1
|
Liu C, Zhang C, Glatt SJ. Psychiatric Genomics 2025: State of the Art and the Path Forward. Psychiatr Clin North Am 2025; 48:217-240. [PMID: 40348414 DOI: 10.1016/j.psc.2025.01.002] [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: 05/14/2025]
Abstract
Psychiatric genetics has evolved from candidate-gene studies to whole-genome sequencing efforts. With hundreds of disease-associated loci now identified, functional interpretation of the associated loci becomes the critical next step toward translational applications. The article discusses achievements, challenges, and opportunities in psychiatric genomics associated with complexity and heterogeneity. Brain expression quantitative trait loci, single-cell ribonucleic acid-sequence, and functional genomics technologies are highlighted. It also covers newly developed techniques with improved spatiotemporal resolution, quality and sensitivity, coupled with advanced analytical methods and artificial intelligence. The power of collaborative research and inclusion of diverse populations will ensure a bright future for precision psychiatry.
Collapse
Affiliation(s)
- Chunyu Liu
- Department of Psychiatry, SUNY Upstate Medical University, 505 Irving Avenue, Syracuse, NY 13210, USA.
| | - Chunling Zhang
- Department of Neuroscience & Physiology, SUNY Upstate Medical University, 505 Irving Avenue, Syracuse, NY 13210, USA
| | - Stephen J Glatt
- Department of Psychiatry, SUNY Upstate Medical University, 505 Irving Avenue, Syracuse, NY 13210, USA
| |
Collapse
|
2
|
Liu C, Gershon ES. Endophenotype 2.0: updated definitions and criteria for endophenotypes of psychiatric disorders, incorporating new technologies and findings. Transl Psychiatry 2024; 14:502. [PMID: 39719446 PMCID: PMC11668880 DOI: 10.1038/s41398-024-03195-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 11/28/2024] [Accepted: 12/05/2024] [Indexed: 12/26/2024] Open
Abstract
Recent genetic studies have linked numerous loci to psychiatric disorders. However, the biological pathways that connect these genetic associations to psychiatric disorders' specific pathophysiological processes are largely unclear. Endophenotypes, first defined over five decades ago, are heritable traits, independent of disease state that are associated with a disease, encompassing a broad range of neurophysiological, biochemical, endocrinological, neuroanatomical, cognitive, and neuropsychological characteristics. Considering the advancements in genetics and genomics over recent decades, we propose a revised definition of endophenotypes as 'genetically influenced phenotypes linked to disease or treatment characteristics and their related events.' We also updated endophenotype criteria to include (1) reliable measurement, (2) association with the disease or its related events, and (3) genetic mediation. 'Genetic mediation' is introduced to differentiate between causality and pleiotropic effects and allows non-linear relationships. Furthermore, this updated Endophenotype 2.0 framework expands to encompass genetically regulated responses to disease-related factors, including environmental risks, illness progression, treatment responses, and resilience phenotypes, which may be state-dependent. This broadened definition paves the way for developing new endophenotypes crucial for genetic analyses in psychiatric disorders. Integrating genetics, genomics, and diverse endophenotypes into multi-dimensional mechanistic models is vital for advancing our understanding of psychiatric disorders. Crucially, elucidating the biological underpinnings of endophenotypes will enhance our grasp of psychiatric genetics, thereby improving disease risk prediction and treatment approaches.
Collapse
Affiliation(s)
- Chunyu Liu
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, USA.
- School of Life Sciences, Central South University, Changsha, China.
| | - Elliot S Gershon
- Departments of Psychiatry and Human Genetics, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
3
|
Choudhary A, Peles D, Nayak R, Mizrahi L, Stern S. Current progress in understanding schizophrenia using genomics and pluripotent stem cells: A meta-analytical overview. Schizophr Res 2024; 273:24-38. [PMID: 36443183 DOI: 10.1016/j.schres.2022.11.001] [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: 06/23/2022] [Revised: 10/16/2022] [Accepted: 11/01/2022] [Indexed: 11/27/2022]
Abstract
Schizophrenia (SCZ) is a complex, heritable and polygenic neuropsychiatric disease, which disables the patients as well as decreases their life expectancy and quality of life. Common and rare variants studies on SCZ subjects have provided >100 genomic loci that hold importance in the context of SCZ pathophysiology. Transcriptomic studies from clinical samples have informed about the differentially expressed genes (DEGs) and non-coding RNAs in SCZ patients. Despite these advancements, no causative genes for SCZ were found and hence SCZ is difficult to recapitulate in animal models. In the last decade, induced Pluripotent Stem Cells (iPSCs)-based models have helped in understanding the neural phenotypes of SCZ by studying patient iPSC-derived 2D neuronal cultures and 3D brain organoids. Here, we have aimed to provide a simplistic overview of the current progress and advancements after synthesizing the enormous literature on SCZ genetics and SCZ iPSC-based models. Although further understanding of SCZ genetics and pathophysiological mechanisms using these technological advancements is required, the recent approaches have allowed to delineate important cellular mechanisms and biological pathways affected in SCZ.
Collapse
Affiliation(s)
- Ashwani Choudhary
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel
| | - David Peles
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel
| | - Ritu Nayak
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel
| | - Liron Mizrahi
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel
| | - Shani Stern
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel.
| |
Collapse
|
4
|
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
|
5
|
Garone C, De Giorgio F, Carli S. Mitochondrial metabolism in neural stem cells and implications for neurodevelopmental and neurodegenerative diseases. J Transl Med 2024; 22:238. [PMID: 38438847 PMCID: PMC10910780 DOI: 10.1186/s12967-024-05041-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: 12/29/2023] [Accepted: 02/25/2024] [Indexed: 03/06/2024] Open
Abstract
Mitochondria are cytoplasmic organelles having a fundamental role in the regulation of neural stem cell (NSC) fate during neural development and maintenance.During embryonic and adult neurogenesis, NSCs undergo a metabolic switch from glycolytic to oxidative phosphorylation with a rise in mitochondrial DNA (mtDNA) content, changes in mitochondria shape and size, and a physiological augmentation of mitochondrial reactive oxygen species which together drive NSCs to proliferate and differentiate. Genetic and epigenetic modifications of proteins involved in cellular differentiation (Mechanistic Target of Rapamycin), proliferation (Wingless-type), and hypoxia (Mitogen-activated protein kinase)-and all connected by the common key regulatory factor Hypoxia Inducible Factor-1A-are deemed to be responsible for the metabolic shift and, consequently, NSC fate in physiological and pathological conditions.Both primary mitochondrial dysfunction due to mutations in nuclear DNA or mtDNA or secondary mitochondrial dysfunction in oxidative phosphorylation (OXPHOS) metabolism, mitochondrial dynamics, and organelle interplay pathways can contribute to the development of neurodevelopmental or progressive neurodegenerative disorders.This review analyses the physiology and pathology of neural development starting from the available in vitro and in vivo models and highlights the current knowledge concerning key mitochondrial pathways involved in this process.
Collapse
Affiliation(s)
- C Garone
- Department of Medical and Surgical Sciences, Alma Mater Studiorum-University of Bologna, Bologna, Italy.
- IRCCS Istituto Delle Scienze Neurologiche di Bologna, UO Neuropsichiatria Dell'età Pediatrica, Bologna, Italy.
| | - F De Giorgio
- Department of Medical and Surgical Sciences, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - S Carli
- Department of Medical and Surgical Sciences, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| |
Collapse
|
6
|
Heider J, Stahl A, Sperlich D, Hartmann SM, Vogel S, Breitmeyer R, Templin M, Volkmer H. Defined co-cultures of glutamatergic and GABAergic neurons with a mutation in DISC1 reveal aberrant phenotypes in GABAergic neurons. BMC Neurosci 2024; 25:12. [PMID: 38438989 PMCID: PMC10910844 DOI: 10.1186/s12868-024-00858-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 02/26/2024] [Indexed: 03/06/2024] Open
Abstract
BACKGROUND Mutations in the gene DISC1 are associated with increased risk for schizophrenia, bipolar disorder and major depression. The study of mutated DISC1 represents a well-known and comprehensively characterized approach to understand neuropsychiatric disease mechanisms. However, previous studies have mainly used animal models or rather heterogeneous populations of iPSC-derived neurons, generated by undirected differentiation, to study the effects of DISC1 disruption. Since major hypotheses to explain neurodevelopmental, psychiatric disorders rely on altered neuronal connectivity observed in patients, an ideal iPSC-based model requires accurate representation of the structure and complexity of neuronal circuitries. In this study, we made use of an isogenic cell line with a mutation in DISC1 to study neuronal synaptic phenotypes in a culture system comprising a defined ratio of NGN2 and ASCL1/DLX2 (AD2)-transduced neurons, enriched for glutamatergic and GABAergic neurons, respectively, to mimic properties of the cortical microcircuitry. RESULTS In heterozygous DISC1 mutant neurons, we replicated the expected phenotypes including altered neural progenitor proliferation as well as neurite outgrowth, deregulated DISC1-associated signaling pathways, and reduced synaptic densities in cultures composed of glutamatergic neurons. Cultures comprising a defined ratio of NGN2 and AD2 neurons then revealed considerably increased GABAergic synapse densities, which have not been observed in any iPSC-derived model so far. Increased inhibitory synapse densities could be associated with an increased efficiency of GABAergic differentiation, which we observed in AD2-transduced cultures of mutant neurons. Additionally, we found increased neuronal activity in GABAergic neurons through calcium imaging while the activity pattern of glutamatergic neurons remained unchanged. CONCLUSIONS In conclusion, our results demonstrate phenotypic differences in a co-culture comprising a defined ratio of DISC1 mutant NGN2 and AD2 neurons, as compared to culture models comprising only one neuronal cell type. Altered synapse numbers and neuronal activity imply that DISC1 impacts the excitatory/inhibitory balance in NGN2/AD2 co-cultures, mainly through increased GABAergic input.
Collapse
Affiliation(s)
- Johanna Heider
- Department of Pharma and Biotech, NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770, Reutlingen, Germany
| | - Aaron Stahl
- Department of Pharma and Biotech, NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770, Reutlingen, Germany
| | - Denise Sperlich
- Department of Pharma and Biotech, NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770, Reutlingen, Germany
| | - Sophia-Marie Hartmann
- Department of Pharma and Biotech, NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770, Reutlingen, Germany
| | - Sabrina Vogel
- Department of Pharma and Biotech, NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770, Reutlingen, Germany
| | - Ricarda Breitmeyer
- Department of Pharma and Biotech, NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770, Reutlingen, Germany
| | - Markus Templin
- Department of Pharma and Biotech, NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770, Reutlingen, Germany
| | - Hansjürgen Volkmer
- Department of Pharma and Biotech, NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770, Reutlingen, Germany.
| |
Collapse
|
7
|
Perrottelli A, Marzocchi FF, Caporusso E, Giordano GM, Giuliani L, Melillo A, Pezzella P, Bucci P, Mucci A, Galderisi S. Advances in the understanding of the pathophysiology of schizophrenia and bipolar disorder through induced pluripotent stem cell models. J Psychiatry Neurosci 2024; 49:E109-E125. [PMID: 38490647 PMCID: PMC10950363 DOI: 10.1503/jpn.230112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 08/04/2023] [Accepted: 01/08/2024] [Indexed: 03/17/2024] Open
Abstract
The pathophysiology of schizophrenia and bipolar disorder involves a complex interaction between genetic and environmental factors that begins in the early stages of neurodevelopment. Recent advancements in the field of induced pluripotent stem cells (iPSCs) offer a promising tool for understanding the neurobiological alterations involved in these disorders and, potentially, for developing new treatment options. In this review, we summarize the results of iPSC-based research on schizophrenia and bipolar disorder, showing disturbances in neurodevelopmental processes, imbalance in glutamatergic-GABAergic transmission and neuromorphological alterations. The limitations of the reviewed literature are also highlighted, particularly the methodological heterogeneity of the studies, the limited number of studies developing iPSC models of both diseases simultaneously, and the lack of in-depth clinical characterization of the included samples. Further studies are needed to advance knowledge on the common and disease-specific pathophysiological features of schizophrenia and bipolar disorder and to promote the development of new treatment options.
