1
|
Santarriaga S, Gerlovin K, Layadi Y, Karmacharya R. Human stem cell-based models to study synaptic dysfunction and cognition in schizophrenia: A narrative review. Schizophr Res 2024; 273:78-97. [PMID: 36925354 PMCID: PMC10500041 DOI: 10.1016/j.schres.2023.02.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 02/27/2023] [Accepted: 02/28/2023] [Indexed: 03/18/2023]
Abstract
Cognitive impairment is the strongest predictor of functional outcomes in schizophrenia and is hypothesized to result from synaptic dysfunction. However, targeting synaptic plasticity and cognitive deficits in patients remains a significant clinical challenge. A comprehensive understanding of synaptic plasticity and the molecular basis of learning and memory in a disease context can provide specific targets for the development of novel therapeutics targeting cognitive impairments in schizophrenia. Here, we describe the role of synaptic plasticity in cognition, summarize evidence for synaptic dysfunction in schizophrenia and demonstrate the use of patient derived induced-pluripotent stem cells for studying synaptic plasticity in vitro. Lastly, we discuss current advances and future technologies for bridging basic science research of synaptic dysfunction with clinical and translational research that can be used to predict treatment response and develop novel therapeutics.
Collapse
Affiliation(s)
- Stephanie Santarriaga
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Chemical Biology and Therapeutic Science Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Kaia Gerlovin
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Chemical Biology and Therapeutic Science Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Yasmine Layadi
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Chimie ParisTech, Université Paris Sciences et Lettres, Paris, France
| | - Rakesh Karmacharya
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Chemical Biology and Therapeutic Science Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA; Schizophrenia and Bipolar Disorder Program, McLean Hospital, Belmont, MA, USA.
| |
Collapse
|
2
|
Lizano P, Karmacharya R. Harnessing stem cell-based approaches for clinically meaningful discoveries in schizophrenia. Schizophr Res 2024; 273:1-3. [PMID: 39209607 PMCID: PMC11570341 DOI: 10.1016/j.schres.2024.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024]
Affiliation(s)
- Paulo Lizano
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA; Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, USA; Division Chief of Translational Neuroscience, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| | - Rakesh Karmacharya
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA; Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Chemical Biology Program, Broad Institute of MIT & Harvard, Cambridge, MA, USA; Schizophrenia & Bipolar Disorder Program, McLean Hospital, Belmont, MA, USA
| |
Collapse
|
3
|
Dines M, Kes M, Ailán D, Cetkovich-Bakmas M, Born C, Grunze H. Bipolar disorders and schizophrenia: discrete disorders? Front Psychiatry 2024; 15:1352250. [PMID: 38745778 PMCID: PMC11091416 DOI: 10.3389/fpsyt.2024.1352250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 04/16/2024] [Indexed: 05/16/2024] Open
Abstract
Background With similarities in heritability, neurobiology and symptomatology, the question has been raised whether schizophrenia and bipolar disorder are truly distinctive disorders or belong to a continuum. This narrative review summarizes common and distinctive findings from genetics, neuroimaging, cognition and clinical course that may help to solve this ethiopathogenetic puzzle. Methods The authors conducted a literature search for papers listed in PubMed and Google Scholar, using the search terms "schizophrenia" and "bipolar disorder" combined with different terms such as "genes", "neuroimaging studies", "phenomenology differences", "cognition", "epidemiology". Articles were considered for inclusion if they were written in English or Spanish, published as full articles, if they compared subjects with schizophrenia and bipolar disorder, or subjects with either disorder with healthy controls, addressing differences between groups. Results Several findings support the hypothesis that schizophrenia and bipolar disorder are discrete disorders, yet some overlapping of findings exists. The evidence for heritability of both SZ and BD is obvious, as well as the environmental impact on individual manifestations of both disorders. Neuroimaging studies support subtle differences between disorders, it appears to be rather a pattern of irregularities than an unequivocally unique finding distinguishing schizophrenia from bipolar disorder. The cognitive profile displays differences between disorders in certain domains, such as premorbid intellectual functioning and executive functions. Finally, the timing and trajectory of cognitive impairment in both disorders also differs. Conclusion The question whether SZ and BD belong to a continuum or are separate disorders remains a challenge for further research. Currently, our research tools may be not precise enough to carve out distinctive, unique and undisputable differences between SZ and BD, but current evidence favors separate disorders. Given that differences are subtle, a way to overcome diagnostic uncertainties in the future could be the application of artificial intelligence based on BigData. Limitations Despite the detailed search, this article is not a full and complete review of all available studies on the topic. The search and selection of papers was also limited to articles in English and Spanish. Selection of papers and conclusions may be biased by the personal view and clinical experience of the authors.
