delay and improve outcomes. Schizophrenia is increasingly considered to be a heterogeneous syndrome and not a singular disease entity. There is no necessary or sufficient etiology, pathology, set of clinical features, or treatment that fully circumscribes this syndrome. A single, common pathophysiological pathway appears unlikely. The boundaries of schizophrenia remain fuzzy, suggesting the absence of a categorical fit and need to reconceptualize it as a broader, multi-dimensional and/or spectrum construct.
Collapse
Affiliation(s)
- Rajiv Tandon
- Department of Psychiatry, WMU Homer Stryker School of Medicine, Kalamazoo, MI 49008, United States of America.
| | - Henry Nasrallah
- Department of Psychiatry, University of Cincinnati College of Medicine Cincinnati, OH 45267, United States of America
| | - Schahram Akbarian
- Department of Psychiatry, Icahn School of Medicine at Mt. Sinai, New York, NY 10029, United States of America
| | - William T Carpenter
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21201, United States of America
| | - Lynn E DeLisi
- Department of Psychiatry, Cambridge Health Alliance and Harvard Medical School, Cambridge, MA 02139, United States of America
| | - Wolfgang Gaebel
- Department of Psychiatry and Psychotherapy, LVR-Klinikum Dusseldorf, Heinrich-Heine University, Dusseldorf, Germany
| | - Michael F Green
- Department of Psychiatry and Biobehavioral Sciences, Jane and Terry Semel Institute of Neuroscience and Human Behavior, UCLA, Los Angeles, CA 90024, United States of America; Greater Los Angeles Veterans' Administration Healthcare System, United States of America
| | - Raquel E Gur
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, United States of America
| | - Stephan Heckers
- Department of Psychiatry, Vanderbilt University Medical Center, Nashville, TN 37232, United States of America
| | - John M Kane
- Department of Psychiatry, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Glen Oaks, NY 11004, United States of America
| | - Dolores Malaspina
- Department of Psychiatry, Neuroscience, Genetics, and Genomics, Icahn School of Medicine at Mt. Sinai, New York, NY 10029, United States of America
| | - Andreas Meyer-Lindenberg
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Mannhein/Heidelberg University, Mannheim, Germany
| | - Robin Murray
- Institute of Psychiatry, Psychology, and Neuroscience, Kings College, London, UK
| | - Michael Owen
- Centre for Neuropsychiatric Genetics and Genomics, and Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Jordan W Smoller
- Center for Precision Psychiatry, Department of Psychiatry, Psychiatric and Neurodevelopmental Unit, Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, United States of America
| | - Walid Yassin
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, United States of America
| | - Matcheri Keshavan
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, United States of America
| |
Collapse
|
9
|
Dean B, Duce J, Li QX, Masters CL, Scarr E. Lower levels of soluble β-amyloid precursor protein, but not β-amyloid, in the frontal cortex in schizophrenia. Psychiatry Res 2024; 331:115656. [PMID: 38071879 DOI: 10.1016/j.psychres.2023.115656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 11/28/2023] [Accepted: 12/01/2023] [Indexed: 01/02/2024]
Abstract
We identified a sub-group (25%) of people with schizophrenia (muscarinic receptor deficit schizophrenia (MRDS)) that are characterised because of markedly lower levels of cortical muscarinic M1 receptors (CHRM1) compared to most people with the disorder (non-MRDS). Notably, bioinformatic analyses of our cortical gene expression data shows a disturbance in the homeostasis of a biochemical pathway that regulates levels of CHRM1. A step in this pathway is the processing of β-amyloid precursor protein (APP) and therefore we postulated there would be altered levels of APP in the frontal cortex from people with MRDS. Here we measure levels of CHRM1 using [3H]pirenzepine binding, soluble APP (sAPP) using Western blotting and amyloid beta peptides (Aβ1-40 and Aβ1-42) using ELISA in the frontal cortex (Brodmann's area 6: BA 6; MRDS = 14, non-MRDS = 14, controls = 14). We confirmed the MRDS cohort in this study had the expected low levels of [3H]pirenzepine binding. In addition, we showed that people with schizophrenia, independent of their sub-group status, had lower levels of sAPP compared to controls but did not have altered levels of Aβ1-40 or Aβ1-42. In conclusion, whilst changes in sAPP are not restricted to MRDS our data could indicate a role of APP, which is important in axonal and synaptic pruning, in the molecular pathology of the syndrome of schizophrenia.