Collapse
Affiliation(s)
| | | | | | | | - Luigi Giuliani
- From the University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Antonio Melillo
- From the University of Campania "Luigi Vanvitelli", Naples, Italy
| | | | - Paola Bucci
- From the University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Armida Mucci
- From the University of Campania "Luigi Vanvitelli", Naples, Italy
| | | |
Collapse
|
8
|
Deans PJM, Seah C, Johnson J, Gonzalez JG, Townsley K, Cao E, Schrode N, Stahl E, O’Reilly P, Huckins LM, Brennand KJ. Non-additive effects of schizophrenia risk genes reflect convergent downstream function. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.03.20.23287497. [PMID: 36993466 PMCID: PMC10055596 DOI: 10.1101/2023.03.20.23287497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Genetic studies of schizophrenia (SCZ) reveal a complex polygenic risk architecture comprised of hundreds of risk variants, the majority of which are common in the population at-large and confer only modest increases in disorder risk. Precisely how genetic variants with individually small predicted effects on gene expression combine to yield substantial clinical impacts in aggregate is unclear. Towards this, we previously reported that the combinatorial perturbation of four SCZ risk genes ("eGenes", whose expression is regulated by common variants) resulted in gene expression changes that were not predicted by individual perturbations, being most non-additive among genes associated with synaptic function and SCZ risk. Now, across fifteen SCZ eGenes, we demonstrate that non-additive effects are greatest within groups of functionally similar eGenes. Individual eGene perturbations reveal common downstream transcriptomic effects ("convergence"), while combinatorial eGene perturbations result in changes that are smaller than predicted by summing individual eGene effects ("sub-additive effects"). Unexpectedly, these convergent and sub-additive downstream transcriptomic effects overlap and constitute a large proportion of the genome-wide polygenic risk score, suggesting that functional redundancy of eGenes may be a major mechanism underlying non-additivity. Single eGene perturbations likewise fail to predict the magnitude or directionality of cellular phenotypes resulting from combinatorial perturbations. Overall, our results indicate that polygenic risk cannot be extrapolated from experiments testing one risk gene at a time and must instead be empirically measured. By unravelling the interactions between complex risk variants, it may be possible to improve the clinical utility of polygenic risk scores through more powerful prediction of symptom onset, clinical trajectory, and treatment response, or to identify novel targets for therapeutic intervention.
Collapse
Affiliation(s)
- PJ Michael Deans
- Departments of Psychiatry and Genetics, Division of Molecular Psychiatry, Department of Genetics, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT 06511
| | - Carina Seah
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomics, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Jessica Johnson
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomics, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Judit Garcia Gonzalez
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomics, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Kayla Townsley
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomics, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Evan Cao
- Departments of Psychiatry and Genetics, Division of Molecular Psychiatry, Department of Genetics, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT 06511
| | - Nadine Schrode
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomics, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Eli Stahl
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomics, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Paul O’Reilly
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomics, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Laura M. Huckins
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomics, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Kristen J. Brennand
- Departments of Psychiatry and Genetics, Division of Molecular Psychiatry, Department of Genetics, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT 06511
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomics, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| |
Collapse
|
9
|
Cardo LF, de la Fuente DC, Li M. Impaired neurogenesis and neural progenitor fate choice in a human stem cell model of SETBP1 disorder. Mol Autism 2023; 14:8. [PMID: 36805818 PMCID: PMC9940404 DOI: 10.1186/s13229-023-00540-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 02/07/2023] [Indexed: 02/22/2023] Open
Abstract
BACKGROUND Disruptions of SETBP1 (SET binding protein 1) on 18q12.3 by heterozygous gene deletion or loss-of-function variants cause SETBP1 disorder. Clinical features are frequently associated with moderate to severe intellectual disability, autistic traits and speech and motor delays. Despite the association of SETBP1 with neurodevelopmental disorders, little is known about its role in brain development. METHODS Using CRISPR/Cas9 genome editing technology, we generated a SETBP1 deletion model in human embryonic stem cells (hESCs) and examined the effects of SETBP1-deficiency in neural progenitors (NPCs) and neurons derived from these stem cells using a battery of cellular assays, genome-wide transcriptomic profiling and drug-based phenotypic rescue. RESULTS Neural induction occurred efficiently in all SETBP1 deletion models as indicated by uniform transition into neural rosettes. However, SETBP1-deficient NPCs exhibited an extended proliferative window and a decrease in neurogenesis coupled with a deficiency in their ability to acquire ventral forebrain fate. Genome-wide transcriptome profiling and protein biochemical analysis revealed enhanced activation of Wnt/β-catenin signaling in SETBP1 deleted cells. Crucially, treatment of the SETBP1-deficient NPCs with a small molecule Wnt inhibitor XAV939 restored hyper canonical β-catenin activity and restored both cortical and MGE neuronal differentiation. LIMITATIONS The current study is based on analysis of isogenic hESC lines with genome-edited SETBP1 deletion and further studies would benefit from the use of patient-derived iPSC lines that may harbor additional genetic risk that aggravate brain pathology of SETBP1 disorder. CONCLUSIONS We identified an important role for SETBP1 in controlling forebrain progenitor expansion and neurogenic differentiation. Our study establishes a novel regulatory link between SETBP1 and Wnt/β-catenin signaling during human cortical neurogenesis and provides mechanistic insights into structural abnormalities and potential therapeutic avenues for SETBP1 disorder.
Collapse
Affiliation(s)
- Lucia F Cardo
- Neuroscience and Mental Health Innovation Institute, School of Medicine and School of Bioscience, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK.
| | - Daniel C de la Fuente
- Neuroscience and Mental Health Innovation Institute, School of Medicine and School of Bioscience, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK
| | - Meng Li
- Neuroscience and Mental Health Innovation Institute, School of Medicine and School of Bioscience, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK.
| |
Collapse
|
10
|
Singh M, Agarwal V, Jindal D, Pancham P, Agarwal S, Mani S, Tiwari RK, Das K, Alghamdi BS, Abujamel TS, Ashraf GM, Jha SK. Recent Updates on Corticosteroid-Induced Neuropsychiatric Disorders and Theranostic Advancements through Gene Editing Tools. Diagnostics (Basel) 2023; 13:diagnostics13030337. [PMID: 36766442 PMCID: PMC9914305 DOI: 10.3390/diagnostics13030337] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 09/28/2022] [Accepted: 10/16/2022] [Indexed: 01/19/2023] Open
Abstract
The vast use of corticosteroids (CCSs) globally has led to an increase in CCS-induced neuropsychiatric disorders (NPDs), a very common manifestation in patients after CCS consumption. These neuropsychiatric disorders range from depression, insomnia, and bipolar disorders to panic attacks, overt psychosis, and many other cognitive changes in such subjects. Though their therapeutic importance in treating and improving many clinical symptoms overrides the complications that arise after their consumption, still, there has been an alarming rise in NPD cases in recent years, and they are seen as the greatest public health challenge globally; therefore, these potential side effects cannot be ignored. It has also been observed that many of the neuronal functional activities are regulated and controlled by genomic variants with epigenetic factors (DNA methylation, non-coding RNA, and histone modeling, etc.), and any alterations in these regulatory mechanisms affect normal cerebral development and functioning. This study explores a general overview of emerging concerns of CCS-induced NPDs, the effective molecular biology approaches that can revitalize NPD therapy in an extremely specialized, reliable, and effective manner, and the possible gene-editing-based therapeutic strategies to either prevent or cure NPDs in the future.
Collapse
Affiliation(s)
- Manisha Singh
- Department of Biotechnology, Jaypee Institute of Information Technology (JIIT), Noida 201309, India
- Correspondence: (M.S.); (S.K.J.)
| | - Vinayak Agarwal
- Department of Biotechnology, Jaypee Institute of Information Technology (JIIT), Noida 201309, India
| | - Divya Jindal
- Department of Biotechnology, Jaypee Institute of Information Technology (JIIT), Noida 201309, India
| | - Pranav Pancham
- Department of Biotechnology, Jaypee Institute of Information Technology (JIIT), Noida 201309, India
| | - Shriya Agarwal
- Department of Molecular Sciences, Macquarie University, Macquarie Park, NSW 2109, Australia
| | - Shalini Mani
- Department of Biotechnology, Jaypee Institute of Information Technology (JIIT), Noida 201309, India
| | - Raj Kumar Tiwari
- School of Health Sciences, Pharmaceutical Sciences, UPES, Dehradun 248007, India
| | - Koushik Das
- School of Health Sciences, Pharmaceutical Sciences, UPES, Dehradun 248007, India
| | - Badrah S. Alghamdi
- Department of Physiology, Neuroscience Unit, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Pre-Clinical Research Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Tukri S. Abujamel
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Ghulam Md. Ashraf
- Pre-Clinical Research Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Department of Medical Laboratory Sciences, College of Health Sciences, University of Sharjah, University City, Sharjah 27272, United Arab Emirates
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering & Technology, Sharda University, Greater Noida 201310, India
- Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun 248007, India
- Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali 140413, India
- Correspondence: (M.S.); (S.K.J.)
| |
Collapse
|
11
|
Ishihara K, Takata K, Mizutani KI. Involvement of an Aberrant Vascular System in Neurodevelopmental, Neuropsychiatric, and Neuro-Degenerative Diseases. LIFE (BASEL, SWITZERLAND) 2023; 13:life13010221. [PMID: 36676170 PMCID: PMC9866034 DOI: 10.3390/life13010221] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/06/2023] [Accepted: 01/11/2023] [Indexed: 01/15/2023]
Abstract
The vascular system of the prenatal brain is crucial for the development of the central nervous system. Communication between vessels and neural cells is bidirectional, and dysfunctional communication can lead to neurodevelopmental diseases. In the present review, we introduce neurodevelopmental and neuropsychiatric diseases potentially caused by disturbances in the neurovascular system and discuss candidate genes responsible for neurovascular system impairments. In contrast to diseases that can manifest during the developing stage, we have also summarized the disturbances of the neurovascular system in neurodegenerative diseases including Alzheimer's disease and Parkinson's disease. Furthermore, we discussed the role of abnormal vascularization and dysfunctional vessels in the development of neurovascular-related diseases.
Collapse
Affiliation(s)
- Keiichi Ishihara
- Department of Pathological Biochemistry, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan
- Correspondence: ; Tel.: +81-75-595-4656
| | - Kazuyuki Takata
- Division of Integrated Pharmaceutical Sciences, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan
| | - Ken-ichi Mizutani
- Laboratory of Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Kobe Gakuin University, Kobe 650-8586, Japan
| |
Collapse
|
12
|
Dixon TA, Muotri AR. Advancing preclinical models of psychiatric disorders with human brain organoid cultures. Mol Psychiatry 2023; 28:83-95. [PMID: 35948659 PMCID: PMC9812789 DOI: 10.1038/s41380-022-01708-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 07/05/2022] [Accepted: 07/07/2022] [Indexed: 01/11/2023]
Abstract
Psychiatric disorders are often distinguished from neurological disorders in that the former do not have characteristic lesions or findings from cerebrospinal fluid, electroencephalograms (EEGs), or brain imaging, and furthermore do not have commonly recognized convergent mechanisms. Psychiatric disorders commonly involve clinical diagnosis of phenotypic behavioral disturbances of mood and psychosis, often with a poorly understood contribution of environmental factors. As such, psychiatric disease has been challenging to model preclinically for mechanistic understanding and pharmaceutical development. This review compares commonly used animal paradigms of preclinical testing with evolving techniques of induced pluripotent cell culture with a focus on emerging three-dimensional models. Advances in complexity of 3D cultures, recapitulating electrical activity in utero, and disease modeling of psychosis, mood, and environmentally induced disorders are reviewed. Insights from these rapidly expanding technologies are discussed as they pertain to the utility of human organoid and other models in finding novel research directions, validating pharmaceutical action, and recapitulating human disease.