Collapse
Affiliation(s)
- Micaela Dines
- Department of Psychiatry, Instituto de Neurología Cognitiva (INECO), Buenos Aires, Argentina
- Department of Psychiatry, Instituto de Neurociencia Cognitiva y Traslacional (Consejo Nacional de Investigaciones Científicas y Técnicas - Fundación INECO - Universidad Favaloro), Buenos Aires, Argentina
| | - Mariana Kes
- Department of Psychiatry, Instituto de Neurología Cognitiva (INECO), Buenos Aires, Argentina
- Department of Psychiatry, Instituto de Neurociencia Cognitiva y Traslacional (Consejo Nacional de Investigaciones Científicas y Técnicas - Fundación INECO - Universidad Favaloro), Buenos Aires, Argentina
| | - Delfina Ailán
- Department of Psychiatry, Instituto de Neurología Cognitiva (INECO), Buenos Aires, Argentina
- Department of Psychiatry, Instituto de Neurociencia Cognitiva y Traslacional (Consejo Nacional de Investigaciones Científicas y Técnicas - Fundación INECO - Universidad Favaloro), Buenos Aires, Argentina
| | - Marcelo Cetkovich-Bakmas
- Department of Psychiatry, Instituto de Neurología Cognitiva (INECO), Buenos Aires, Argentina
- Department of Psychiatry, Instituto de Neurociencia Cognitiva y Traslacional (Consejo Nacional de Investigaciones Científicas y Técnicas - Fundación INECO - Universidad Favaloro), Buenos Aires, Argentina
| | - Christoph Born
- Department of Psychiatry, Psychiatrie Schwäbisch Hall, Ringstraße, Germany
- Department of Psychiatry, Paracelsus Medical University, Nuremberg, Germany
| | - Heinz Grunze
- Department of Psychiatry, Psychiatrie Schwäbisch Hall, Ringstraße, Germany
- Department of Psychiatry, Paracelsus Medical University, Nuremberg, Germany
| |
Collapse
|
4
|
Kathuria A, Lopez-Lengowski K, Watmuff B, Karmacharya R. Morphological and transcriptomic analyses of stem cell-derived cortical neurons reveal mechanisms underlying synaptic dysfunction in schizophrenia. Genome Med 2023; 15:58. [PMID: 37507766 PMCID: PMC10375745 DOI: 10.1186/s13073-023-01203-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 06/16/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND Postmortem studies in schizophrenia consistently show reduced dendritic spines in the cerebral cortex but the mechanistic underpinnings of these deficits remain unknown. Recent genome-wide association studies and exome sequencing investigations implicate synaptic genes and processes in the disease biology of schizophrenia. METHODS We generated human cortical pyramidal neurons by differentiating iPSCs of seven schizophrenia patients and seven healthy subjects, quantified dendritic spines and synapses in different cortical neuron subtypes, and carried out transcriptomic studies to identify differentially regulated genes and aberrant cellular processes in schizophrenia. RESULTS Cortical neurons expressing layer III marker CUX1, but not those expressing layer V marker CTIP2, showed significant reduction in dendritic spine density in schizophrenia, mirroring findings in postmortem studies. Transcriptomic experiments in iPSC-derived cortical neurons showed that differentially expressed genes in schizophrenia were enriched for genes implicated in schizophrenia in genome-wide association and exome sequencing studies. Moreover, most of the differentially expressed genes implicated in schizophrenia genetic studies had lower expression levels in schizophrenia cortical neurons. Network analysis of differentially expressed genes led to identification of NRXN3 as a hub gene, and follow-up experiments showed specific reduction of the NRXN3 204 isoform in schizophrenia neurons. Furthermore, overexpression of the NRXN3 204 isoform in schizophrenia neurons rescued the spine and synapse deficits in the cortical neurons while knockdown of NRXN3 204 in healthy neurons phenocopied spine and synapse deficits seen in schizophrenia cortical neurons. The antipsychotic clozapine increased expression of the NRXN3 204 isoform in schizophrenia cortical neurons and rescued the spine and synapse density deficits. CONCLUSIONS Taken together, our findings in iPSC-derived cortical neurons recapitulate cell type-specific findings in postmortem studies in schizophrenia and have led to the identification of a specific isoform of NRXN3 that modulates synaptic deficits in schizophrenia neurons.
Collapse
Affiliation(s)
- Annie Kathuria
- Harvard University, MGH Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, CPZN6, Boston, MA, 02114, USA
- Chemical Biology Program, Broad Institute of MIT & Harvard, Cambridge, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Kara Lopez-Lengowski
- Harvard University, MGH Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, CPZN6, Boston, MA, 02114, USA
- Chemical Biology Program, Broad Institute of MIT & Harvard, Cambridge, MA, USA
| | - Bradley Watmuff
- Harvard University, MGH Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, CPZN6, Boston, MA, 02114, USA
- Chemical Biology Program, Broad Institute of MIT & Harvard, Cambridge, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Rakesh Karmacharya
- Harvard University, MGH Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, CPZN6, Boston, MA, 02114, USA.
- Chemical Biology Program, Broad Institute of MIT & Harvard, Cambridge, MA, USA.
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA.
- Program in Neuroscience, Harvard University, Cambridge, MA, USA.
- Schizophrenia & Bipolar Disorder Program, McLean Hospital, Belmont, MA, USA.
- Program in Chemical Biology, Harvard University, Cambridge, MA, USA.
- Harvard Stem Cell Institute, Cambridge, MA, USA.
| |
Collapse
|
5
|
Kathuria A, Lopez-Lengowski K, McPhie D, Cohen BM, Karmacharya R. Disease-specific differences in gene expression, mitochondrial function and mitochondria-endoplasmic reticulum interactions in iPSC-derived cerebral organoids and cortical neurons in schizophrenia and bipolar disorder. DISCOVER MENTAL HEALTH 2023; 3:8. [PMID: 36915374 PMCID: PMC9998323 DOI: 10.1007/s44192-023-00031-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 01/04/2023] [Indexed: 03/12/2023]
Abstract
We compared transcriptomic profiles of cerebral organoids differentiated from induced pluripotent stem cells of eight schizophrenia and eight bipolar disorder patients to identify genes that were differentially expressed in cerebral organoids between two disorders. Gene ontology analysis showed relative up-regulation in schizophrenia organoids of genes related to response to cytokines, antigen binding and clathrin-coated vesicles, while showing up-regulation in bipolar disorder of genes involved in calcium binding. Gene set enrichment analysis revealed enrichment in schizophrenia of genes involved in mitochondrial and oxidative phosphorylation while showing enrichment in bipolar disorder of genes involved in long term potentiation and neuro-transporters. We compared mitochondrial function in cerebral organoids from schizophrenia and bipolar disorder subjects and found that while schizophrenia organoids showed deficits in basal oxygen consumption rate and ATP production when compared to healthy control organoids, while bipolar disorder organoids did not show these deficits. Gene ontology analyses also revealed enrichment in bipolar disorder of genes in ion binding and regulation of transport. Experiments examining the interaction between mitochondria and endoplasmic reticulum in cortical neurons from bipolar disorder subjects showed a significantly lower number of contact sites between mitochondria and endoplasmic reticulum when compared to cortical neurons from schizophrenia patients. These results point to disease-specific deficits in mitochondrial respiration in schizophrenia and in mitochondrial-endoplasmic reticulum interactions in bipolar disorder. Supplementary Information The online version contains supplementary material available at 10.1007/s44192-023-00031-8.