Collapse
Affiliation(s)
- Brian Dean
- The Florey, Parkville, Victoria, Australia; The University of Melbourne of Melbourne Florey Department of Neuroscience and Mental Health, Parkville, Victoria, Australia.
| | - James Duce
- MSD Discovery Centre, 120 Moorgate, London, UK
| | - Qiao-Xin Li
- The Florey, Parkville, Victoria, Australia; The University of Melbourne of Melbourne Florey Department of Neuroscience and Mental Health, Parkville, Victoria, Australia
| | - Colin L Masters
- The Florey, Parkville, Victoria, Australia; The University of Melbourne of Melbourne Florey Department of Neuroscience and Mental Health, Parkville, Victoria, Australia
| | - Elizabeth Scarr
- The Department of Psychiatry, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
10
|
Wang B, Irizar H, Thygesen JH, Zartaloudi E, Austin-Zimmerman I, Bhat A, Harju-Seppänen J, Pain O, Bass N, Gkofa V, Alizadeh BZ, van Amelsvoort T, Arranz MJ, Bender S, Cahn W, Stella Calafato M, Crespo-Facorro B, Di Forti M, Giegling I, de Haan L, Hall J, Hall MH, van Haren N, Iyegbe C, Kahn RS, Kravariti E, Lawrie SM, Lin K, Luykx JJ, Mata I, McDonald C, McIntosh AM, Murray RM, Picchioni M, Powell J, Prata DP, Rujescu D, Rutten BPF, Shaikh M, Simons CJP, Toulopoulou T, Weisbrod M, van Winkel R, Kuchenbaecker K, McQuillin A, Bramon E. Psychosis Endophenotypes: A Gene-Set-Specific Polygenic Risk Score Analysis. Schizophr Bull 2023; 49:1625-1636. [PMID: 37582581 PMCID: PMC10686343 DOI: 10.1093/schbul/sbad088] [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] [Indexed: 08/17/2023]
Abstract
BACKGROUND AND HYPOTHESIS Endophenotypes can help to bridge the gap between psychosis and its genetic predispositions, but their underlying mechanisms remain largely unknown. This study aims to identify biological mechanisms that are relevant to the endophenotypes for psychosis, by partitioning polygenic risk scores into specific gene sets and testing their associations with endophenotypes. STUDY DESIGN We computed polygenic risk scores for schizophrenia and bipolar disorder restricted to brain-related gene sets retrieved from public databases and previous publications. Three hundred and seventy-eight gene-set-specific polygenic risk scores were generated for 4506 participants. Seven endophenotypes were also measured in the sample. Linear mixed-effects models were fitted to test associations between each endophenotype and each gene-set-specific polygenic risk score. STUDY RESULTS After correction for multiple testing, we found that a reduced P300 amplitude was associated with a higher schizophrenia polygenic risk score of the forebrain regionalization gene set (mean difference per SD increase in the polygenic risk score: -1.15 µV; 95% CI: -1.70 to -0.59 µV; P = 6 × 10-5). The schizophrenia polygenic risk score of forebrain regionalization also explained more variance of the P300 amplitude (R2 = 0.032) than other polygenic risk scores, including the genome-wide polygenic risk scores. CONCLUSIONS Our finding on reduced P300 amplitudes suggests that certain genetic variants alter early brain development thereby increasing schizophrenia risk years later. Gene-set-specific polygenic risk scores are a useful tool to elucidate biological mechanisms of psychosis and endophenotypes, offering leads for experimental validation in cellular and animal models.
Collapse
Affiliation(s)
- Baihan Wang
- Department of Mental Health Neuroscience, Division of Psychiatry, University College London, London, UK
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Haritz Irizar
- Department of Mental Health Neuroscience, Division of Psychiatry, University College London, London, UK
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Johan H Thygesen
- Department of Mental Health Neuroscience, Division of Psychiatry, University College London, London, UK
- Institute of Health Informatics, University College London, London, UK
| | - Eirini Zartaloudi
- Department of Mental Health Neuroscience, Division of Psychiatry, University College London, London, UK
| | - Isabelle Austin-Zimmerman
- Department of Mental Health Neuroscience, Division of Psychiatry, University College London, London, UK
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Anjali Bhat
- Department of Mental Health Neuroscience, Division of Psychiatry, University College London, London, UK
| | - Jasmine Harju-Seppänen
- Department of Mental Health Neuroscience, Division of Psychiatry, University College London, London, UK
| | - Oliver Pain
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Nick Bass
- Department of Mental Health Neuroscience, Division of Psychiatry, University College London, London, UK
| | - Vasiliki Gkofa
- Department of Mental Health Neuroscience, Division of Psychiatry, University College London, London, UK
| | - Behrooz Z Alizadeh
- University of Groningen, University Medical Center Groningen, University Center for Psychiatry, Rob Giel Research Center, Groningen, The Netherlands
- Department of Epidemiology, University Medical Center Groningen, Groningen, The Netherlands
| | - Therese van Amelsvoort
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Maria J Arranz
- Fundació Docència i Recerca Mutua Terrassa, Terrassa, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Institut de Recerca Biomédica Sant Pau (IIB-Sant Pau), Barcelona, Spain
| | - Stephan Bender
- Department of Child and Adolescent Psychiatry, Psychosomatic Medicine and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Wiepke Cahn
- Department of Psychiatry, Brain Centre Rudolf Magnus, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Altrecht, General Mental Health Care, Utrecht, The Netherlands
| | - Maria Stella Calafato
- Department of Mental Health Neuroscience, Division of Psychiatry, University College London, London, UK
| | - Benedicto Crespo-Facorro
- CIBERSAM, Centro Investigación Biomédica en Red Salud Mental, Sevilla, Spain
- Department of Psychiatry, University Hospital Virgen del Rocio, School of Medicine, University of Sevilla–IBiS, Sevilla, Spain
| | - Marta Di Forti