Collapse
Affiliation(s)
- Thomas Anthony Dixon
- grid.266100.30000 0001 2107 4242Department of Psychiatry, University of California San Diego, La Jolla, CA 92093 USA
| | - Alysson R. Muotri
- grid.266100.30000 0001 2107 4242Department of Pediatrics and Department of Cellular & Molecular Medicine, University of California San Diego, School of Medicine, Center for Academic Research and Training in Anthropogeny (CARTA), Kavli Institute for Brain and Mind, Archealization Center (ArchC), La Jolla, CA 92037 USA
| |
Collapse
|
13
|
Dubonyte U, Asenjo-Martinez A, Werge T, Lage K, Kirkeby A. Current advancements of modelling schizophrenia using patient-derived induced pluripotent stem cells. Acta Neuropathol Commun 2022; 10:183. [PMID: 36527106 PMCID: PMC9756764 DOI: 10.1186/s40478-022-01460-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 10/12/2022] [Indexed: 12/23/2022] Open
Abstract
Schizophrenia (SZ) is a severe psychiatric disorder, with a prevalence of 1-2% world-wide and substantial health- and social care costs. The pathology is influenced by both genetic and environmental factors, however the underlying cause still remains elusive. SZ has symptoms including delusions, hallucinations, confused thoughts, diminished emotional responses, social withdrawal and anhedonia. The onset of psychosis is usually in late adolescence or early adulthood. Multiple genome-wide association and whole exome sequencing studies have provided extraordinary insights into the genetic variants underlying familial as well as polygenic forms of the disease. Nonetheless, a major limitation in schizophrenia research remains the lack of clinically relevant animal models, which in turn hampers the development of novel effective therapies for the patients. The emergence of human induced pluripotent stem cell (hiPSC) technology has allowed researchers to work with SZ patient-derived neuronal and glial cell types in vitro and to investigate the molecular basis of the disorder in a human neuronal context. In this review, we summarise findings from available studies using hiPSC-based neural models and discuss how these have provided new insights into molecular and cellular pathways of SZ. Further, we highlight different examples of how these models have shown alterations in neurogenesis, neuronal maturation, neuronal connectivity and synaptic impairment as well as mitochondrial dysfunction and dysregulation of miRNAs in SZ patient-derived cultures compared to controls. We discuss the pros and cons of these models and describe the potential of using such models for deciphering the contribution of specific human neural cell types to the development of the disease.
Collapse
Affiliation(s)
- Ugne Dubonyte
- Department of Neuroscience and Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), University of Copenhagen, Copenhagen, Denmark
| | - Andrea Asenjo-Martinez
- Department of Neuroscience and Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), University of Copenhagen, Copenhagen, Denmark
| | - Thomas Werge
- Institute of Biological Psychiatry, Mental Health Services, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine and Lundbeck Foundation Center for GeoGenetics, GLOBE Institute, University of Copenhagen, Copenhagen, Denmark
| | - Kasper Lage
- Institute of Biological Psychiatry, Mental Health Services, Copenhagen University Hospital, Copenhagen, Denmark
- Stanley Center for Psychiatric Research and The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Agnete Kirkeby
- Department of Neuroscience and Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), University of Copenhagen, Copenhagen, Denmark.
- Department of Experimental Medical Science and Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden.
| |
Collapse
|
14
|
Chen H, Jin X, Li T, Ye Z. Brain organoids: Establishment and application. Front Cell Dev Biol 2022; 10:1029873. [PMID: 36506083 PMCID: PMC9726712 DOI: 10.3389/fcell.2022.1029873] [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: 08/28/2022] [Accepted: 11/10/2022] [Indexed: 11/24/2022] Open
Abstract
Brain organoids are produced by the differentiation of pluripotent stem cells under three-dimensional culture conditions by adding neurodevelopment-related regulatory signals. They are similar to the cell composition and anatomical structure of the brain, and can reflect the developmental process of the brain, as well as their physiology, pathology, and pharmacology. Brain organoids are good models to study human brain development and brain-related diseases in vitro. Here, we mainly focus on the construction of brain organoids and review the application of brain organoids in disease modelingand drug screening.
Collapse
Affiliation(s)
- Hao Chen
- Department of Neurovascular Surgery, First Hospital, Jilin University, Changchun, China
| | - Xin Jin
- Department of Oncology and Hematology, Second Hospital, Jilin University, Changchun, China
| | - Tie Li
- Department of Rheumatology, First Hospital, Jilin University, Changchun, China
| | - Zhuang Ye
- Department of Rheumatology, First Hospital, Jilin University, Changchun, China,*Correspondence: Zhuang Ye,
| |
Collapse
|
15
|
Alciati A, Reggiani A, Caldirola D, Perna G. Human-Induced Pluripotent Stem Cell Technology: Toward the Future of Personalized Psychiatry. J Pers Med 2022; 12:1340. [PMID: 36013289 PMCID: PMC9410334 DOI: 10.3390/jpm12081340] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/15/2022] [Accepted: 08/18/2022] [Indexed: 11/16/2022] Open
Abstract
The polygenic and multifactorial nature of many psychiatric disorders has hampered implementation of the personalized medicine approach in clinical practice. However, induced pluripotent stem cell (iPSC) technology has emerged as an innovative tool for patient-specific disease modeling to expand the pathophysiology knowledge and treatment perspectives in the last decade. Current technologies enable adult human somatic cell reprogramming into iPSCs to generate neural cells and direct neural cell conversion to model organisms that exhibit phenotypes close to human diseases, thereby effectively representing relevant aspects of neuropsychiatric disorders. In this regard, iPSCs reflect patient pathophysiology and pharmacological responsiveness, particularly when cultured under conditions that emulate spatial tissue organization in brain organoids. Recently, the application of iPSCs has been frequently associated with gene editing that targets the disease-causing gene to deepen the illness pathophysiology and to conduct drug screening. Moreover, gene editing has provided a unique opportunity to repair the putative causative genetic lesions in patient-derived cells. Here, we review the use of iPSC technology to model and potentially treat neuropsychiatric disorders by illustrating the key studies on a series of mental disorders, including schizophrenia, major depressive disorder, bipolar disorder, and autism spectrum disorder. Future perspectives will involve the development of organ-on-a-chip platforms that control the microenvironmental conditions so as to reflect individual pathophysiological by adjusting physiochemical parameters according to personal health data. This strategy could open new ways by which to build a disease model that considers individual variability and tailors personalized treatments.
Collapse
Affiliation(s)
- Alessandra Alciati
- Department of Clinical Neurosciences, Villa San Benedetto Menni—Hermanas Hospitalarias, Via Roma 16, 22032 Albese con Cassano, Como, Italy
- Humanitas Clinical and Research Center, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Angelo Reggiani
- D3 Validation Research Line, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Daniela Caldirola
- Department of Clinical Neurosciences, Villa San Benedetto Menni—Hermanas Hospitalarias, Via Roma 16, 22032 Albese con Cassano, Como, Italy
- Department of Biological Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20090 Milan, Italy
| | - Giampaolo Perna
- Department of Clinical Neurosciences, Villa San Benedetto Menni—Hermanas Hospitalarias, Via Roma 16, 22032 Albese con Cassano, Como, Italy
- Department of Biological Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20090 Milan, Italy
| |
Collapse
|
16
|
Whiteley JT, Fernandes S, Sharma A, Mendes APD, Racha V, Benassi SK, Marchetto MC. Reaching into the toolbox: Stem cell models to study neuropsychiatric disorders. Stem Cell Reports 2022; 17:187-210. [PMID: 35063127 PMCID: PMC8828548 DOI: 10.1016/j.stemcr.2021.12.015] [Citation(s) in RCA: 21] [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: 12/06/2020] [Revised: 12/17/2021] [Accepted: 12/20/2021] [Indexed: 12/26/2022] Open
Abstract
Recent advances in genetics, molecular biology, and stem cell biology have accelerated our understanding of neuropsychiatric disorders, like autism spectrum disorder (ASD), major depressive disorder (MDD), bipolar disorder (BD), and schizophrenia (SZ). This progress highlights the incredible complexity of both the human brain and mental illnesses from the biochemical to the cellular level. Contributing to the complexity of neuropsychiatric disorders are their polygenic nature, cellular and brain region interconnectivity, and dysregulation of human-specific neurodevelopmental processes. Here, we discuss available tools, including CRISPR-Cas9, and the applications of these tools to develop cell-based two-dimensional (2D) models and 3D brain organoid models that better represent and unravel the intricacies of neuropsychiatric disorder pathophysiology.
Collapse
Affiliation(s)
- Jack T Whiteley
- Laboratory of Genetics, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA; Doctoral Program in Neurobiology and Behavior, Department of Neuroscience, Columbia University, Jerome L. Greene Science Center, 3227 Broadway, L7-028, MC 9872, New York, NY 10027, USA
| | - Sarah Fernandes
- Laboratory of Genetics, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA; Department of Biological Sciences, University of California, San Diego, 9500 Gilman Dr, La Jolla, CA 92093, USA
| | - Amandeep Sharma
- Laboratory of Genetics, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Ana Paula D Mendes
- Laboratory of Genetics, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Vipula Racha
- Laboratory of Genetics, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Simone K Benassi
- Laboratory of Genetics, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Maria C Marchetto
- Laboratory of Genetics, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA; Department of Anthropology, University of California, San Diego, 9500 Gilman Dr, La Jolla, CA 92093, USA.
| |
Collapse
|
17
|
Page SC, Sripathy SR, Farinelli F, Ye Z, Wang Y, Hiler DJ, Pattie EA, Nguyen CV, Tippani M, Moses RL, Chen HY, Tran MN, Eagles NJ, Stolz JM, Catallini JL, Soudry OR, Dickinson D, Berman KF, Apud JA, Weinberger DR, Martinowich K, Jaffe AE, Straub RE, Maher BJ. Electrophysiological measures from human iPSC-derived neurons are associated with schizophrenia clinical status and predict individual cognitive performance. Proc Natl Acad Sci U S A 2022; 119:e2109395119. [PMID: 35017298 PMCID: PMC8784142 DOI: 10.1073/pnas.2109395119] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 11/10/2021] [Indexed: 12/11/2022] Open
Abstract
Neurons derived from human induced pluripotent stem cells (hiPSCs) have been used to model basic cellular aspects of neuropsychiatric disorders, but the relationship between the emergent phenotypes and the clinical characteristics of donor individuals has been unclear. We analyzed RNA expression and indices of cellular function in hiPSC-derived neural progenitors and cortical neurons generated from 13 individuals with high polygenic risk scores (PRSs) for schizophrenia (SCZ) and a clinical diagnosis of SCZ, along with 15 neurotypical individuals with low PRS. We identified electrophysiological measures in the patient-derived neurons that implicated altered Na+ channel function, action potential interspike interval, and gamma-aminobutyric acid-ergic neurotransmission. Importantly, electrophysiological measures predicted cardinal clinical and cognitive features found in these SCZ patients. The identification of basic neuronal physiological properties related to core clinical characteristics of illness is a potentially critical step in generating leads for novel therapeutics.