Collapse
Affiliation(s)
- Annie Kathuria
- Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114 USA
- Chemical Biology Program, Broad Institute of MIT & Harvard, Cambridge, MA USA
- Department of Psychiatry, Harvard Medical School, Boston, MA USA
| | - Kara Lopez-Lengowski
- Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114 USA
- Chemical Biology Program, Broad Institute of MIT & Harvard, Cambridge, MA USA
| | - Donna McPhie
- Department of Psychiatry, Harvard Medical School, Boston, MA USA
- Schizophrenia and Bipolar Disorder Program, McLean Hospital, Belmont, MA USA
| | - Bruce M. Cohen
- Department of Psychiatry, Harvard Medical School, Boston, MA USA
- Schizophrenia and Bipolar Disorder Program, McLean Hospital, Belmont, MA USA
| | - Rakesh Karmacharya
- Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114 USA
- Chemical Biology Program, Broad Institute of MIT & Harvard, Cambridge, MA USA
- Department of Psychiatry, Harvard Medical School, Boston, MA USA
- Schizophrenia and Bipolar Disorder Program, McLean Hospital, Belmont, MA USA
- Program in Neuroscience, Harvard University, Cambridge, MA USA
- Program in Chemical Biology, Harvard University, Cambridge, MA USA
- Harvard Stem Cell Institute, Cambridge, MA USA
| |
Collapse
|
6
|
Distinct effects of interleukin-6 and interferon-γ on differentiating human cortical neurons. Brain Behav Immun 2022; 103:97-108. [PMID: 35429607 PMCID: PMC9278892 DOI: 10.1016/j.bbi.2022.04.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/29/2022] [Accepted: 04/10/2022] [Indexed: 01/17/2023] Open
Abstract
Translational evidence suggests that cytokines involved in maternal immune activation (MIA), such as interleukin-6 (IL-6) and interferon-γ (IFN-γ), can cross the placenta, injure fetal brain, and predispose to neuropsychiatric disorders. To elaborate developmental neuronal sequelae of MIA, we differentiated human pluripotent stem cells to cortical neurons over a two-month period, exposing them to IL-6 or IFN-γ. IL-6 impacted expression of genes regulating extracellular matrix, actin cytoskeleton and TGF-β signaling while IFN-γ impacted genes regulating antigen processing, major histocompatibility complex and endoplasmic reticulum biology. IL-6, but not IFN-γ, altered mitochondrial respiration while IFN-γ, but not IL-6, induced reduction in dendritic spine density. Pre-treatment with folic acid, which has known neuroprotective and anti-inflammatory properties, ameliorated IL-6 effects on mitochondrial respiration and IFN-γ effects on dendritic spine density. These findings suggest distinct mechanisms for how fetal IL-6 and IFN-γ exposure influence risk for neuropsychiatric disorders, and how folic acid can mitigate such risk.
Collapse
|
7
|
Pong S, Karmacharya R, Sofman M, Bishop JR, Lizano P. The Role of Brain Microvascular Endothelial Cell and Blood-Brain Barrier Dysfunction in Schizophrenia. Complex Psychiatry 2020; 6:30-46. [PMID: 34883503 PMCID: PMC7673590 DOI: 10.1159/000511552] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 09/10/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Despite decades of research, little clarity exists regarding pathogenic mechanisms related to schizophrenia. Investigations on the disease biology of schizophrenia have primarily focused on neuronal alterations. However, there is substantial evidence pointing to a significant role for the brain's microvasculature in mediating neuroinflammation in schizophrenia. SUMMARY Brain microvascular endothelial cells (BMEC) are a central element of the microvasculature that forms the blood-brain barrier (BBB) and shields the brain against toxins and immune cells via paracellular, transcellular, transporter, and extracellular matrix proteins. While evidence for BBB dysfunction exists in brain disorders, including schizophrenia, it is not known if BMEC themselves are functionally compromised and lead to BBB dysfunction. KEY MESSAGES Genome-wide association studies, postmortem investigations, and gene expression analyses have provided some insights into the role of the BBB in schizophrenia pathophysiology. However, there is a significant gap in our understanding of the role that BMEC play in BBB dysfunction. Recent advances differentiating human BMEC from induced pluripotent stem cells (iPSC) provide new avenues to examine the role of BMEC in BBB dysfunction in schizophrenia.
Collapse
Affiliation(s)
- Sovannarath Pong
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Rakesh Karmacharya
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, USA
| | - Marianna Sofman
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Jeffrey R. Bishop
- Departments of Clinical and Experimental Pharmacology and Psychiatry, University of Minnesota, Minneapolis, Minnesota, USA
| | - Paulo Lizano
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
8
|
Kathuria A, Lopez-Lengowski K, Watmuff B, Karmacharya R. Comparative Transcriptomic Analysis of Cerebral Organoids and Cortical Neuron Cultures Derived from Human Induced Pluripotent Stem Cells. Stem Cells Dev 2020; 29:1370-1381. [PMID: 32862797 DOI: 10.1089/scd.2020.0069] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Human induced pluripotent stem cells (iPSCs) can be differentiated along various neuronal lineages to generate two-dimensional neuronal cultures as well as three-dimensional brain organoids. Such iPSC-derived cellular models are being utilized to study the basic biology of human neuronal function and to interrogate the molecular underpinnings of disease biology. The different cellular models generated from iPSCs have varying properties in terms of the diversity and organization of the cells as well as the cellular functions that are present. To understand transcriptomic differences in iPSC-derived monolayer neuronal cultures and three-dimensional brain organoids, we differentiated eight human iPSC lines from healthy control subjects to generate cerebral organoids and cortical neuron monolayer cultures from the same set of iPSC lines. We undertook RNA-seq experiments in these model systems and analyzed the gene expression data to identify genes that are differentially expressed in cerebral organoids and two-dimensional cortical neuron cultures. In cerebral organoids, gene ontology analysis showed enrichment of genes involved in tissue development, response to stimuli, and the interferon-γ pathway, while two-dimensional cortical neuron cultures showed enrichment of genes involved in nervous system development and neurogenesis. We also undertook comparative analysis of these gene expression profiles with transcriptomic data from the human fetal prefrontal cortex (PFC). This analysis showed greater overlap of the fetal PFC transcriptome with cerebral organoid gene expression profiles compared to monolayer cortical neuron culture profiles. Our studies delineate the transcriptomic differences between cortical neuron monolayer cultures and three-dimensional cerebral organoids and can help inform the appropriate use of these model systems to address specific scientific questions.