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | | | - Ina Giegling
- Comprehensive Centers for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Austria
| | - Lieuwe de Haan
- Department of Psychiatry, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Arkin, Institute for Mental Health, Amsterdam, The Netherlands
| | - Jeremy Hall
- Neuroscience and Mental Health Innovation Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Mandy Road, Cardiff, UK
| | - Mei-Hua Hall
- Psychosis Neurobiology Laboratory, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Neeltje van Haren
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus University Medical Center, Sophia’s Children Hospital, Rotterdam, The Netherlands
| | - Conrad Iyegbe
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - René S Kahn
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eugenia Kravariti
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Stephen M Lawrie
- Division of Psychiatry, University of Edinburgh, Royal Edinburgh Hospital, Edinburgh, UK
| | - Kuang Lin
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Jurjen J Luykx
- Department of Psychiatry, Brain Centre Rudolf Magnus, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Ignacio Mata
- Fundacion Argibide, Pamplona, Spain
- CIBERSAM, Centro Investigación Biomédica en Red Salud Mental, Madrid, Spain
| | - Colm McDonald
- The Centre for Neuroimaging & Cognitive Genomics (NICOG) and NCBES Galway Neuroscience Centre, University of Galway, Galway, Ireland
| | - Andrew M McIntosh
- Division of Psychiatry, University of Edinburgh, Royal Edinburgh Hospital, Edinburgh, UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
| | - Robin M Murray
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | | | - Marco Picchioni
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
- St Magnus Hospital, Surrey, UK
| | - John Powell
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Diana P Prata
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
- Instituto de Biofísica e Engenharia Biomédica, Faculdade de Ciencias da Universidade de Lisboa, Portugal
| | - Dan Rujescu
- Department of Psychiatry, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Division of General Psychiatry, Medical University of Vienna, Austria
| | - Bart P F Rutten
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Madiha Shaikh
- North East London Foundation Trust, London, UK
- Research Department of Clinical, Educational and Health Psychology, University College London, London, UK
| | - Claudia J P Simons
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, The Netherlands
- GGzE Institute for Mental Health Care, Eindhoven, The Netherlands
| | - Timothea Toulopoulou
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
- Interdisciplinary Program in Neuroscience, Aysel Sabuncu Brain Research Center, Bilkent University, Ankara, Türkiye
- National Magnetic Resonance Research Center (UMRAM), Bilkent University, Ankara, Türkiye
- Department of Psychology, Bilkent University, Ankara, Türkiye
- School of Medicine, Department of Psychiatry, National and Kapodistrian University of Athens, Athens, Greece
- Department of Psychiatry and Behavioral Health System, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Matthias Weisbrod
- Department of General Psychiatry, Center of Psychosocial Medicine, University of Heidelberg, Germany
- SRH Klinikum, Karlsbad-Langensteinbach, Germany
| | - Ruud van Winkel
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, The Netherlands
- KU Leuven, Department of Neuroscience, Research Group Psychiatry, Leuven, Belgium
| | - Karoline Kuchenbaecker
- Department of Mental Health Neuroscience, Division of Psychiatry, University College London, London, UK
- UCL Genetics Institute, Division of Biosciences, University College London, London, UK
| | - Andrew McQuillin
- Department of Mental Health Neuroscience, Division of Psychiatry, University College London, London, UK
| | - Elvira Bramon
- Department of Mental Health Neuroscience, Division of Psychiatry, University College London, London, UK
- Institute of Cognitive Neuroscience, University College London, London, UK
| |
Collapse
|
11
|
Tung VSK, Mathews F, Boruk M, Suppa G, Foronjy R, Pato MT, Pato CN, Knowles JA, Evgrafov OV. Cultured Mesenchymal Cells from Nasal Turbinate as a Cellular Model of the Neurodevelopmental Component of Schizophrenia Etiology. Int J Mol Sci 2023; 24:15339. [PMID: 37895019 PMCID: PMC10607243 DOI: 10.3390/ijms242015339] [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: 08/14/2023] [Revised: 10/02/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
The study of neurodevelopmental molecular mechanisms in schizophrenia requires the development of adequate biological models such as patient-derived cells and their derivatives. We previously utilized cell lines with neural progenitor properties (CNON) derived from the superior or middle turbinates of patients with schizophrenia and control groups to study schizophrenia-specific gene expression. In this study, we analyzed single-cell RNA seq data from two CNON cell lines (one derived from an individual with schizophrenia (SCZ) and the other from a control group) and two biopsy samples from the middle turbinate (MT) (also from an individual with SCZ and a control). We compared our data with previously published data regarding the olfactory neuroepithelium and demonstrated that CNON originated from a single cell type present both in middle turbinate and the olfactory neuroepithelium and expressed in multiple markers of mesenchymal cells. To define the relatedness of CNON to the developing human brain, we also compared CNON datasets with scRNA-seq data derived from an embryonic brain and found that the expression profile of the CNON closely matched the expression profile one of the cell types in the embryonic brain. Finally, we evaluated the differences between SCZ and control samples to assess the utility and potential benefits of using CNON single-cell RNA seq to study the etiology of schizophrenia.