Collapse
Affiliation(s)
| | | | | | - Zengyou Ye
- Lieber Institute for Brain Development, Baltimore, MD 21205
| | - Yanhong Wang
- Lieber Institute for Brain Development, Baltimore, MD 21205
| | - Daniel J Hiler
- Lieber Institute for Brain Development, Baltimore, MD 21205
| | | | | | | | | | - Huei-Ying Chen
- Lieber Institute for Brain Development, Baltimore, MD 21205
| | - Matthew Nguyen Tran
- Lieber Institute for Brain Development, Baltimore, MD 21205
- McKusick-Nathans Institute, Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205
| | | | - Joshua M Stolz
- Lieber Institute for Brain Development, Baltimore, MD 21205
| | - Joseph L Catallini
- Lieber Institute for Brain Development, Baltimore, MD 21205
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205
| | | | - Dwight Dickinson
- Clinical and Translational Neuroscience Branch, National Institute of Mental Health Intramural Research Program, NIH, Bethesda, MD 20892
| | - Karen F Berman
- Clinical and Translational Neuroscience Branch, National Institute of Mental Health Intramural Research Program, NIH, Bethesda, MD 20892
| | - Jose A Apud
- Clinical and Translational Neuroscience Branch, National Institute of Mental Health Intramural Research Program, NIH, Bethesda, MD 20892
| | - Daniel R Weinberger
- Lieber Institute for Brain Development, Baltimore, MD 21205
- McKusick-Nathans Institute, Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21205
| | - Keri Martinowich
- Lieber Institute for Brain Development, Baltimore, MD 21205
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205
| | - Andrew E Jaffe
- Lieber Institute for Brain Development, Baltimore, MD 21205
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205
| | | | - Brady J Maher
- Lieber Institute for Brain Development, Baltimore, MD 21205;
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205
| |
Collapse
|
18
|
Golimbet V, Kostyuk G. Genotype — phenotype relationships in view of recent advances in the understanding of genetic causes of schizophrenia. Zh Nevrol Psikhiatr Im S S Korsakova 2022; 122:20-25. [DOI: 10.17116/jnevro202212201220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
19
|
Saia-Cereda VM. Modulating Specific Pathways In Vitro to Understand the Synaptic Dysfunction of Schizophrenia. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1400:121-127. [DOI: 10.1007/978-3-030-97182-3_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
20
|
Räsänen N, Tiihonen J, Koskuvi M, Lehtonen Š, Koistinaho J. The iPSC perspective on schizophrenia. Trends Neurosci 2021; 45:8-26. [PMID: 34876311 DOI: 10.1016/j.tins.2021.11.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 09/29/2021] [Accepted: 11/10/2021] [Indexed: 12/17/2022]
Abstract
Over a decade of schizophrenia research using human induced pluripotent stem cell (iPSC)-derived neural models has provided substantial data describing neurobiological characteristics of the disorder in vitro. Simultaneously, translation of the results into general mechanistic concepts underlying schizophrenia pathophysiology has been trailing behind. Given that modeling brain function using cell cultures is challenging, the gap between the in vitro models and schizophrenia as a clinical disorder has remained wide. In this review, we highlight reproducible findings and emerging trends in recent schizophrenia-related iPSC studies. We illuminate the relevance of the results in the context of human brain development, with a focus on processes coinciding with critical developmental periods for schizophrenia.
Collapse
Affiliation(s)
- Noora Räsänen
- Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Jari Tiihonen
- Neuroscience Center, University of Helsinki, Helsinki, Finland; Department of Clinical Neuroscience, Karolinska Institutet, Solna, Sweden; Center for Psychiatric Research, Stockholm City Council, Stockholm, Sweden; Department of Forensic Psychiatry, University of Eastern Finland, Niuvanniemi Hospital, Kuopio, Finland
| | - Marja Koskuvi
- Neuroscience Center, University of Helsinki, Helsinki, Finland; A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Šárka Lehtonen
- Neuroscience Center, University of Helsinki, Helsinki, Finland; A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Jari Koistinaho
- Neuroscience Center, University of Helsinki, Helsinki, Finland; A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
21
|
Chandrasekaran A, Jensen P, Mohamed FA, Lancaster M, Benros ME, Larsen MR, Freude KK. A protein-centric view of in vitro biological model systems for schizophrenia. Stem Cells 2021; 39:1569-1578. [PMID: 34431581 DOI: 10.1002/stem.3447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 08/10/2021] [Indexed: 01/10/2023]
Abstract
Schizophrenia (SCZ) is a severe brain disorder, characterized by psychotic, negative, and cognitive symptoms, affecting 1% of the population worldwide. The precise etiology of SCZ is still unknown; however, SCZ has a high heritability and is associated with genetic, environmental, and social risk factors. Even though the genetic contribution is indisputable, the discrepancies between transcriptomics and proteomics in brain tissues are consistently challenging the field to decipher the disease pathology. Here we provide an overview of the state of the art of neuronal two-dimensional and three-dimensional model systems that can be combined with proteomics analyses to decipher specific brain pathology and detection of alternative entry points for drug development.
Collapse
Affiliation(s)
- Abinaya Chandrasekaran
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Pia Jensen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Fadumo A Mohamed
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Madeline Lancaster
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, UK
| | - Michael E Benros
- Biological and Precision Psychiatry, Copenhagen Research Centre for Mental Health, Mental Health Centre Copenhagen, Copenhagen University Hospital, Hellerup, Denmark
- Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N, Denmark
| | - Martin R Larsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Kristine K Freude
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
22
|
Abashkin DA, Kurishev AO, Karpov DS, Golimbet VE. Cellular Models in Schizophrenia Research. Int J Mol Sci 2021; 22:ijms22168518. [PMID: 34445221 PMCID: PMC8395162 DOI: 10.3390/ijms22168518] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/04/2021] [Accepted: 08/05/2021] [Indexed: 12/11/2022] Open
Abstract
Schizophrenia (SZ) is a prevalent functional psychosis characterized by clinical behavioural symptoms and underlying abnormalities in brain function. Genome-wide association studies (GWAS) of schizophrenia have revealed many loci that do not directly identify processes disturbed in the disease. For this reason, the development of cellular models containing SZ-associated variations has become a focus in the post-GWAS research era. The application of revolutionary clustered regularly interspaced palindromic repeats CRISPR/Cas9 gene-editing tools, along with recently developed technologies for cultivating brain organoids in vitro, have opened new perspectives for the construction of these models. In general, cellular models are intended to unravel particular biological phenomena. They can provide the missing link between schizophrenia-related phenotypic features (such as transcriptional dysregulation, oxidative stress and synaptic dysregulation) and data from pathomorphological, electrophysiological and behavioural studies. The objectives of this review are the systematization and classification of cellular models of schizophrenia, based on their complexity and validity for understanding schizophrenia-related phenotypes.
Collapse
Affiliation(s)
- Dmitrii A. Abashkin
- Mental Health Research Center, Clinical Genetics Laboratory, Kashirskoe Sh. 34, 115522 Moscow, Russia; (D.A.A.); (A.O.K.); (D.S.K.)
| | - Artemii O. Kurishev
- Mental Health Research Center, Clinical Genetics Laboratory, Kashirskoe Sh. 34, 115522 Moscow, Russia; (D.A.A.); (A.O.K.); (D.S.K.)
| | - Dmitry S. Karpov
- Mental Health Research Center, Clinical Genetics Laboratory, Kashirskoe Sh. 34, 115522 Moscow, Russia; (D.A.A.); (A.O.K.); (D.S.K.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Str. 32, 119991 Moscow, Russia
| | - Vera E. Golimbet
- Mental Health Research Center, Clinical Genetics Laboratory, Kashirskoe Sh. 34, 115522 Moscow, Russia; (D.A.A.); (A.O.K.); (D.S.K.)
- Correspondence:
| |
Collapse
|
23
|
Tomaskovic-Crook E, Guerrieri-Cortesi K, Crook JM. Induced pluripotent stem cells for 2D and 3D modelling the biological basis of schizophrenia and screening possible therapeutics. Brain Res Bull 2021; 175:48-62. [PMID: 34273422 DOI: 10.1016/j.brainresbull.2021.07.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 07/05/2021] [Accepted: 07/08/2021] [Indexed: 12/22/2022]
Abstract
Induced pluripotent stem cells (iPSCs) are providing unprecedented insight into complex neuropsychiatric disorders such as schizophrenia (SZ). Here we review the use of iPSCs for investigating the etiopathology and treatment of SZ, beginning with conventional in vitro two-dimensional (2D; monolayer) cell modelling, through to more advanced 3D tissue studies. With the advent of 3D modelling, utilising advanced differentiation paradigms and additive manufacturing technologies, inclusive of patient-specific cerebral/neural organoids and bioprinted neural tissues, such live disease-relevant tissue systems better recapitulate "within-body" tissue function and pathobiology. We posit that by enabling better understanding of biological causality, these evolving strategies will yield novel therapeutic targets and accordingly, drug candidates.
Collapse
Affiliation(s)
- Eva Tomaskovic-Crook
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, University of Wollongong, 2500, Wollongong, Australia; Illawarra Health and Medical Research Institute, University of Wollongong, 2500, Wollongong, Australia.
| | - Kyle Guerrieri-Cortesi
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, University of Wollongong, 2500, Wollongong, Australia
| | - Jeremy Micah Crook
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, University of Wollongong, 2500, Wollongong, Australia; Illawarra Health and Medical Research Institute, University of Wollongong, 2500, Wollongong, Australia; Chris O'Brien Lifehouse Hospital, Camperdown, NSW, 2050, Australia; Department of Surgery, St Vincent's Hospital, The University of Melbourne, 3065, Fitzroy, Australia.
| |
Collapse
|
24
|
Bodnar B, Zhang Y, Liu J, Lin Y, Wang P, Wei Z, Saribas S, Zhu Y, Li F, Wang X, Yang W, Li Q, Ho WZ, Hu W. Novel Scalable and Simplified System to Generate Microglia-Containing Cerebral Organoids From Human Induced Pluripotent Stem Cells. Front Cell Neurosci 2021; 15:682272. [PMID: 34290591 PMCID: PMC8288463 DOI: 10.3389/fncel.2021.682272] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 05/07/2021] [Indexed: 12/18/2022] Open
Abstract
Human cerebral organoid (CO) is a three-dimensional (3D) cell culture system that recapitulates the developing human brain. While CO has proved an invaluable tool for studying neurological disorders in a more clinically relevant matter, there have still been several shortcomings including CO variability and reproducibility as well as lack of or underrepresentation of certain cell types typically found in the brain. As the technology to generate COs has continued to improve, more efficient and streamlined protocols have addressed some of these issues. Here we present a novel scalable and simplified system to generate microglia-containing CO (MCO). We characterize the cell types and dynamic development of MCOs and validate that these MCOs harbor microglia, astrocytes, neurons, and neural stem/progenitor cells, maturing in a manner that reflects human brain development. We introduce a novel technique for the generation of embryoid bodies (EBs) directly from induced pluripotent stem cells (iPSCs) that involves simplified steps of transitioning directly from 3D cultures as well as orbital shaking culture in a standard 6-well culture plate. This allows for the generation of MCOs with an easy-to-use system that is affordable and accessible by any general lab.
Collapse
Affiliation(s)
- Brittany Bodnar
- Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
- Center for Metabolic Disease Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Yongang Zhang
- Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
- Center for Metabolic Disease Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
- Center for Stem Cell Research and Application, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS and PUMC), Chengdu, China
| | - Jinbiao Liu
- Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Yuan Lin
- Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
- Center for Metabolic Disease Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Peng Wang
- Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Zhengyu Wei
- Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
- Center for Metabolic Disease Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Sami Saribas
- Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
- Center for Metabolic Disease Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Yuanjun Zhu
- Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
- Center for Metabolic Disease Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Fang Li
- Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
- Center for Metabolic Disease Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Xu Wang
- Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Wenli Yang
- Institute for Regenerative Medicine and Department of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Qingsheng Li
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Wen-Zhe Ho
- Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Wenhui Hu
- Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
- Center for Metabolic Disease Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| |
Collapse
|
25
|
Michael Deans PJ, Brennand KJ. Applying stem cells and CRISPR engineering to uncover the etiology of schizophrenia. Curr Opin Neurobiol 2021; 69:193-201. [PMID: 34010781 DOI: 10.1016/j.conb.2021.04.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/06/2021] [Accepted: 04/12/2021] [Indexed: 12/13/2022]
Abstract
Schizophrenia is a highly heritable, polygenic disorder. A growing list of common genetic variants have been associated with schizophrenia; there is a clear need to understand the role of these risk factors in the etiology of disease. The majority of these variants occur in noncoding regions of the genome and are thought to regulate the expression of one or more genes in a cell type-specific fashion. Recent advances in stem cell biology and molecular genetics have resulted in two invaluable advances: hiPSC technology makes possible the generation of donor-specific disease-relevant neural cell types, whereas CRISPR-based techniques can be applied to manipulate individual variants and/or their gene targets. New multiplexed gene manipulation and CRISPR screening techniques show great promise toward dissecting the complex interactions between the myriad disease-associated variants. This review outlines key advances in hiPSC and CRISPR technology, describing their applications and future potential in the field of schizophrenia research.