Collapse
Affiliation(s)
- Annie Kathuria
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA.,Chemical Biology Program, Broad Institute of MIT & Harvard, Cambridge, Massachusetts, USA.,Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, USA
| | - Kara Lopez-Lengowski
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA.,Chemical Biology Program, Broad Institute of MIT & Harvard, Cambridge, Massachusetts, USA
| | - Bradley Watmuff
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA.,Chemical Biology Program, Broad Institute of MIT & Harvard, Cambridge, Massachusetts, USA.,Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, USA
| | - Rakesh Karmacharya
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA.,Chemical Biology Program, Broad Institute of MIT & Harvard, Cambridge, Massachusetts, USA.,Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, USA.,Schizophrenia and Bipolar Disorder Program, McLean Hospital, Belmont, Massachusetts, USA.,Program in Neuroscience, Harvard University, Cambridge, Massachusetts, USA.,Program in Chemical Biology, Harvard University, Cambridge, Massachusetts, USA.,Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| |
Collapse
|
9
|
Kathuria A, Lopez-Lengowski K, Jagtap SS, McPhie D, Perlis RH, Cohen BM, Karmacharya R. Transcriptomic Landscape and Functional Characterization of Induced Pluripotent Stem Cell-Derived Cerebral Organoids in Schizophrenia. JAMA Psychiatry 2020; 77:745-754. [PMID: 32186681 PMCID: PMC7081156 DOI: 10.1001/jamapsychiatry.2020.0196] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
IMPORTANCE Three-dimensional cerebral organoids generated from patient-derived induced pluripotent stem cells (iPSCs) may be used to interrogate cellular-molecular underpinnings of schizophrenia. OBJECTIVE To determine transcriptomic profiles and functional characteristics of cerebral organoids from patients with schizophrenia using gene expression studies, complemented with investigations of mitochondrial function through measurement of real-time oxygen consumption rate, and functional studies of neuronal firing with microelectrode arrays. DESIGN, SETTING, AND PARTICIPANTS This case-control study was conducted at Massachusetts General Hospital between 2017 and 2019. Transcriptomic profiling of iPSC-derived cerebral organoids from 8 patients with schizophrenia and 8 healthy control individuals was undertaken to identify cellular pathways that are aberrant in schizophrenia. Induced pluripotent stem cells and cerebral organoids were generated from patients who had been diagnosed as having schizophrenia and from heathy control individuals. MAIN OUTCOMES AND MEASURES Transcriptomic analysis of iPSC-derived cerebral organoids from patients with schizophrenia show differences in expression of genes involved in synaptic biology and neurodevelopment and are enriched for genes implicated in schizophrenia genome-wide association studies (GWAS). RESULTS The study included iPSC lines generated from 11 male and 5 female white participants, with a mean age of 38.8 years. RNA sequencing data from iPSC-derived cerebral organoids in schizophrenia showed differential expression of genes involved in synapses, in nervous system development, and in antigen processing. The differentially expressed genes were enriched for genes implicated in schizophrenia, with 23% of GWAS genes showing differential expression in schizophrenia and control organoids: 10 GWAS genes were upregulated in schizophrenia organoids while 15 GWAS genes were downregulated. Analysis of the gene expression profiles suggested dysregulation of genes involved in mitochondrial function and those involved in modulation of excitatory and inhibitory pathways. Studies of mitochondrial respiration showed lower basal consumption rate, adenosine triphosphate production, proton leak, and nonmitochondrial oxygen consumption in schizophrenia cerebral organoids, without any differences in the extracellular acidification rate. Microelectrode array studies of cerebral organoids showed no differences in baseline electrical activity in schizophrenia but revealed a diminished response to stimulation and depolarization. CONCLUSIONS AND RELEVANCE Investigations of patient-derived cerebral organoids in schizophrenia revealed gene expression patterns suggesting dysregulation of a number of pathways in schizophrenia, delineated differences in mitochondrial function, and showed deficits in response to stimulation and depolarization in schizophrenia.