Collapse
Affiliation(s)
- Victoria Sook Keng Tung
- Department of Cell Biology, State University of New York at Downstate, Brooklyn, NY 11203, USA
| | - Fasil Mathews
- Department of Otolaryngology, State University of New York at Downstate, Brooklyn, NY 11203, USA
| | - Marina Boruk
- Department of Otolaryngology, State University of New York at Downstate, Brooklyn, NY 11203, USA
| | - Gabrielle Suppa
- Department of Cell Biology, State University of New York at Downstate, Brooklyn, NY 11203, USA
| | - Robert Foronjy
- Department of Cell Biology, State University of New York at Downstate, Brooklyn, NY 11203, USA
| | - Michele T. Pato
- Department of Psychiatry, Rutgers University, Piscataway, NJ 08854, USA (C.N.P.)
| | - Carlos N. Pato
- Department of Psychiatry, Rutgers University, Piscataway, NJ 08854, USA (C.N.P.)
| | - James A. Knowles
- Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ 08854, USA;
| | - Oleg V. Evgrafov
- Department of Cell Biology, State University of New York at Downstate, Brooklyn, NY 11203, USA
- Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ 08854, USA;
| |
Collapse
|
12
|
Réthelyi JM, Vincze K, Schall D, Glennon J, Berkel S. The role of insulin/IGF1 signalling in neurodevelopmental and neuropsychiatric disorders - Evidence from human neuronal cell models. Neurosci Biobehav Rev 2023; 153:105330. [PMID: 37516219 DOI: 10.1016/j.neubiorev.2023.105330] [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: 09/30/2022] [Revised: 07/15/2023] [Accepted: 07/26/2023] [Indexed: 07/31/2023]
Abstract
Insulin and insulin-like growth factor 1 (IGF1) signalling play a central role in the development and maintenance of neurons in the brain, and human neurodevelopmental as well as neuropsychiatric disorders have been linked to impaired insulin and IGF1 signalling. This review focuses on the impairments of the insulin and IGF1 signalling cascade in the context of neurodevelopmental and neuropsychiatric disorders, based on evidence from human neuronal cell models. Clear evidence was obtained for impaired insulin and IGF1 receptor downstream signalling in neurodevelopmental disorders, while the evidence for its role in neuropsychiatric disorders was less substantial. Human neuronal model systems can greatly add to our knowledge about insulin/IGF1 signalling in the brain, its role in restoring dendritic maturity, and complement results from clinical studies and animal models. Moreover, they represent a useful model for the development of new therapeutic strategies. Further research is needed to systematically investigate the exact role of the insulin/IGF1 signalling cascades in neurodevelopmental and neuropsychiatric disorders, and to elucidate the respective therapeutic implications.
Collapse
Affiliation(s)
- János M Réthelyi
- Department of Psychiatry and Psychotherapy, Semmelweis University, Budapest, Hungary
| | - Katalin Vincze
- Department of Psychiatry and Psychotherapy, Semmelweis University, Budapest, Hungary; Doctoral School of Mental Health Sciences, Semmelweis University, Budapest, Hungary
| | - Dorothea Schall
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Jeffrey Glennon
- Conway Institute of Biomedical and Biomolecular Research, School of Medicine, University College Dublin, Dublin, Ireland
| | - Simone Berkel
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany; Interdisciplinary Centre of Neurosciences (IZN), Heidelberg University, Germany.
| |
Collapse
|
13
|
Kathuria A, Lopez-Lengowski K, Watmuff B, Karmacharya R. Morphological and transcriptomic analyses of stem cell-derived cortical neurons reveal mechanisms underlying synaptic dysfunction in schizophrenia. Genome Med 2023; 15:58. [PMID: 37507766 PMCID: PMC10375745 DOI: 10.1186/s13073-023-01203-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 06/16/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND Postmortem studies in schizophrenia consistently show reduced dendritic spines in the cerebral cortex but the mechanistic underpinnings of these deficits remain unknown. Recent genome-wide association studies and exome sequencing investigations implicate synaptic genes and processes in the disease biology of schizophrenia. METHODS We generated human cortical pyramidal neurons by differentiating iPSCs of seven schizophrenia patients and seven healthy subjects, quantified dendritic spines and synapses in different cortical neuron subtypes, and carried out transcriptomic studies to identify differentially regulated genes and aberrant cellular processes in schizophrenia. RESULTS Cortical neurons expressing layer III marker CUX1, but not those expressing layer V marker CTIP2, showed significant reduction in dendritic spine density in schizophrenia, mirroring findings in postmortem studies. Transcriptomic experiments in iPSC-derived cortical neurons showed that differentially expressed genes in schizophrenia were enriched for genes implicated in schizophrenia in genome-wide association and exome sequencing studies. Moreover, most of the differentially expressed genes implicated in schizophrenia genetic studies had lower expression levels in schizophrenia cortical neurons. Network analysis of differentially expressed genes led to identification of NRXN3 as a hub gene, and follow-up experiments showed specific reduction of the NRXN3 204 isoform in schizophrenia neurons. Furthermore, overexpression of the NRXN3 204 isoform in schizophrenia neurons rescued the spine and synapse deficits in the cortical neurons while knockdown of NRXN3 204 in healthy neurons phenocopied spine and synapse deficits seen in schizophrenia cortical neurons. The antipsychotic clozapine increased expression of the NRXN3 204 isoform in schizophrenia cortical neurons and rescued the spine and synapse density deficits. CONCLUSIONS Taken together, our findings in iPSC-derived cortical neurons recapitulate cell type-specific findings in postmortem studies in schizophrenia and have led to the identification of a specific isoform of NRXN3 that modulates synaptic deficits in schizophrenia neurons.