Collapse
Affiliation(s)
- Peter James Michael Deans
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomics, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kristen J Brennand
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomics, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
26
|
Imbalance of Excitatory/Inhibitory Neuron Differentiation in Neurodevelopmental Disorders with an NR2F1 Point Mutation. Cell Rep 2021; 31:107521. [PMID: 32320667 DOI: 10.1016/j.celrep.2020.03.085] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 12/13/2019] [Accepted: 03/24/2020] [Indexed: 12/27/2022] Open
Abstract
Recent studies have revealed an essential role for embryonic cortical development in the pathophysiology of neurodevelopmental disorders, including autism spectrum disorder (ASD). However, the genetic basis and underlying mechanisms remain unclear. Here, we generate mutant human embryonic stem cell lines (Mut hESCs) carrying an NR2F1-R112K mutation that has been identified in a patient with ASD features and investigate their neurodevelopmental alterations. Mut hESCs overproduce ventral telencephalic neuron progenitors (ventral NPCs) and underproduce dorsal NPCs, causing the imbalance of excitatory/inhibitory neurons. These alterations can be mainly attributed to the aberrantly activated Hedgehog signaling pathway. Moreover, the corresponding Nr2f1 point-mutant mice display a similar excitatory/inhibitory neuron imbalance and abnormal behaviors. Antagonizing the increased inhibitory synaptic transmission partially alleviates their behavioral deficits. Together, our results suggest that the NR2F1-dependent imbalance of excitatory/inhibitory neuron differentiation caused by the activated Hedgehog pathway is one precursor of neurodevelopmental disorders and may enlighten the therapeutic approaches.
Collapse
|
27
|
Liput M, Magliaro C, Kuczynska Z, Zayat V, Ahluwalia A, Buzanska L. Tools and approaches for analyzing the role of mitochondria in health, development and disease using human cerebral organoids. Dev Neurobiol 2021; 81:591-607. [PMID: 33725382 DOI: 10.1002/dneu.22818] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 03/08/2021] [Accepted: 03/09/2021] [Indexed: 12/16/2022]
Abstract
Mitochondria are cellular organelles involved in generating energy to power various processes in the cell. Although the pivotal role of mitochondria in neurogenesis was demonstrated (first in animal models), very little is known about their role in human embryonic neurodevelopment and its pathology. In this respect human-induced pluripotent stem cells (hiPSC)-derived cerebral organoids provide a tractable, alternative model system of the early neural development and disease that is responsive to pharmacological and genetic manipulations, not possible to apply in humans. Although the involvement of mitochondria in the pathogenesis and progression of neurodegenerative diseases and brain dysfunction has been demonstrated, the precise role they play in cell life and death remains unknown, compromising the development of new mitochondria-targeted approaches to treat human diseases. The cerebral organoid model of neurogenesis and disease in vitro provides an unprecedented opportunity to answer some of the most fundamental questions about mitochondrial function in early human neurodevelopment and neural pathology. Largely an unexplored territory due to the lack of tools and approaches, this review focuses on recent technological advancements in fluorescent and molecular tools, imaging systems, and computational approaches for quantitative and qualitative analyses of mitochondrial structure and function in three-dimensional cellular assemblies-cerebral organoids. Future developments in this direction will further facilitate our understanding of the important role or mitochondrial dynamics and energy requirements during early embryonic development. This in turn will provide a further understanding of how dysfunctional mitochondria contribute to disease processes.
Collapse
Affiliation(s)
- Michał Liput
- Department of Stem Cell Bioengineering, Mossakowski Medical Research Institute Polish Academy of Sciences, Warsaw, Poland
| | - Chiara Magliaro
- Research Centre "E. Piaggio", and Department of Information Engineering, University of Pisa, Pisa, Italy
| | - Zuzanna Kuczynska
- Department of Stem Cell Bioengineering, Mossakowski Medical Research Institute Polish Academy of Sciences, Warsaw, Poland
| | - Valery Zayat
- Department of Stem Cell Bioengineering, Mossakowski Medical Research Institute Polish Academy of Sciences, Warsaw, Poland
| | - Arti Ahluwalia
- Research Centre "E. Piaggio", and Department of Information Engineering, University of Pisa, Pisa, Italy
| | - Leonora Buzanska
- Department of Stem Cell Bioengineering, Mossakowski Medical Research Institute Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
28
|
Dell' Amico C, Tata A, Pellegrino E, Onorati M, Conti L. Genome editing in stem cells for genetic neurodisorders. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2021; 182:403-438. [PMID: 34175049 DOI: 10.1016/bs.pmbts.2020.12.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The recent advent of genome editing techniques and their rapid improvement paved the way in establishing innovative human neurological disease models and in developing new therapeutic opportunities. Human pluripotent (both induced or naive) stem cells and neural stem cells represent versatile tools to be applied to multiple research needs and, together with genomic snip and fix tools, have recently made possible the creation of unique platforms to directly investigate several human neural affections. In this chapter, we will discuss genome engineering tools, and their recent improvements, applied to the stem cell field, focusing on how these two technologies may be pivotal instruments to deeply unravel molecular mechanisms underlying development and function, as well as disorders, of the human brain. We will review how these frontier technologies may be exploited to investigate or treat severe neurodevelopmental disorders, such as microcephaly, autism spectrum disorder, schizophrenia, as well as neurodegenerative conditions, including Parkinson's disease, Huntington's disease, Alzheimer's disease, and spinal muscular atrophy.
Collapse
Affiliation(s)
- Claudia Dell' Amico
- Unit of Cell and Developmental Biology, Department of Biology, University of Pisa, Pisa, Italy
| | - Alice Tata
- Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy
| | - Enrica Pellegrino
- Unit of Cell and Developmental Biology, Department of Biology, University of Pisa, Pisa, Italy; Host-Pathogen Interactions in Tuberculosis Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Marco Onorati
- Unit of Cell and Developmental Biology, Department of Biology, University of Pisa, Pisa, Italy.
| | - Luciano Conti
- Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy.
| |
Collapse
|
29
|
Szabó E, Juhász F, Hathy E, Reé D, Homolya L, Erdei Z, Réthelyi JM, Apáti Á. Functional Comparison of Blood-Derived Human Neural Progenitor Cells. Int J Mol Sci 2020; 21:E9118. [PMID: 33266139 PMCID: PMC7730078 DOI: 10.3390/ijms21239118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 11/21/2020] [Accepted: 11/26/2020] [Indexed: 12/16/2022] Open
Abstract
Induced pluripotent stem cell (iPSC)-derived neural progenitor cells (NPCs) are promising tools to model complex neurological or psychiatric diseases, including schizophrenia. Multiple studies have compared patient-derived and healthy control NPCs derived from iPSCs in order to investigate cellular phenotypes of this disease, although the establishment, stabilization, and directed differentiation of iPSC lines are rather expensive and time-demanding. However, interrupted reprogramming by omitting the stabilization of iPSCs may allow for the generation of a plastic stage of the cells and thus provide a shortcut to derive NPSCs directly from tissue samples. Here, we demonstrate a method to generate shortcut NPCs (sNPCs) from blood mononuclear cells and present a detailed comparison of these sNPCs with NPCs obtained from the same blood samples through stable iPSC clones and a subsequent neural differentiation (classical NPCs-cNPCs). Peripheral blood cells were obtained from a schizophrenia patient and his two healthy parents (a case-parent trio), while a further umbilical cord blood sample was obtained from the cord of a healthy new-born. The expression of stage-specific markers in sNPCs and cNPCs were compared both at the protein and RNA levels. We also performed functional tests to investigate Wnt and glutamate signaling and the oxidative stress, as these pathways have been suggested to play important roles in the pathophysiology of schizophrenia. We found similar responses in the two types of NPCs, suggesting that the shortcut procedure provides sNPCs, allowing an efficient screening of disease-related phenotypes.
Collapse
Affiliation(s)
- Eszter Szabó
- Institute of Enzymology, Research Centre for Natural Sciences, 1117 Budapest, Hungary; (E.S.); (F.J.); (D.R.); (L.H.); (Z.E.)
| | - Flóra Juhász
- Institute of Enzymology, Research Centre for Natural Sciences, 1117 Budapest, Hungary; (E.S.); (F.J.); (D.R.); (L.H.); (Z.E.)
| | - Edit Hathy
- Department of Psychiatry and Psychotherapy, Faculty of Medicine, Semmelweis University, 1083 Budapest, Hungary;
- National Brain Research Project (NAP) Molecular Psychiatry Research Group, Hungarian Academy of Sciences and Faculty of Medicine, Semmelweis University, 1083 Budapest, Hungary
| | - Dóra Reé
- Institute of Enzymology, Research Centre for Natural Sciences, 1117 Budapest, Hungary; (E.S.); (F.J.); (D.R.); (L.H.); (Z.E.)
| | - László Homolya
- Institute of Enzymology, Research Centre for Natural Sciences, 1117 Budapest, Hungary; (E.S.); (F.J.); (D.R.); (L.H.); (Z.E.)
| | - Zsuzsa Erdei
- Institute of Enzymology, Research Centre for Natural Sciences, 1117 Budapest, Hungary; (E.S.); (F.J.); (D.R.); (L.H.); (Z.E.)
| | - János M. Réthelyi
- Department of Psychiatry and Psychotherapy, Faculty of Medicine, Semmelweis University, 1083 Budapest, Hungary;
- National Brain Research Project (NAP) Molecular Psychiatry Research Group, Hungarian Academy of Sciences and Faculty of Medicine, Semmelweis University, 1083 Budapest, Hungary
| | - Ágota Apáti
- Institute of Enzymology, Research Centre for Natural Sciences, 1117 Budapest, Hungary; (E.S.); (F.J.); (D.R.); (L.H.); (Z.E.)
| |
Collapse
|
30
|
iPSC-derived homogeneous populations of developing schizophrenia cortical interneurons have compromised mitochondrial function. Mol Psychiatry 2020; 25:2873-2888. [PMID: 31019265 PMCID: PMC6813882 DOI: 10.1038/s41380-019-0423-3] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 03/23/2019] [Accepted: 04/03/2019] [Indexed: 02/05/2023]
Abstract
Schizophrenia (SCZ) is a neurodevelopmental disorder. Thus, studying pathogenetic mechanisms underlying SCZ requires studying the development of brain cells. Cortical interneurons (cINs) are consistently observed to be abnormal in SCZ postmortem brains. These abnormalities may explain altered gamma oscillation and cognitive function in patients with SCZ. Of note, currently used antipsychotic drugs ameliorate psychosis, but they are not very effective in reversing cognitive deficits. Characterizing mechanisms of SCZ pathogenesis, especially related to cognitive deficits, may lead to improved treatments. We generated homogeneous populations of developing cINs from 15 healthy control (HC) iPSC lines and 15 SCZ iPSC lines. SCZ cINs, but not SCZ glutamatergic neurons, show dysregulated Oxidative Phosphorylation (OxPhos) related gene expression, accompanied by compromised mitochondrial function. The OxPhos deficit in cINs could be reversed by Alpha Lipoic Acid/Acetyl-L-Carnitine (ALA/ALC) but not by other chemicals previously identified as increasing mitochondrial function. The restoration of mitochondrial function by ALA/ALC was accompanied by a reversal of arborization deficits in SCZ cINs. OxPhos abnormality, even in the absence of any circuit environment with other neuronal subtypes, appears to be an intrinsic deficit in SCZ cINs.