Collapse
Affiliation(s)
- Annie Kathuria
- Center for Genomic Medicine, Massachusetts
General Hospital, Boston,Chemical Biology Program, Broad Institute of
Massachusetts Institute of Technology and Harvard, Cambridge,Department of Psychiatry, Harvard Medical
School, Boston, Massachusetts
| | - Kara Lopez-Lengowski
- Center for Genomic Medicine, Massachusetts
General Hospital, Boston,Chemical Biology Program, Broad Institute of
Massachusetts Institute of Technology and Harvard, Cambridge
| | - Smita S. Jagtap
- Center for Genomic Medicine, Massachusetts
General Hospital, Boston
| | - Donna McPhie
- Department of Psychiatry, Harvard Medical
School, Boston, Massachusetts,Schizophrenia and Bipolar Disorder Program,
McLean Hospital, Belmont, Massachusetts
| | - Roy H. Perlis
- Center for Genomic Medicine, Massachusetts
General Hospital, Boston,Department of Psychiatry, Harvard Medical
School, Boston, Massachusetts
| | - Bruce M. Cohen
- Department of Psychiatry, Harvard Medical
School, Boston, Massachusetts,Schizophrenia and Bipolar Disorder Program,
McLean Hospital, Belmont, Massachusetts
| | - Rakesh Karmacharya
- Center for Genomic Medicine, Massachusetts
General Hospital, Boston,Chemical Biology Program, Broad Institute of
Massachusetts Institute of Technology and Harvard, Cambridge,Department of Psychiatry, Harvard Medical
School, Boston, Massachusetts,Schizophrenia and Bipolar Disorder Program,
McLean Hospital, Belmont, Massachusetts,Program in Neuroscience, Harvard University,
Cambridge, Massachusetts,Program in Chemical Biology, Harvard University,
Cambridge, Massachusetts,Harvard Stem Cell Institute, Cambridge,
Massachusetts
| |
Collapse
|
10
|
Silva MC, Haggarty SJ. Human pluripotent stem cell-derived models and drug screening in CNS precision medicine. Ann N Y Acad Sci 2020; 1471:18-56. [PMID: 30875083 PMCID: PMC8193821 DOI: 10.1111/nyas.14012] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 01/02/2019] [Accepted: 01/07/2019] [Indexed: 12/12/2022]
Abstract
Development of effective therapeutics for neurological disorders has historically been challenging partly because of lack of accurate model systems in which to investigate disease etiology and test new therapeutics at the preclinical stage. Human stem cells, particularly patient-derived induced pluripotent stem cells (iPSCs) upon differentiation, have the ability to recapitulate aspects of disease pathophysiology and are increasingly recognized as robust scalable systems for drug discovery. We review advances in deriving cellular models of human central nervous system (CNS) disorders using iPSCs along with strategies for investigating disease-relevant phenotypes, translatable biomarkers, and therapeutic targets. Given their potential to identify novel therapeutic targets and leads, we focus on phenotype-based, small-molecule screens employing human stem cell-derived models. Integrated efforts to assemble patient iPSC-derived cell models with deeply annotated clinicopathological data, along with molecular and drug-response signatures, may aid in the stratification of patients, diagnostics, and clinical trial success, shifting translational science and precision medicine approaches. A number of remaining challenges, including the optimization of cost-effective, large-scale culture of iPSC-derived cell types, incorporation of aging into neuronal models, as well as robustness and automation of phenotypic assays to support quantitative drug efficacy, toxicity, and metabolism testing workflows, are covered. Continued advancement of the field is expected to help fully humanize the process of CNS drug discovery.
Collapse
Affiliation(s)
- M. Catarina Silva
- Chemical Neurobiology Laboratory, Departments of Neurology and Psychiatry, Massachusetts General Hospital, Center for Genomic Medicine, Harvard Medical School, Boston MA, USA
| | - Stephen J. Haggarty
- Chemical Neurobiology Laboratory, Departments of Neurology and Psychiatry, Massachusetts General Hospital, Center for Genomic Medicine, Harvard Medical School, Boston MA, USA
| |
Collapse
|
11
|
Abstract
With the seminal discovery of somatic cell reprogramming with defined genetic factors, it is now a routine laboratory procedure to reprogram somatic cells to generate patient-specific induced pluripotent stem cells (iPSCs) [1] Patient-specific iPSCs can be differentiated to generate mature neurons as well as three-dimensional brain organoids that show appropriate functional activity in electrophysiological studies [2,3]. However, there is a significant gap in the thoughtful incorporation of patient-derived neuronal cells in clinical studies addressing disease risk.
Collapse
Affiliation(s)
- Rakesh Karmacharya
- Program in Neuroscience and Chemical Biology, Center for Genomic Medicine, Massachusetts General Hospital & McLean Hospital, Harvard University, Boston, MA, USA.,Chemical Biology and Therapeutic Science Program, Broad Institute of Harvard & MIT, Cambridge, MA, USA
| | - Christian Kieling
- Department of Psychiatry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil.,Child & Adolescent Psychiatry Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - Valeria Mondelli
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom.,National Institute for Health Research Mental Health Biomedical Research Centre, South London and Maudsley NHS Foundation Trust, and King's College London,London, United Kingdom
| |
Collapse
|
12
|
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] [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
|
13
|
Kathuria A, Lopez-Lengowski K, Vater M, McPhie D, Cohen BM, Karmacharya R. Transcriptome analysis and functional characterization of cerebral organoids in bipolar disorder. Genome Med 2020; 12:34. [PMID: 32306996 PMCID: PMC7168850 DOI: 10.1186/s13073-020-00733-6] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 04/02/2020] [Indexed: 02/06/2023] Open
Abstract
Background Reprogramming human induced pluripotent stem cells (iPSCs) from somatic cells and generating three-dimensional brain organoids from these iPSCs provide access to live human neuronal tissue with disease-specific genetic backgrounds. Methods Cerebral organoids were generated from iPSCs of eight bipolar disorder (BPI) patients and eight healthy control individuals. RNA-seq experiments were undertaken using RNA isolated from the cerebral organoids. Functional activity in the cerebral organoids was studied using microelectrode arrays. Results RNA-seq data comparing gene expression profiles in the cerebral organoids showed downregulation of pathways involved in cell adhesion, neurodevelopment, and synaptic biology in bipolar disorder along with upregulation of genes involved in immune signaling. The central hub in the network analysis was neurocan (NCAN), which is located in a locus with evidence for genome-wide significant association in BPI. Gene ontology analyses suggested deficits related to endoplasmic reticulum biology in BPI, which was supported by cellular characterization of ER–mitochondria interactions. Functional studies with microelectrode arrays revealed specific deficits in response to stimulation and depolarization in BPI cerebral organoids. Conclusions Our studies in cerebral organoids from bipolar disorder showed dysregulation in genes involved in cell adhesion, immune signaling, and endoplasmic reticulum biology; implicated a central role for the GWAS hit NCAN in the biology of BPI; and showed evidence of deficits in neurotransmission.