Collapse
Affiliation(s)
- Annie Kathuria
- Harvard University, MGH Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, CPZN6, Boston, MA, 02114, USA
- Chemical Biology Program, Broad Institute of MIT & Harvard, Cambridge, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Kara Lopez-Lengowski
- Harvard University, MGH Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, CPZN6, Boston, MA, 02114, USA
- Chemical Biology Program, Broad Institute of MIT & Harvard, Cambridge, MA, USA
| | - Bradley Watmuff
- Harvard University, MGH Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, CPZN6, Boston, MA, 02114, USA
- Chemical Biology Program, Broad Institute of MIT & Harvard, Cambridge, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Rakesh Karmacharya
- Harvard University, MGH Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, CPZN6, Boston, MA, 02114, USA.
- Chemical Biology Program, Broad Institute of MIT & Harvard, Cambridge, MA, USA.
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA.
- Program in Neuroscience, Harvard University, Cambridge, MA, USA.
- Schizophrenia & Bipolar Disorder Program, McLean Hospital, Belmont, MA, USA.
- Program in Chemical Biology, Harvard University, Cambridge, MA, USA.
- Harvard Stem Cell Institute, Cambridge, MA, USA.
| |
Collapse
|
14
|
Stogsdill JA, Harwell CC, Goldman SA. Astrocytes as master modulators of neural networks: Synaptic functions and disease-associated dysfunction of astrocytes. Ann N Y Acad Sci 2023; 1525:41-60. [PMID: 37219367 DOI: 10.1111/nyas.15004] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Astrocytes are the most abundant glial cell type in the central nervous system and are essential to the development, plasticity, and maintenance of neural circuits. Astrocytes are heterogeneous, with their diversity rooted in developmental programs modulated by the local brain environment. Astrocytes play integral roles in regulating and coordinating neural activity extending far beyond their metabolic support of neurons and other brain cell phenotypes. Both gray and white matter astrocytes occupy critical functional niches capable of modulating brain physiology on time scales slower than synaptic activity but faster than those adaptive responses requiring a structural change or adaptive myelination. Given their many associations and functional roles, it is not surprising that astrocytic dysfunction has been causally implicated in a broad set of neurodegenerative and neuropsychiatric disorders. In this review, we focus on recent discoveries concerning the contributions of astrocytes to the function of neural networks, with a dual focus on the contribution of astrocytes to synaptic development and maturation, and on their role in supporting myelin integrity, and hence conduction and its regulation. We then address the emerging roles of astrocytic dysfunction in disease pathogenesis and on potential strategies for targeting these cells for therapeutic purposes.
Collapse
Affiliation(s)
| | - Corey C Harwell
- Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Steven A Goldman
- Sana Biotechnology Inc., Cambridge, Massachusetts, USA
- Center for Translational Neuromedicine, University of Rochester, Rochester, New York, USA
- University of Copenhagen Faculty of Health and Medical Sciences, Copenhagen, Denmark
| |
Collapse
|
15
|
Arioka Y, Okumura H, Sakaguchi H, Ozaki N. Shedding light on latent pathogenesis and pathophysiology of mental disorders: The potential of iPS cell technology. Psychiatry Clin Neurosci 2023; 77:308-314. [PMID: 36929185 PMCID: PMC11488641 DOI: 10.1111/pcn.13545] [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: 10/04/2022] [Revised: 03/04/2023] [Accepted: 03/13/2023] [Indexed: 03/18/2023]
Abstract
Mental disorders are considered as one of the major healthcare issues worldwide owing to their significant impact on the quality of life of patients, causing serious social burdens. However, it is hard to examine the living brain-a source of psychiatric symptoms-at the cellular, subcellular, and molecular levels, which poses difficulty in determining the pathogenesis and pathophysiology of mental disorders. Recently, induced pluripotent stem cell (iPSC) technology has been used as a novel tool for research on mental disorders. We believe that the iPSC-based studies will address the limitations of other research approaches, such as human genome, postmortem brain study, brain imaging, and animal model analysis. Notably, studies using integrated iPSC technology with genetic information have provided significant novel findings to date. This review aimed to discuss the history, current trends, potential, and future of iPSC technology in the field of mental disorders. Although iPSC technology has several limitations, this technology can be used in combination with the other approaches to facilitate studies on mental disorders.