Collapse
|
31
|
McNeill RV, Ziegler GC, Radtke F, Nieberler M, Lesch KP, Kittel-Schneider S. Mental health dished up-the use of iPSC models in neuropsychiatric research. J Neural Transm (Vienna) 2020; 127:1547-1568. [PMID: 32377792 PMCID: PMC7578166 DOI: 10.1007/s00702-020-02197-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 04/20/2020] [Indexed: 12/13/2022]
Abstract
Genetic and molecular mechanisms that play a causal role in mental illnesses are challenging to elucidate, particularly as there is a lack of relevant in vitro and in vivo models. However, the advent of induced pluripotent stem cell (iPSC) technology has provided researchers with a novel toolbox. We conducted a systematic review using the PRISMA statement. A PubMed and Web of Science online search was performed (studies published between 2006-2020) using the following search strategy: hiPSC OR iPSC OR iPS OR stem cells AND schizophrenia disorder OR personality disorder OR antisocial personality disorder OR psychopathy OR bipolar disorder OR major depressive disorder OR obsessive compulsive disorder OR anxiety disorder OR substance use disorder OR alcohol use disorder OR nicotine use disorder OR opioid use disorder OR eating disorder OR anorexia nervosa OR attention-deficit/hyperactivity disorder OR gaming disorder. Using the above search criteria, a total of 3515 studies were found. After screening, a final total of 56 studies were deemed eligible for inclusion in our study. Using iPSC technology, psychiatric disease can be studied in the context of a patient's own unique genetic background. This has allowed great strides to be made into uncovering the etiology of psychiatric disease, as well as providing a unique paradigm for drug testing. However, there is a lack of data for certain psychiatric disorders and several limitations to present iPSC-based studies, leading us to discuss how this field may progress in the next years to increase its utility in the battle to understand psychiatric disease.
Collapse
Affiliation(s)
- Rhiannon V McNeill
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, University of Würzburg, Margarete-Höppel-Platz 1, 97080, Würzburg, Germany
| | - Georg C Ziegler
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, University of Würzburg, Margarete-Höppel-Platz 1, 97080, Würzburg, Germany
| | - Franziska Radtke
- Department of Child and Adolescent Psychiatry, Psychosomatic Medicine and Psychotherapy University Hospital, University of Würzburg, Würzburg, Germany
| | - Matthias Nieberler
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, University of Würzburg, Margarete-Höppel-Platz 1, 97080, Würzburg, Germany
| | - Klaus-Peter Lesch
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, University of Würzburg, Margarete-Höppel-Platz 1, 97080, Würzburg, Germany
| | - Sarah Kittel-Schneider
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, University of Würzburg, Margarete-Höppel-Platz 1, 97080, Würzburg, Germany.
| |
Collapse
|
32
|
Identification of Molecular Signatures in Neural Differentiation and Neurological Diseases Using Digital Color-Coded Molecular Barcoding. Stem Cells Int 2020; 2020:8852313. [PMID: 33005195 PMCID: PMC7503121 DOI: 10.1155/2020/8852313] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 08/26/2020] [Accepted: 08/28/2020] [Indexed: 12/04/2022] Open
Abstract
Human pluripotent stem cells (PSCs), including embryonic stem cells and induced pluripotent stem cells, represent powerful tools for disease modeling and for therapeutic applications. PSCs are particularly useful for the study of development and diseases of the nervous system. However, generating in vitro models that recapitulate the architecture and the full variety of subtypes of cells that make the complexity of our brain remains a challenge. In order to fully exploit the potential of PSCs, advanced methods that facilitate the identification of molecular signatures in neural differentiation and neurological diseases are highly demanded. Here, we review the literature on the development and application of digital color-coded molecular barcoding as a potential tool for standardizing PSC research and applications in neuroscience. We will also describe relevant examples of the use of this technique for the characterization of the heterogeneous composition of the brain tumor glioblastoma multiforme.
Collapse
|
33
|
Fu X, Zhang G, Liu Y, Zhang L, Zhang F, Zhou C. Altered expression of the DISC1 gene in peripheral blood of patients with schizophrenia. BMC MEDICAL GENETICS 2020; 21:194. [PMID: 33008326 PMCID: PMC7532617 DOI: 10.1186/s12881-020-01132-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Accepted: 09/24/2020] [Indexed: 11/17/2022]
Abstract
Background Schizophrenia is a severe, heritable, and refractory psychiatric disorder. Several studies have shown that the disrupted in schizophrenia 1 (DISC1) gene is closely associated with schizophrenia by its role in neuronal morphology, synaptic function, brain development, and dopamine homeostasis etc. This study intended to investigate the expression levels of DISC1 gene in schizophrenia patients compared with healthy controls, and the expression variation of DISC1 gene before and after antipsychotic treatment in schizophrenia patients. Methods In this study, we compared DISC1 expression levels in blood of 48 healthy controls, and 32 schizophrenia patients before and after 12 weeks of antipsychotic treatment using real-time quantitative PCR (RT-qPCR) analysis. Results The expression levels of DISC1 gene in peripheral blood mononuclear cells of schizophrenia patients before antipsychotic treatment were higher than those in healthy controls (P < 0.01); whereas after antipsychotic treatment, the expression levels of DISC1 gene in peripheral blood mononuclear cells of schizophrenia patients still remained increased (P < 0.01). Conclusions Our study provided further support for the involvement of DISC1 in the development of schizophrenia.
Collapse
Affiliation(s)
- Xiaoqian Fu
- Department of Clinical Psychology, Suzhou Guangji Hospital, The Affiliated Guangji Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Guofu Zhang
- Wuxi Mental Health Center of Nanjing Medical University, 156 Qianrong Road, Wuxi, China.
| | - Yansong Liu
- Department of Clinical Psychology, Suzhou Guangji Hospital, The Affiliated Guangji Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Ling Zhang
- Department of Clinical Psychology, Suzhou Guangji Hospital, The Affiliated Guangji Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Fuquan Zhang
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, 264 Guangzhou Road, Nanjing, Jiangsu Province, China.
| | - Conghua Zhou
- School of Computer Science and Telecommunication Engineering, Jiangsu University, Zhenjiang, China.
| |
Collapse
|
34
|
Integrating CRISPR Engineering and hiPSC-Derived 2D Disease Modeling Systems. J Neurosci 2020; 40:1176-1185. [PMID: 32024766 DOI: 10.1523/jneurosci.0518-19.2019] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 10/23/2019] [Accepted: 10/23/2019] [Indexed: 12/20/2022] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) have revolutionized research on human diseases, particularly neurodegenerative and psychiatric disorders, making it possible to study mechanisms of disease risk and initiation in otherwise inaccessible patient-specific cells. Today, the integration of CRISPR engineering approaches with hiPSC-based models permits precise isogenic comparisons of human neurons and glia. This review is intended as a guideline for neuroscientists and clinicians interested in translating their research to hiPSC-based studies. It offers state-of-the-art approaches to tackling the challenges that are unique to human in vitro disease models, particularly interdonor and intradonor variability, and limitations in neuronal maturity and circuit complexity. Finally, we provide a detailed overview of the immense possibilities the field has to offer, highlighting efficient neural differentiation and induction strategies for the major brain cell types and providing perspective into integrating CRISPR-based methods into study design. The combination of hiPSC-based disease modeling, CRISPR technology, and high-throughput approaches promises to advance our scientific knowledge and accelerate progress in drug discovery.Dual Perspectives Companion Paper: Studying Human Neurodevelopment and Diseases Using 3D Brain Organoids, by Ai Tian, Julien Muffat, and Yun Li.
Collapse
|
35
|
Evgrafov OV, Armoskus C, Wrobel BB, Spitsyna VN, Souaiaia T, Herstein JS, Walker CP, Nguyen JD, Camarena A, Weitz JR, Kim JMH, Lopez Duarte E, Wang K, Simpson GM, Sobell JL, Medeiros H, Pato MT, Pato CN, Knowles JA. Gene Expression in Patient-Derived Neural Progenitors Implicates WNT5A Signaling in the Etiology of Schizophrenia. Biol Psychiatry 2020; 88:236-247. [PMID: 32143829 PMCID: PMC10947993 DOI: 10.1016/j.biopsych.2020.01.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 01/03/2020] [Accepted: 01/06/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Genome-wide association studies of schizophrenia have demonstrated that variations in noncoding regions are responsible for most of the common variation heritability of the disease. It is hypothesized that these risk variants alter gene expression. Therefore, studying alterations in gene expression in schizophrenia may provide a direct approach to understanding the etiology of the disease. In this study we use cultured neural progenitor cells derived from olfactory neuroepithelium (CNON cells) as a genetically unaltered cellular model to elucidate the neurodevelopmental aspects of schizophrenia. METHODS We performed a gene expression study using RNA sequencing of CNON cells from 111 control subjects and 144 individuals with schizophrenia. Differentially expressed genes were identified with DESeq2 software, using covariates to correct for sex, age, library batches, and 1 surrogate variable component. RESULTS A total of 80 genes were differentially expressed (false discovery rate < 10%), showing enrichment in cell migration, cell adhesion, developmental process, synapse assembly, cell proliferation, and related Gene Ontology categories. Cadherin and Wnt signaling pathways were positive in overrepresentation test, and, in addition, many genes were specifically involved in WNT5A signaling. The differentially expressed genes were modestly, but significantly, enriched in the genes overlapping single nucleotide polymorphisms with genome-wide significant association from the Psychiatric Genomics Consortium genome-wide association study of schizophrenia. We also found substantial overlap with genes associated with other psychiatric disorders or brain development, enrichment in the same Gene Ontology categories as genes with mutations de novo in schizophrenia, and studies of induced pluripotent stem cell-derived neural progenitor cells. CONCLUSIONS CNON cells are a good model of the neurodevelopmental aspects of schizophrenia and can be used to elucidate the etiology of the disorder.
Collapse
Affiliation(s)
- Oleg V Evgrafov
- College of Medicine, SUNY Downstate Health Sciences University, Brooklyn, New York.
| | - Chris Armoskus
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Bozena B Wrobel
- Caruso Department of Otolaryngology, Head and Neck Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Valeria N Spitsyna
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Tade Souaiaia
- College of Medicine, SUNY Downstate Health Sciences University, Brooklyn, New York
| | - Jennifer S Herstein
- Department of Psychiatry and the Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Christopher P Walker
- Department of Psychiatry and the Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Joseph D Nguyen
- Department of Psychiatry and the Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Adrian Camarena
- Department of Psychiatry and the Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Jonathan R Weitz
- Department of Psychiatry and the Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Jae Mun Hugo Kim
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Edder Lopez Duarte
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Kai Wang
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania; Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - George M Simpson
- Department of Psychiatry and the Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Janet L Sobell
- Department of Psychiatry and the Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Helena Medeiros
- College of Medicine, SUNY Downstate Health Sciences University, Brooklyn, New York
| | - Michele T Pato
- College of Medicine, SUNY Downstate Health Sciences University, Brooklyn, New York
| | - Carlos N Pato
- College of Medicine, SUNY Downstate Health Sciences University, Brooklyn, New York
| | - James A Knowles
- College of Medicine, SUNY Downstate Health Sciences University, Brooklyn, New York
| |
Collapse
|
36
|
Zhang S, Zhang H, Zhou Y, Qiao M, Zhao S, Kozlova A, Shi J, Sanders AR, Wang G, Luo K, Sengupta S, West S, Qian S, Streit M, Avramopoulos D, Cowan CA, Chen M, Pang ZP, Gejman PV, He X, Duan J. Allele-specific open chromatin in human iPSC neurons elucidates functional disease variants. Science 2020; 369:561-565. [PMID: 32732423 PMCID: PMC7773145 DOI: 10.1126/science.aay3983] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 09/16/2019] [Accepted: 06/01/2020] [Indexed: 12/12/2022]
Abstract
Most neuropsychiatric disease risk variants are in noncoding sequences and lack functional interpretation. Because regulatory sequences often reside in open chromatin, we reasoned that neuropsychiatric disease risk variants may affect chromatin accessibility during neurodevelopment. Using human induced pluripotent stem cell (iPSC)-derived neurons that model developing brains, we identified thousands of genetic variants exhibiting allele-specific open chromatin (ASoC). These neuronal ASoCs were partially driven by altered transcription factor binding, overrepresented in brain gene enhancers and expression quantitative trait loci, and frequently associated with distal genes through chromatin contacts. ASoCs were enriched for genetic variants associated with brain disorders, enabling identification of functional schizophrenia risk variants and their cis-target genes. This study highlights ASoC as a functional mechanism of noncoding neuropsychiatric risk variants, providing a powerful framework for identifying disease causal variants and genes.