Collapse
Affiliation(s)
- Annie Kathuria
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Chemical Biology Program, Broad Institute of MIT & Harvard, Cambridge, MA, USA.,Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Kara Lopez-Lengowski
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Chemical Biology Program, Broad Institute of MIT & Harvard, Cambridge, MA, USA
| | - Magdalena Vater
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Chemical Biology Program, Broad Institute of MIT & Harvard, Cambridge, MA, USA.,Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Donna McPhie
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA.,Schizophrenia and Bipolar Disorder Program, McLean Hospital, Belmont, MA, USA
| | - Bruce M Cohen
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA.,Schizophrenia and Bipolar Disorder Program, McLean Hospital, Belmont, MA, USA
| | - Rakesh Karmacharya
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA. .,Chemical Biology Program, Broad Institute of MIT & Harvard, Cambridge, MA, USA. .,Department of Psychiatry, Harvard Medical School, Boston, MA, USA. .,Schizophrenia and Bipolar Disorder Program, McLean Hospital, Belmont, MA, USA. .,Program in Neuroscience, Harvard University, Cambridge, MA, USA. .,Program in Chemical Biology, Harvard University, Cambridge, MA, USA. .,Harvard Stem Cell Institute, Cambridge, MA, USA.
| |
Collapse
|
14
|
Kathuria A, Lopez-Lengowski K, Watmuff B, McPhie D, Cohen BM, Karmacharya R. Synaptic deficits in iPSC-derived cortical interneurons in schizophrenia are mediated by NLGN2 and rescued by N-acetylcysteine. Transl Psychiatry 2019; 9:321. [PMID: 31780643 PMCID: PMC6882825 DOI: 10.1038/s41398-019-0660-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 09/06/2019] [Accepted: 10/20/2019] [Indexed: 02/08/2023] Open
Abstract
Human postmortem studies suggest a major role for abnormalities in GABAergic interneurons in the prefrontal cortex in schizophrenia. Cortical interneurons differentiated from induced pluripotent stem cells (iPSCs) of schizophrenia subjects showed significantly lower levels of glutamate decarboxylase 67 (GAD67), replicating findings from multiple postmortem studies, as well as reduced levels of synaptic proteins gehpyrin and NLGN2. Co-cultures of the interneurons with excitatory cortical pyramidal neurons from schizophrenia iPSCs showed reduced synaptic puncta density and lower action potential frequency. NLGN2 overexpression in schizophrenia neurons rescued synaptic puncta deficits while NLGN2 knockdown in healthy neurons resulted in reduced synaptic puncta density. Schizophrenia interneurons also had significantly smaller nuclear area, suggesting an innate oxidative stressed state. The antioxidant N-acetylcysteine increased the nuclear area in schizophrenia interneurons, increased NLGN2 expression and rescued synaptic deficits. These results implicate specific deficiencies in the synaptic machinery in cortical interneurons as critical regulators of synaptic connections in schizophrenia and point to a nexus between oxidative stress and NLGN2 expression in mediating synaptic deficits in schizophrenia.
Collapse
Affiliation(s)
- Annie Kathuria
- 0000 0004 0386 9924grid.32224.35Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA USA ,grid.66859.34Chemical Biology and Therapeutic Science Program, Broad Institute of MIT & Harvard, Cambridge, MA USA ,000000041936754Xgrid.38142.3cDepartment of Psychiatry, Harvard Medical School, Boston, MA USA
| | - Kara Lopez-Lengowski
- 0000 0004 0386 9924grid.32224.35Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA USA ,grid.66859.34Chemical Biology and Therapeutic Science Program, Broad Institute of MIT & Harvard, Cambridge, MA USA
| | - Bradley Watmuff
- 0000 0004 0386 9924grid.32224.35Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA USA ,grid.66859.34Chemical Biology and Therapeutic Science Program, Broad Institute of MIT & Harvard, Cambridge, MA USA ,000000041936754Xgrid.38142.3cDepartment of Psychiatry, Harvard Medical School, Boston, MA USA
| | - Donna McPhie
- 000000041936754Xgrid.38142.3cDepartment of Psychiatry, Harvard Medical School, Boston, MA USA ,0000 0000 8795 072Xgrid.240206.2Schizophrenia and Bipolar Disorder Program, McLean Hospital, Belmont, MA USA
| | - Bruce M. Cohen
- 000000041936754Xgrid.38142.3cDepartment of Psychiatry, Harvard Medical School, Boston, MA USA ,0000 0000 8795 072Xgrid.240206.2Schizophrenia and Bipolar Disorder Program, McLean Hospital, Belmont, MA USA
| | - Rakesh Karmacharya
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA. .,Chemical Biology and Therapeutic Science Program, Broad Institute of MIT & Harvard, Cambridge, MA, USA. .,Department of Psychiatry, Harvard Medical School, Boston, MA, USA. .,Schizophrenia and Bipolar Disorder Program, McLean Hospital, Belmont, MA, USA. .,Graduate Program in Chemical Biology, Harvard University, Cambridge, MA, USA. .,Program in Neuroscience, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
15
|
In Vitro Modeling of the Bipolar Disorder and Schizophrenia Using Patient-Derived Induced Pluripotent Stem Cells with Copy Number Variations of PCDH15 and RELN. eNeuro 2019; 6:ENEURO.0403-18.2019. [PMID: 31540999 PMCID: PMC6800292 DOI: 10.1523/eneuro.0403-18.2019] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 08/30/2019] [Accepted: 09/03/2019] [Indexed: 12/12/2022] Open
Abstract
Bipolar disorder (BP) and schizophrenia (SCZ) are major psychiatric disorders, but the molecular mechanisms underlying the complicated pathologies of these disorders remain unclear. It is difficult to establish adequate in vitro models for pathological analysis because of the heterogeneity of these disorders. In the present study, to recapitulate the pathologies of these disorders in vitro, we established in vitro models by differentiating mature neurons from human induced pluripotent stem cells (hiPSCs) derived from BP and SCZ patient with contributive copy number variations, as follows: two BP patients with PCDH15 deletion and one SCZ patient with RELN deletion. Glutamatergic neurons and GABAergic neurons were induced from hiPSCs under optimized conditions. Both types of induced neurons from both hiPSCs exhibited similar phenotypes of MAP2 (microtubule-associated protein 2)-positive dendrite shortening and decreasing synapse numbers. Additionally, we analyzed isogenic PCDH15- or RELN-deleted cells. The dendrite and synapse phenotypes of isogenic neurons were partially similar to those of patient-derived neurons. These results suggest that the observed phenotypes are general phenotypes of psychiatric disorders, and our in vitro models using hiPSC-based technology may be suitable for analysis of the pathologies of psychiatric disorders.