Collapse
Affiliation(s)
- Yuko Arioka
- Pathophysiology of Mental DisordersNagoya University Graduate School of MedicineNagoyaJapan
- Center for Advanced Medicine and Clinical ResearchNagoya University HospitalNagoyaJapan
| | - Hiroki Okumura
- Pathophysiology of Mental DisordersNagoya University Graduate School of MedicineNagoyaJapan
- Hospital PharmacyNagoya University HospitalNagoyaJapan
| | - Hideya Sakaguchi
- BDR‐Otsuka Pharmaceutical Collaboration Center, RIKEN Center for Biosystems Dynamics ResearchKobeJapan
| | - Norio Ozaki
- Pathophysiology of Mental DisordersNagoya University Graduate School of MedicineNagoyaJapan
- Institute for Glyco‐core Research (iGCORE)Nagoya UniversityNagoyaJapan
| |
Collapse
|
16
|
Rukh S, Meechan DW, Maynard TM, Lamantia AS. Out of Line or Altered States? Neural Progenitors as a Target in a Polygenic Neurodevelopmental Disorder. Dev Neurosci 2023; 46:1-21. [PMID: 37231803 DOI: 10.1159/000530898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 04/19/2023] [Indexed: 05/27/2023] Open
Abstract
The genesis of a mature complement of neurons is thought to require, at least in part, precursor cell lineages in which neural progenitors have distinct identities recognized by exclusive expression of one or a few molecular markers. Nevertheless, limited progenitor types distinguished by specific markers and lineal progression through such subclasses cannot easily yield the magnitude of neuronal diversity in most regions of the nervous system. The late Verne Caviness, to whom this edition of Developmental Neuroscience is dedicated, recognized this mismatch. In his pioneering work on the histogenesis of the cerebral cortex, he acknowledged the additional flexibility required to generate multiple classes of cortical projection and interneurons. This flexibility may be accomplished by establishing cell states in which levels rather than binary expression or repression of individual genes vary across each progenitor's shared transcriptome. Such states may reflect local, stochastic signaling via soluble factors or coincidence of cell surface ligand/receptor pairs in subsets of neighboring progenitors. This probabilistic, rather than determined, signaling could modify transcription levels via multiple pathways within an apparently uniform population of progenitors. Progenitor states, therefore, rather than lineal relationships between types may underlie the generation of neuronal diversity in most regions of the nervous system. Moreover, mechanisms that influence variation required for flexible progenitor states may be targets for pathological changes in a broad range of neurodevelopmental disorders, especially those with polygenic origins.
Collapse
Affiliation(s)
- Shah Rukh
- Fralin Biomedical Research Institute, Virginia Tech Carilion School of Medicine, Roanoke, Virginia, USA
| | - Daniel W Meechan
- Fralin Biomedical Research Institute, Virginia Tech Carilion School of Medicine, Roanoke, Virginia, USA
| | - Thomas M Maynard
- Fralin Biomedical Research Institute, Virginia Tech Carilion School of Medicine, Roanoke, Virginia, USA
| | - Anthony-Samuel Lamantia
- Fralin Biomedical Research Institute, Virginia Tech Carilion School of Medicine, Roanoke, Virginia, USA
- Department of Biological Sciences, Virginia Tech, Blacksburg, Virginia, USA
| |
Collapse
|
17
|
Puigdevall P, Jerber J, Danecek P, Castellano S, Kilpinen H. Somatic mutations alter the differentiation outcomes of iPSC-derived neurons. CELL GENOMICS 2023; 3:100280. [PMID: 37082143 PMCID: PMC10112289 DOI: 10.1016/j.xgen.2023.100280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 11/11/2022] [Accepted: 02/23/2023] [Indexed: 04/22/2023]
Abstract
The use of induced pluripotent stem cells (iPSC) as models for development and human disease has enabled the study of otherwise inaccessible tissues. A remaining challenge in developing reliable models is our limited understanding of the factors driving irregular differentiation of iPSCs, particularly the impact of acquired somatic mutations. We leveraged data from a pooled dopaminergic neuron differentiation experiment of 238 iPSC lines profiled with single-cell RNA and whole-exome sequencing to study how somatic mutations affect differentiation outcomes. We found that deleterious somatic mutations in key developmental genes, notably the BCOR gene, are strongly associated with failure in dopaminergic neuron differentiation and a larger proliferation rate in culture. We further identified broad differences in cell type composition between incorrectly and successfully differentiating lines, as well as significant changes in gene expression contributing to the inhibition of neurogenesis. Our work calls for caution in interpreting differentiation-related phenotypes in disease-modeling experiments.
Collapse
Affiliation(s)
- Pau Puigdevall
- UCL Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
- Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Haartmaninkatu 8, PO Box 63, Helsinki 00014, Finland
| | - Julie Jerber
- Open Targets, Wellcome Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Petr Danecek
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Sergi Castellano
- UCL Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
| | - Helena Kilpinen
- UCL Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
- Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Haartmaninkatu 8, PO Box 63, Helsinki 00014, Finland
- Faculty of Biological and Environmental Sciences, University of Helsinki, Viikinkaari 1, PO Box 65, Helsinki 00014, Finland
| |
Collapse
|
18
|
Abstract
Schizophrenia is a neurodevelopmental disorder with genetic and environmental factors involved in its aetiology. Genetic liability contributing to the development of schizophrenia is a subject of extensive research activity, as reliable data regarding its aetiology would enable the improvement of its therapy and the development of new methods of treatment. A multitude of studies in this field focus on genetic variants, such as copy number variations (CNVs) or single-nucleotide variants (SNVs). Certain genetic disorders caused by CNVs including 22q11.2 microdeletion syndrome, Burnside-Butler syndrome (15q11.2 BP1-BP2 microdeletion) or 1q21.1 microduplication/microdeletion syndrome are associated with a higher risk of developing schizophrenia. In this article, we provide a unifying framework linking these CNVs and their associated genetic disorders with schizophrenia and its various neural and behavioural abnormalities.