Collapse
Affiliation(s)
- Siwei Zhang
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA
| | - Hanwen Zhang
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA
| | - Yifan Zhou
- Department of Human Genetics, University of Chicago, Chicago, IL 60637, USA
- The Graduate Program in Biophysical Sciences, University of Chicago, Chicago, IL 60637, USA
| | - Min Qiao
- Department of Human Genetics, University of Chicago, Chicago, IL 60637, USA
- Department of Biostatistics and Data Science, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Siming Zhao
- Department of Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Alena Kozlova
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA
| | - Jianxin Shi
- Biostatistics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20892, USA
| | - Alan R Sanders
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL 60637, USA
| | - Gao Wang
- Department of Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Kaixuan Luo
- Department of Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Subhajit Sengupta
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA
| | - Siobhan West
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA
| | - Sheng Qian
- Department of Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Michael Streit
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA
| | - Dimitrios Avramopoulos
- Department of Genetic Medicine and Psychiatry, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Chad A Cowan
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Mengjie Chen
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Zhiping P Pang
- Department of Neuroscience and Cell Biology, Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Pablo V Gejman
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL 60637, USA
| | - Xin He
- Department of Human Genetics, University of Chicago, Chicago, IL 60637, USA.
- Grossman Institute for Neuroscience, Quantitative Biology and Human Behavior, University of Chicago, Chicago, IL 60637, USA
| | - Jubao Duan
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA.
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
37
|
Hirano K, Kubo M, Fukuyama Y, Namihira M. Indonesian Ginger (Bangle) Extract Promotes Neurogenesis of Human Neural Stem Cells through WNT Pathway Activation. Int J Mol Sci 2020; 21:E4772. [PMID: 32635647 PMCID: PMC7369972 DOI: 10.3390/ijms21134772] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 06/26/2020] [Accepted: 07/03/2020] [Indexed: 11/16/2022] Open
Abstract
Indonesian ginger (Zingiber purpureum Rosc.), also known as Bangle, exhibits neurotrophic effects on cultured murine cortical neurons and in the adult mouse brain, but the underlying mechanisms remain unknown. Here, using human fetal neural stem cells (hfNSCs) as a model system for in vitro human neurogenesis, we show that Bangle extracts activate canonical WNT/β-catenin signaling. Bangle extract-treatment of hfNSCs not only promoted neuronal differentiation, but also accelerated neurite outgrowth from immature neurons. Furthermore, Bangle extracts induced expression of neurogenic genes and WNT signaling-target genes, and facilitated the accumulation of β-catenin in nuclei of hfNSC. Interestingly, altered histone modifications were also observed in Bangle-treated hfNSCs. Together, these findings demonstrate that Bangle contributes to hfNSC neurogenesis by WNT pathway and epigenetic regulation.
Collapse
Affiliation(s)
- Kazumi Hirano
- Molecular Neurophysiology Research Group, Biomedical Research Institute, The National Institute of Advanced Industrial Science and Technology (AIST), Ibaraki 305-8566, Japan
| | - Miwa Kubo
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima 770-8514, Japan; (M.K.); (Y.F.)
| | - Yoshiyasu Fukuyama
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima 770-8514, Japan; (M.K.); (Y.F.)
| | - Masakazu Namihira
- Molecular Neurophysiology Research Group, Biomedical Research Institute, The National Institute of Advanced Industrial Science and Technology (AIST), Ibaraki 305-8566, Japan
| |
Collapse
|
38
|
Powell SK, O'Shea CP, Shannon SR, Akbarian S, Brennand KJ. Investigation of Schizophrenia with Human Induced Pluripotent Stem Cells. ADVANCES IN NEUROBIOLOGY 2020; 25:155-206. [PMID: 32578147 DOI: 10.1007/978-3-030-45493-7_6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Schizophrenia is a chronic and severe neuropsychiatric condition manifested by cognitive, emotional, affective, perceptual, and behavioral abnormalities. Despite decades of research, the biological substrates driving the signs and symptoms of the disorder remain elusive, thus hampering progress in the development of treatments aimed at disease etiologies. The recent emergence of human induced pluripotent stem cell (hiPSC)-based models has provided the field with a highly innovative approach to generate, study, and manipulate living neural tissue derived from patients, making possible the exploration of fundamental roles of genes and early-life stressors in disease-relevant cell types. Here, we begin with a brief overview of the clinical, epidemiological, and genetic aspects of the condition, with a focus on schizophrenia as a neurodevelopmental disorder. We then highlight relevant technical advancements in hiPSC models and assess novel findings attained using hiPSC-based approaches and their implications for disease biology and treatment innovation. We close with a critical appraisal of the developments necessary for both further expanding knowledge of schizophrenia and the translation of new insights into therapeutic innovations.
Collapse
Affiliation(s)
- Samuel K Powell
- Medical Scientist Training Program, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Callan P O'Shea
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sara Rose Shannon
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Schahram Akbarian
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kristen J Brennand
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
39
|
Zhang DY, Song H, Ming GL. Modeling neurological disorders using brain organoids. Semin Cell Dev Biol 2020; 111:4-14. [PMID: 32561297 DOI: 10.1016/j.semcdb.2020.05.026] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/08/2020] [Accepted: 05/27/2020] [Indexed: 12/18/2022]
Abstract
Neurological disorders are challenging to study given the complexity and species-specific features of the organ system. Brain organoids are three dimensional structured aggregates of neural tissue that are generated by self-organization and differentiation from pluripotent stem cells under optimized culture conditions. These brain organoids exhibit similar features of structural organization and cell type diversity as the developing human brain, creating opportunities to recapitulate disease phenotypes that are not otherwise accessible. Here we review the initial attempt in the field to apply brain organoid models for the study of many different types of human neurological disorders across a wide range of etiologies and pathophysiologies. Forthcoming advancements in both brain organoid technology as well as analytical methods have significant potentials to advance the understanding of neurological disorders and to uncover opportunities for meaningful therapeutic intervention.
Collapse
Affiliation(s)
- Daniel Y Zhang
- Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Hongjun Song
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA; Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA; The Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Guo-Li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA; Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA; Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
40
|
Modeling Brain Disorders Using Induced Pluripotent Stem Cells. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a035659. [PMID: 31767646 DOI: 10.1101/cshperspect.a035659] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Brain disorders, from neurodegenerative to psychiatric disorders, are among the most challenging conditions to study because of the intricate nature of the human brain and the limitations of existing model systems in recapitulating all these intricacies. However, innovations in stem cell technologies now allow us to reprogram patient somatic cells to induced pluripotent stem cells (iPSCs), which can then be differentiated to disease-relevant neural and glial cells. iPSCs are a valuable tool to model brain disorders, as they can be derived from patients with known symptom histories, genetics, and drug-response profiles. Here, we discuss the premise and validity of the iPSC-based in vitro model system and highlight key findings from the most commonly studied neurodegenerative and psychiatric disorders.
Collapse
|
41
|
Zhang Y, Zhao Y, Song X, Luo H, Sun J, Han C, Gu X, Li J, Cai G, Zhu Y, Liu Z, Wei L, Wei ZZ. Modulation of Stem Cells as Therapeutics for Severe Mental Disorders and Cognitive Impairments. Front Psychiatry 2020; 11:80. [PMID: 32425815 PMCID: PMC7205035 DOI: 10.3389/fpsyt.2020.00080] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 01/31/2020] [Indexed: 12/11/2022] Open
Abstract
Severe mental illnesses (SMI) such as schizophrenia and bipolar disorder affect 2-4% of the world population. Current medications and diagnostic methods for mental illnesses are not satisfying. In animal studies, stem cell therapy is promising for some neuropsychiatric disorders and cognitive/social deficits, not only treating during development (targeting modulation and balancing) but also following neurodegeneration (cell replacement and regenerating support). We believe that novel interventions such as modulation of particular cell populations to develop cell-based treatment can improve cognitive and social functions in SMI. With pathological synaptic/myelin damage, oligodendrocytes seem to play a role. In this review, we have summarized oligodendrogenesis mechanisms and some related calcium signals in neural cells and stem/progenitor cells. The related benefits from endogenous stem/progenitor cells within the brain and exogenous stem cells, including multipotent mesenchymal-derived stromal cells (MSC), fetal neural stem cells (NSC), pluripotent stem cells (PSC), and differentiated progenitors, are discussed. These also include stimulating mechanisms of oligodendrocyte proliferation, maturation, and myelination, responsive to the regenerative effects by both endogenous stem cells and transplanted cells. Among the mechanisms, calcium signaling regulates the neuronal/glial progenitor cell (NPC/GPC)/oligodendrocyte precursor cell (OPC) proliferation, migration, and differentiation, dendrite development, and synaptic plasticity, which are involved in many neuropsychiatric diseases in human. On the basis of numerous protein annotation and protein-protein interaction databases, a total of 119 calcium-dependent/activated proteins that are related to neuropsychiatry in human are summarized in this investigation. One of the advanced methods, the calcium/cation-channel-optogenetics-based stimulation of stem cells and transplanted cells, can take advantage of calcium signaling regulations. Intranasal-to-brain delivery of drugs and stem cells or local delivery with the guidance of brain imaging techniques may provide a unique new approach for treating psychiatric disorders. It is also expected that preconditioning stem cell therapy following precise brain imaging as pathological confirmation has high potential if translated to cell clinic use. Generally, modulable cell transplantation followed by stimulations should provide paracrine protection, synaptic modulation, and myelin repair for the brain in SMI.
Collapse
Affiliation(s)
- Yongbo Zhang
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yingying Zhao
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, United States
| | - Xiaopeng Song
- McLean Imaging Center, McLean Hospital, Harvard Medical School, Belmont, MA, United States
| | - Hua Luo
- Emory Critical Care Center, Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States
| | - Jinmei Sun
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, United States
| | - Chunyu Han
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xiaohuan Gu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, United States
| | - Jun Li
- Department of Biological Psychiatry, Peking University Sixth Hospital, Beijing, China
- Department of Biological Psychiatry, Peking University Institute of Mental Health, Beijing, China
- Department of Biological Psychiatry, NHC Key Laboratory of Mental Health (Peking University), Beijing, China
- Department of Biological Psychiatry, National Clinical Research Center for Mental Disorders, Beijing, China
| | - Guilan Cai
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yanbing Zhu
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhandong Liu
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Ling Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, United States
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, United States
| | - Zheng Zachory Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
42
|
Abstract
PURPOSE OF REVIEW We review the ways in which stem cells are used in psychiatric disease research, including the related advances in gene editing and directed cell differentiation. RECENT FINDINGS The recent development of induced pluripotent stem cell (iPSC) technologies has created new possibilities for the study of psychiatric disease. iPSCs can be derived from patients or controls and differentiated to an array of neuronal and non-neuronal cell types. Their genomes can be edited as desired, and they can be assessed for a variety of phenotypes. This makes them especially interesting for studying genetic variation, which is particularly useful today now that our knowledge on the genetics of psychiatric disease is quickly expanding. The recent advances in cell engineering have led to powerful new methods for studying psychiatric illness including schizophrenia, bipolar disorder, and autism. There is a wide array of possible applications as illustrated by the many examples from the literature, most of which are cited here.
Collapse
Affiliation(s)
- Debamitra Das
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kyra Feuer
- Predoctoral Training Program in Human Genetics, Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Marah Wahbeh
- Predoctoral Training Program in Human Genetics, Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dimitrios Avramopoulos
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
43
|
Rajarajan P, Flaherty E, Akbarian S, Brennand KJ. CRISPR-based functional evaluation of schizophrenia risk variants. Schizophr Res 2020; 217:26-36. [PMID: 31277978 PMCID: PMC6939156 DOI: 10.1016/j.schres.2019.06.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 06/14/2019] [Accepted: 06/17/2019] [Indexed: 02/06/2023]
Abstract
As expanding genetic and genomic studies continue to implicate a growing list of variants contributing risk to neuropsychiatric disease, an important next step is to understand the functional impact and points of convergence of these risk factors. Here, with a focus on schizophrenia, we survey the most recent findings of the rare and common variants underlying genetic risk for schizophrenia. We discuss the ongoing efforts to validate these variants in post-mortem brain tissue, as well as new approaches to combine CRISPR-based genome engineering with patient-specific human induced pluripotent stem cell (hiPSC)-based models, in order to identify putative causal schizophrenia loci that regulate gene expression and cellular function. We consider the current limitations of hiPSC-based approaches as well as the future advances necessary to improve the fidelity of this human model. With the objective of utilizing patient genotype data to improve diagnosis and predict treatment response, the integration of CRISPR-genome engineering and hiPSC-based models represent an important strategy with which to systematically demonstrate the cell-type-specific effects of schizophrenia-associated variants.