Collapse
|
16
|
Iaconelli J, Xuan L, Karmacharya R. HDAC6 Modulates Signaling Pathways Relevant to Synaptic Biology and Neuronal Differentiation in Human Stem-Cell-Derived Neurons. Int J Mol Sci 2019; 20:ijms20071605. [PMID: 30935091 PMCID: PMC6480207 DOI: 10.3390/ijms20071605] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 03/12/2019] [Accepted: 03/18/2019] [Indexed: 12/18/2022] Open
Abstract
Recent studies show that histone deacetylase 6 (HDAC6) has important roles in the human brain, especially in the context of a number of nervous system disorders. Animal models of neurodevelopmental, neurodegenerative, and neuropsychiatric disorders show that HDAC6 modulates important biological processes relevant to disease biology. Pan-selective histone deacetylase (HDAC) inhibitors had been studied in animal behavioral assays and shown to induce synaptogenesis in rodent neuronal cultures. While most studies of HDACs in the nervous system have focused on class I HDACs located in the nucleus (e.g., HDACs 1,2,3), recent findings in rodent models suggest that the cytoplasmic class IIb HDAC, HDAC6, plays an important role in regulating mood-related behaviors. Human studies suggest a significant role for synaptic dysfunction in the prefrontal cortex (PFC) and hippocampus in depression. Studies of HDAC inhibitors (HDACi) in human neuronal cells show that HDAC6 inhibitors (HDAC6i) increase the acetylation of specific lysine residues in proteins involved in synaptogenesis. This has led to the hypothesis that HDAC6i may modulate synaptic biology not through effects on the acetylation of histones, but by regulating acetylation of non-histone proteins.
Collapse
Affiliation(s)
- Jonathan Iaconelli
- Center for Genomic Medicine, Harvard Medical School and Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA.
- Chemical Biology Program, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA.
| | - Lucius Xuan
- Center for Genomic Medicine, Harvard Medical School and Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA.
- Chemical Biology Program, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA.
| | - Rakesh Karmacharya
- Center for Genomic Medicine, Harvard Medical School and Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA.
- Chemical Biology Program, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA.
- Schizophrenia and Bipolar Disorder Program, McLean Hospital, Belmont, MA 02478, USA.
- Program in Neuroscience, Harvard University, Cambridge, MA 02138, USA.
- Chemical Biology PhD Program, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
17
|
Zhou J, Dong XH, Zhang FZ, Zhu HM, Hao T, Jiang XX, Zheng WB, Zhang T, Wang PZ, Li H, Na J, Wang CY. Real microgravity condition promoted regeneration capacity of induced pluripotent stem cells during the TZ-1 space mission. Cell Prolif 2019; 52:e12574. [PMID: 30724402 PMCID: PMC6536455 DOI: 10.1111/cpr.12574] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) are reprogrammed somatic cells that gained self‐renewal and differentiation capacity similar to embryonic stem cells. Taking the precious opportunity of the TianZhou‐1 spacecraft mission, we studied the effect of space microgravity (µg) on the self‐renewal capacity of iPSCs. Murine iPSCs carrying pluripotency reporter Oct4‐GFP were used. The Oct4‐EGFP‐iPSCs clones were loaded into the bioreactor and exposed to μg in outer space for 14 days. The control experiment was performed in identical device but on the ground in earth gravity (1 g). iPSCs clones were compact and highly expressed Oct4 before launch. In μg condition, cells in iPSC clones spread out more rapidly than those in ground 1 g condition during the first 3 days after launch. However, in 1 g condition, as the cell density increases, the Oct4‐GFP signal dropped significantly during the following 3 days. Interestingly, in μg condition, iPSCs originated from the spread‐out clones during the first 3 days appeared to cluster together and reform colonies that activated strong Oct4 expression. On the other hand, iPSC clones in 1 g condition were not able to recover Oct4 expression after overgrown. Our study for the first time performed real‐time imaging on the proliferation process of iPSCs in space and found that in μg condition, cell behaviour appeared to be more dynamic than on the ground.