Collapse
|
19
|
Yde Ohki CM, Walter NM, Bender A, Rickli M, Ruhstaller S, Walitza S, Grünblatt E. Growth rates of human induced pluripotent stem cells and neural stem cells from attention-deficit hyperactivity disorder patients: a preliminary study. J Neural Transm (Vienna) 2023; 130:243-252. [PMID: 36800023 PMCID: PMC10033475 DOI: 10.1007/s00702-023-02600-1] [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: 11/18/2022] [Accepted: 02/06/2023] [Indexed: 02/18/2023]
Abstract
Attention-deficit hyperactivity disorder (ADHD) is a neurodevelopmental polygenic disorder that affects more than 5% of children and adolescents around the world. Genetic and environmental factors play important roles in ADHD etiology, which leads to a wide range of clinical outcomes and biological phenotypes across the population. Brain maturation delays of a 4-year lag are commonly found in patients, when compared to controls of the same age. Possible differences in cellular growth rates might reflect the clinical observations in ADHD patients. However, the cellular mechanisms are still not elucidated. To test this hypothesis, we analysed the proliferation of induced pluripotent stem cells (iPSCs) and neural stem cells (NSCs) derived from male children and adolescents diagnosed with ADHD and with genetic predisposition to it (assessed using polygenic risk scores), as well as their respective matched controls. In the current pilot study, it was noticeable that NSCs from the ADHD group proliferate less than controls, while no differences were seen at the iPSC developmental stage. Our results from two distinct proliferation methods indicate that the functional and structural delays found in patients might be associated with these in vitro phenotypic differences, but start at a distinct neurodevelopmental stage. These findings are the first ones in the field of disease modelling of ADHD and might be crucial to better understand the pathophysiology of this disorder.
Collapse
Affiliation(s)
- Cristine Marie Yde Ohki
- Department of Child and Adolescent Psychiatry and Psychotherapy, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Biomedicine PhD Program, University of Zurich, Zurich, Switzerland
| | - Natalie Monet Walter
- Department of Child and Adolescent Psychiatry and Psychotherapy, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Audrey Bender
- Department of Child and Adolescent Psychiatry and Psychotherapy, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Michelle Rickli
- Department of Child and Adolescent Psychiatry and Psychotherapy, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Sina Ruhstaller
- Department of Child and Adolescent Psychiatry and Psychotherapy, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Susanne Walitza
- Department of Child and Adolescent Psychiatry and Psychotherapy, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and the ETH Zurich, Zurich, Switzerland
- Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Edna Grünblatt
- Department of Child and Adolescent Psychiatry and Psychotherapy, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland.
- Neuroscience Center Zurich, University of Zurich and the ETH Zurich, Zurich, Switzerland.
- Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland.
- Department of Child and Adolescent Psychiatry and Psychotherapy, Translational Molecular Psychiatry, Psychiatric University Hospital Zurich, University Zurich, University of Zurich, Wagistrasse 12, 8952, Schlieren, Switzerland.
| |
Collapse
|
20
|
Yuan Yin X, Hua Zhu Z, Feng Chen R, Li J, Ya Zhang G, Long Hou W, Nan Zhuang N, Cai Y, Bin Dai N, Liu F, Wang J, Fang Jia Q, Hui L. Generation and characterization of iPSCs from a patient with obsessive-compulsive disorder, his mother with schizophrenia and his healthy father. Stem Cell Res 2023; 66:102993. [PMID: 36502622 DOI: 10.1016/j.scr.2022.102993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 11/28/2022] [Accepted: 12/03/2022] [Indexed: 12/12/2022] Open
Abstract
Schizophrenia (SCZ) and obsessive-compulsive disorder (OCD) are complex polygenic disorders with brain morphology abnormalities. The etiologies and relationship of both disorders remain elusive, and should be further investigated. Thus, induced pluripotent stem cells (iPSCs) were generated from peripheral blood mononuclear cells (PBMCs) from an OCD patient, his mother with SCZ and his healthy father with reprograming method. All iPSCs were characterized to have normal karyotype and expression of pluripotency makers. These iPSCs will be a valuable model to elucidate the pathophysiological mechanisms and association of both diseases.