Collapse
Affiliation(s)
- Prashanth Rajarajan
- Graduate School of Biomedical Science, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America; Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America
| | - Erin Flaherty
- Graduate School of Biomedical Science, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America; Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America
| | - Schahram Akbarian
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America
| | - Kristen J Brennand
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America; Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America; Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America.
| |
Collapse
|
44
|
Hoffmann A, Ziller M, Spengler D. Focus on Causality in ESC/iPSC-Based Modeling of Psychiatric Disorders. Cells 2020; 9:E366. [PMID: 32033412 PMCID: PMC7072492 DOI: 10.3390/cells9020366] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/31/2020] [Accepted: 02/03/2020] [Indexed: 12/14/2022] Open
Abstract
Genome-wide association studies (GWAS) have identified an increasing number of genetic variants that significantly associate with psychiatric disorders. Despite this wealth of information, our knowledge of which variants causally contribute to disease, how they interact, and even more so of the functions they regulate, is still poor. The availability of embryonic stem cells (ESCs) and the advent of patient-specific induced pluripotent stem cells (iPSCs) has opened new opportunities to investigate genetic risk variants in living disease-relevant cells. Here, we analyze how this progress has contributed to the analysis of causal relationships between genetic risk variants and neuronal phenotypes, especially in schizophrenia (SCZ) and bipolar disorder (BD). Studies on rare, highly penetrant risk variants have originally led the field, until more recently when the development of (epi-) genetic editing techniques spurred studies on cause-effect relationships between common low risk variants and their associated neuronal phenotypes. This reorientation not only offers new insights, but also raises issues on interpretability. Concluding, we consider potential caveats and upcoming developments in the field of ESC/iPSC-based modeling of causality in psychiatric disorders.
Collapse
Affiliation(s)
| | | | - Dietmar Spengler
- Department of Translational Research in Psychiatry, Max-Planck Institute of Psychiatry, 80804 Munich, Germany; (A.H.); (M.Z.)
| |
Collapse
|
45
|
Chen C, Yang Y, Yao Y. HBO Promotes the Differentiation of Neural Stem Cells via Interactions Between the Wnt3/β-Catenin and BMP2 Signaling Pathways. Cell Transplant 2019; 28:1686-1699. [PMID: 31694396 PMCID: PMC6923559 DOI: 10.1177/0963689719883578] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Hyperbaric oxygen (HBO) therapy may promote neurological recovery from hypoxic-ischemic
encephalopathy (HIE). However, the therapeutic effects of HBO and its associated
mechanisms remain unknown. The canonical Wnt/β-catenin signaling pathways and bone
morphogenetic protein (BMP) play important roles in mammalian nervous system development.
The present study examined whether HBO stimulates the differentiation of neural stem cells
(NSCs) and its effect on Wnt3/β-catenin and BMP2 signaling pathways. We showed HBO
treatment (2 ATA, 60 min) promoted differentiation of NSCs into neurons and
oligodendrocytes in vitro. In addition, rat hypoxic-ischemic brain damage (HIBD) tissue
extracts also promoted the differentiation of NSCs into neurons and oligodendrocytes, with
the advantage of reducing the number of astrocytes. These effects were most pronounced
when these two were combined together. In addition, the expression of Wnt3a, BMP2, and
β-catenin nuclear proteins were increased after HBO treatment. However, blockade of
Wnt/β-catenin or BMP signaling inhibited NSC differentiation and reduced the expression of
Wnt3a, BMP2, and β-catenin nuclear proteins. In conclusion, HBO promotes differentiation
of NSCs into neurons and oligodendrocytes and reduced the number of astrocytes in vitro
possibly through regulation of Wnt3/β-catenin and BMP2 signaling pathways. HBO may serve
as a potential therapeutic strategy for treating HIE.
Collapse
Affiliation(s)
- Chongfeng Chen
- Department of Pediatrics, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou City, Guangdong, China
| | - Yujia Yang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha City, Hunan, P.R. China
| | - Yue Yao
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha City, Hunan, P.R. China
| |
Collapse
|
46
|
Balan S, Toyoshima M, Yoshikawa T. Contribution of induced pluripotent stem cell technologies to the understanding of cellular phenotypes in schizophrenia. Neurobiol Dis 2019; 131:104162. [DOI: 10.1016/j.nbd.2018.04.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 04/23/2018] [Accepted: 04/28/2018] [Indexed: 02/07/2023] Open
|
47
|
Moslem M, Olive J, Falk A. Stem cell models of schizophrenia, what have we learned and what is the potential? Schizophr Res 2019; 210:3-12. [PMID: 30587427 DOI: 10.1016/j.schres.2018.12.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 12/14/2018] [Accepted: 12/16/2018] [Indexed: 12/13/2022]
Abstract
Schizophrenia is a complex disorder with clinical manifestations in early adulthood. However, it may start with disruption of brain development caused by genetic or environmental factors, or both. Early deteriorating effects of genetic/environmental factors on neural development might be key to described disease causing mechanisms. Establishing cellular models with cells from affected individual using the induced pluripotent stem cells (iPSC) technology could be used to mimic early neurodevelopment alterations caused by risk genes or environmental stressors. Indeed, cellular models have allowed identification and further study of risk factors and the biological pathways in which they are involved. New advancements in differentiation methods such as defined and robust monolayer protocols and cerebral 3D organoids have made it possible to faithfully mimic neural development and neuronal functionality while CRISPR-editing tools assist to engineer isogenic cell lines to precisely explore genetic variation in polygenic diseases such as schizophrenia. Here we review the current field of iPSC models of schizophrenia and how risk factors can be modelled as well as discussing the common biological pathways involved.
Collapse
Affiliation(s)
- Mohsen Moslem
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | - Jessica Olive
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden; Department of Life Sciences, Imperial College London, United Kingdom.
| | - Anna Falk
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
48
|
Johnstone M, Hillary RF, St Clair D. Stem Cells to Inform the Neurobiology of Mental Illness. Curr Top Behav Neurosci 2019; 40:13-43. [PMID: 30030769 DOI: 10.1007/7854_2018_57] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The inception of human-induced pluripotent stem cell (hiPSCs) technology has provided an exciting platform upon which the modelling and treatment of human neurodevelopmental and neuropsychiatric disorders may be expedited. Although the genetic architecture of these disorders is far more complex than previously imagined, many key loci have at last been identified. This has allowed in vivo and in vitro technologies to be refined to model specific high-penetrant genetic loci involved in both disorders. Animal models of neurodevelopmental disorders, such as schizophrenia and autism spectrum disorders, show limitations in recapitulating the full complexity and heterogeneity of human neurodevelopmental disease states. Indeed, patient-derived hiPSCs offer distinct advantages over classical animal models in the study of human neuropathologies. Here we have discussed the current, relative translational merit of hiPSCs in investigating human neurodevelopmental and neuropsychiatric disorders with a specific emphasis on the utility of such systems to aid in the identification of biomarkers. We have highlighted the promises and pitfalls of reprogramming cell fate for the study of these disorders and provide recommendations for future directions in this field in order to overcome current limitations. Ultimately, this will aid in the development of effective clinical strategies for diverse patient populations affected by these disorders with the aim of also leading to biomarker identification.
Collapse
Affiliation(s)
- Mandy Johnstone
- Division of Psychiatry, Royal Edinburgh Hospital, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, Scotland, UK.
| | - Robert F Hillary
- Division of Psychiatry, Royal Edinburgh Hospital, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, Scotland, UK
| | - David St Clair
- Division of Psychiatry, Royal Edinburgh Hospital, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, Scotland, UK
| |
Collapse
|
49
|
Pruski M, Lang B. Primary Cilia-An Underexplored Topic in Major Mental Illness. Front Psychiatry 2019; 10:104. [PMID: 30886591 PMCID: PMC6409319 DOI: 10.3389/fpsyt.2019.00104] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 02/12/2019] [Indexed: 12/20/2022] Open
Abstract
Though much progress has been made in recent years towards understanding the function and physiology of primary cilia, they remain a somewhat elusive organelle. Some studies have explored the role of primary cilia in the developing nervous system, and their dysfunction has been linked with several neurosensory deficits. Yet, very little has been written on their potential role in psychiatric disorders. This article provides an overview of some of the functions of primary cilia in signalling pathways, and demonstrates that they are a worthy candidate in psychiatric research. The links between primary cilia and major mental illness have been demonstrated to exist at several levels, spanning genetics, signalling pathways, and pharmacology as well as cell division and migration. The primary focus of this review is on the sensory role of the primary cilium and the neurodevelopmental hypothesis of psychiatric disease. As such, the primary cilium is demonstrated to be a key link between the cellular environment and cell behaviour, and hence of key importance in the considerations of the nature and nurture debate in psychiatric research.
Collapse
Affiliation(s)
- Michal Pruski
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China
- Critical Care Laboratory, Critical Care Directorate, Manchester Royal Infirmary, Manchester University NHS Foundation Trust, Manchester, United Kingdom
- School of Healthcare Science, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, United Kingdom
| | - Bing Lang
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| |
Collapse
|
50
|
Wilkinson B, Evgrafov O, Zheng D, Hartel N, Knowles JA, Graham NA, Ichida J, Coba MP. Endogenous Cell Type-Specific Disrupted in Schizophrenia 1 Interactomes Reveal Protein Networks Associated With Neurodevelopmental Disorders. Biol Psychiatry 2019; 85:305-316. [PMID: 29961565 PMCID: PMC6251761 DOI: 10.1016/j.biopsych.2018.05.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 04/03/2018] [Accepted: 05/03/2018] [Indexed: 11/17/2022]
Abstract
BACKGROUND Disrupted in schizophrenia 1 (DISC1) has been implicated in a number of psychiatric diseases along with neurodevelopmental phenotypes such as the proliferation and differentiation of neural progenitor cells. While there has been significant effort directed toward understanding the function of DISC1 through the determination of its protein-protein interactions within an in vitro setting, endogenous interactions involving DISC1 within a cell type-specific setting relevant to neural development remain unclear. METHODS Using CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/CRISPR-associated protein 9) genome engineering technology, we inserted an endogenous 3X-FLAG tag at the C-terminus of the canonical DISC1 gene in human induced pluripotent stem cells (iPSCs). We further differentiated these cells and used affinity purification to determine protein-protein interactions involving DISC1 in iPSC-derived neural progenitor cells and astrocytes. RESULTS We were able to determine 151 novel cell type-specific proteins present in DISC1 endogenous interactomes. The DISC1 interactomes can be clustered into several subcomplexes that suggest novel DISC1 cell-specific functions. In addition, the DISC1 interactome in iPSC-derived neural progenitor cells associates in a connected network containing proteins found to harbor de novo mutations in patients affected by schizophrenia and contains a subset of novel interactions that are known to harbor syndromic mutations in neurodevelopmental disorders. CONCLUSIONS Endogenous DISC1 interactomes within iPSC-derived human neural progenitor cells and astrocytes are able to provide context to DISC1 function in a cell type-specific setting relevant to neural development and enables the integration of psychiatric disease risk factors within a set of defined molecular functions.
Collapse
Affiliation(s)
- Brent Wilkinson
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Oleg Evgrafov
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY 11203, USA
| | - DongQing Zheng
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90033, USA
| | - Nicolas Hartel
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90033, USA
| | - James A. Knowles
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY 11203, USA
| | - Nicholas A. Graham
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90033, USA
| | - Justin Ichida
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA,Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA,Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC
| | - Marcelo P. Coba
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA,Department of Psychiatry and Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA,Corresponding Author: Marcelo P. Coba, Keck School of Medicine, University of Southern California, Zilkha Neurogenetic Institute, 1501 San Pablo St, Los Angeles, CA 90033, USA. Phone: 323-442-4345.
| |
Collapse
|