Collapse
Affiliation(s)
- Xu Yuan Yin
- Research Center of Biological Psychiatry, Suzhou Psychiatric Hospital, Suzhou Medical College, Soochow University, Suzhou 215137, Jiangsu, China
| | - Zhen Hua Zhu
- Research Center of Biological Psychiatry, Suzhou Psychiatric Hospital, Suzhou Medical College, Soochow University, Suzhou 215137, Jiangsu, China
| | - Ru Feng Chen
- Research Center of Biological Psychiatry, Suzhou Psychiatric Hospital, Suzhou Medical College, Soochow University, Suzhou 215137, Jiangsu, China
| | - Jian Li
- Changshu No. 2 People's Hospital The Affiliated Changshu Hospital of Xuzhou Medical University, Changshu 215500, China
| | - Guang Ya Zhang
- Research Center of Biological Psychiatry, Suzhou Psychiatric Hospital, Suzhou Medical College, Soochow University, Suzhou 215137, Jiangsu, China
| | - Wen Long Hou
- Research Center of Biological Psychiatry, Suzhou Psychiatric Hospital, Suzhou Medical College, Soochow University, Suzhou 215137, Jiangsu, China
| | - Nan Nan Zhuang
- Research Center of Biological Psychiatry, Suzhou Psychiatric Hospital, Suzhou Medical College, Soochow University, Suzhou 215137, Jiangsu, China
| | - Yuan Cai
- Research Center of Biological Psychiatry, Suzhou Psychiatric Hospital, Suzhou Medical College, Soochow University, Suzhou 215137, Jiangsu, China
| | - Ning Bin Dai
- Suzhou Center of Disease Control and Prevention, Suzhou 215004, RP China
| | - Fang Liu
- Suzhou Center of Disease Control and Prevention, Suzhou 215004, RP China
| | - Jing Wang
- Research Center of Biological Psychiatry, Suzhou Psychiatric Hospital, Suzhou Medical College, Soochow University, Suzhou 215137, Jiangsu, China
| | - Qiu Fang Jia
- Research Center of Biological Psychiatry, Suzhou Psychiatric Hospital, Suzhou Medical College, Soochow University, Suzhou 215137, Jiangsu, China.
| | - Li Hui
- Research Center of Biological Psychiatry, Suzhou Psychiatric Hospital, Suzhou Medical College, Soochow University, Suzhou 215137, Jiangsu, China.
| |
Collapse
|
21
|
Abulaiti X, Wang A, Zhang H, Su H, Gao R, Chen J, Gao S, Li L. Disrupted mossy fiber connections from defective embryonic neurogenesis contribute to SOX11-associated schizophrenia. Cell Mol Life Sci 2022; 79:180. [PMID: 35254515 PMCID: PMC11072709 DOI: 10.1007/s00018-022-04206-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/29/2022] [Accepted: 02/09/2022] [Indexed: 11/26/2022]
Abstract
Abnormal mossy fiber connections in the hippocampus have been implicated in schizophrenia. However, it remains unclear whether this abnormality in the patients is genetically determined and whether it contributes to the onset of schizophrenia. Here, we showed that iPSC-derived hippocampal NPCs from schizophrenia patients with the A/A allele at SNP rs16864067 exhibited abnormal NPC polarity, resulting from the downregulation of SOX11 by this high-risk allele. In the SOX11-deficient mouse brain, abnormal NPC polarity was also observed in the hippocampal dentate gyrus, and this abnormal NPC polarity led to defective hippocampal neurogenesis-specifically, irregular neuroblast distribution and disrupted granule cell morphology. As granule cell synapses, the mossy fiber pathway was disrupted, and this disruption was resistant to activity-induced mossy fiber remodeling in SOX11 mutant mice. Moreover, these mutant mice exhibited diminished PPI and schizophrenia-like behaviors. Activation of hippocampal neurogenesis in the embryonic brain, but not in the adult brain, partially alleviated disrupted mossy fiber connections and improved schizophrenia-related behaviors in mutant mice. We conclude that disrupted mossy fiber connections are genetically determined and strongly correlated with schizophrenia-like behaviors in SOX11-deficient mice. This disruption may reflect the pathological substrate of SOX11-associated schizophrenia.
Collapse
Affiliation(s)
- Xianmixinuer Abulaiti
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
- Shanghai Advanced Research Institute Chinese Academy of Sciences, Shanghai, 201210, China
| | - Aifang Wang
- Shanghai Advanced Research Institute Chinese Academy of Sciences, Shanghai, 201210, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Han Zhang
- Shanghai Advanced Research Institute Chinese Academy of Sciences, Shanghai, 201210, China
| | - Hang Su
- Shanghai Advanced Research Institute Chinese Academy of Sciences, Shanghai, 201210, China
- Henan Provincial People's Hospital of Zhengzhou University, Zhengzhou, 450003, Henan, China
| | - Rui Gao
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Jiayu Chen
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Shaorong Gao
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| | - Lingsong Li
- Shanghai Advanced Research Institute Chinese Academy of Sciences, Shanghai, 201210, China.
- Henan Provincial People's Hospital of Zhengzhou University, Zhengzhou, 450003, Henan, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
22
|
Karpov D, Golimbet V. Cellular and supracellular models in the study of molecular mechanisms associated with schizophrenia. Zh Nevrol Psikhiatr Im S S Korsakova 2022; 122:46-50. [DOI: 10.17116/jnevro202212211146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|