Collapse
Affiliation(s)
- Jin Zhou
- Tissue Engineering Research Center, Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing, China
| | - Xiao-Hui Dong
- Tissue Engineering Research Center, Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing, China
| | - Feng-Zhi Zhang
- Tissue Engineering Research Center, Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing, China
| | - Hui-Min Zhu
- Tissue Engineering Research Center, Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing, China
| | - Tong Hao
- Tissue Engineering Research Center, Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing, China
| | - Xiao-Xia Jiang
- Tissue Engineering Research Center, Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing, China
| | - Wei-Bo Zheng
- Shanghai Institute of Technical Physics, Chinese Academy of Sciences, Shanghai, China
| | - Tao Zhang
- Shanghai Institute of Technical Physics, Chinese Academy of Sciences, Shanghai, China
| | - Pei-Zhe Wang
- School of Medicine, Tsinghua University, Beijing, China
| | - Hong Li
- Tissue Engineering Research Center, Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing, China
| | - Jie Na
- School of Medicine, Tsinghua University, Beijing, China
| | - Chang-Yong Wang
- Tissue Engineering Research Center, Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing, China
| |
Collapse
|
18
|
Scalable Measurements of Intrinsic Excitability in Human iPS Cell-Derived Excitatory Neurons Using All-Optical Electrophysiology. Neurochem Res 2019; 44:714-725. [PMID: 30603979 DOI: 10.1007/s11064-018-2694-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 12/02/2018] [Accepted: 12/04/2018] [Indexed: 12/27/2022]
Abstract
Induced pluripotent stem (iPS) cells offer the exciting opportunity for modeling neurological disorders in vitro in the context of a human genetic background. While significant progress has been made in advancing the use of iPS cell-based disease models, there remains an unmet need to characterize the electrophysiological profile of individual neurons with sufficient throughput to enable statistically robust assessment of disease phenotypes and pharmacological modulation. Here, we describe the Optopatch platform technology that utilizes optogenetics to both stimulate and record action potentials (APs) from human iPS cell-derived excitatory neurons with similar information content to manual patch clamp electrophysiology, but with ~ 3 orders of magnitude greater throughput. Cortical excitatory neurons were produced using the NGN2 transcriptional programming approach and cultured in the presence of rodent glial cells. Characterization of the neuronal preparations using immunocytochemistry and qRT-PCR assays reveals an enrichment of neuronal and glutamatergic markers as well as select ion channels. We demonstrate the scale of our intrinsic cellular excitability assay using pharmacological assessment with select ion channel modulators quinidine and retigabine, by measuring changes in both spike timing and waveform properties. The Optopatch platform in human iPS cell-derived cortical excitatory neurons has the potential for detailed phenotype and pharmacology evaluation, which can serve as the basis of cellular disease model exploration for drug discovery and phenotypic screening efforts.
Collapse
|
19
|
van Hugte E, Nadif Kasri N. Modeling Psychiatric Diseases with Induced Pluripotent Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1192:297-312. [PMID: 31705501 DOI: 10.1007/978-981-32-9721-0_15] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neuropsychiatric disorders are a heterogeneous group of disorders that are challenging to model and treat, due to their underlying complex genetic architecture and clinical variability. Presently, increasingly more studies are making use of induced pluripotent stem cell (iPSC)-derived neurons, reprogrammed from patient somatic cells, to model neuropsychiatric disorders. iPSC-derived neurons offer the possibility to recapitulate relevant disease biology in the context of the individual patient genetic background. In addition to disease modeling, iPSC-derived neurons offer unprecedented opportunities in drug screening. In this chapter, the current status of iPSC disease modeling for neuropsychiatric disorders is presented. Both 2D and 3D disease modeling approaches are discussed as well as the generation of different neuronal cell types that are relevant for studying neuropsychiatric disorders. Moreover, the advantages and limitations are highlighted in addition to the future perspectives of using iPSC-derived neurons in the uncovering of robust cellular phenotypes that consecutively have the potential to lead to clinical developments.
Collapse
Affiliation(s)
- Eline van Hugte
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition, and Behaviour, 6500 HB, Nijmegen, The Netherlands
- Academic Center for Epileptology Kempenhaeghe, Heeze, The Netherlands
| | - Nael Nadif Kasri
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition, and Behaviour, 6500 HB, Nijmegen, The Netherlands.
- Department of Cognitive Neuroscience, Radboudumc, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands.
| |
Collapse
|
20
|
McPhie DL, Nehme R, Ravichandran C, Babb SM, Ghosh SD, Staskus A, Kalinowski A, Kaur R, Douvaras P, Du F, Ongur D, Fossati V, Eggan K, Cohen BM. Oligodendrocyte differentiation of induced pluripotent stem cells derived from subjects with schizophrenias implicate abnormalities in development. Transl Psychiatry 2018; 8:230. [PMID: 30352993 PMCID: PMC6199264 DOI: 10.1038/s41398-018-0284-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 09/26/2018] [Indexed: 12/22/2022] Open
Abstract
Abnormalities of brain connectivity and signal transduction are consistently observed in individuals with schizophrenias (SZ). Underlying these anomalies, convergent in vivo, post mortem, and genomic evidence suggest abnormal oligodendrocyte (OL) development and function and lower myelination in SZ. Our primary hypothesis was that there would be abnormalities in the number of induced pluripotent stem (iPS) cell-derived OLs from subjects with SZ. Our secondary hypothesis was that these in vitro abnormalities would correlate with measures of white matter (WM) integrity and myelination in the same subjects in vivo, estimated from magnetic resonance imaging. Six healthy control (HC) and six SZ iPS cell lines, derived from skin fibroblasts from well-characterized subjects, were differentiated into OLs. FACS analysis of the oligodendrocyte-specific surface, glycoprotein O4, was performed at three time points of development (days 65, 75, and 85) to quantify the number of late oligodendrocyte progenitor cells (OPCs) and OLs in each line. Significantly fewer O4-positive cells developed from SZ versus HC lines (95% CI 1.0: 8.6, F1,10 = 8.06, p = 0.02). The difference was greater when corrected for age (95% CI 5.4:10.4, F1,8 = 53.6, p < 0.001). A correlation between myelin content in WM in vivo, estimated by magnetization transfer ratio (MTR) and number of O4-positive cells in vitro was also observed across all time points (F1,9 = 4.3, p = 0.07), reaching significance for mature OLs at day 85 in culture (r = 0.70, p < 0.02). Low production of OPCs may be a contributing mechanism underlying WM reduction in SZ.
Collapse
Affiliation(s)
- Donna L. McPhie
- 000000041936754Xgrid.38142.3cHarvard Medical School, Boston, MA USA ,0000 0000 8795 072Xgrid.240206.2McLean Hospital, 115 Mill St., Belmont, MA 02478 USA
| | - Ralda Nehme
- grid.66859.34Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142 USA ,000000041936754Xgrid.38142.3cDepartment of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138 USA
| | - Caitlin Ravichandran
- 000000041936754Xgrid.38142.3cHarvard Medical School, Boston, MA USA ,0000 0000 8795 072Xgrid.240206.2McLean Hospital, 115 Mill St., Belmont, MA 02478 USA
| | - Suzann M. Babb
- 000000041936754Xgrid.38142.3cHarvard Medical School, Boston, MA USA ,0000 0000 8795 072Xgrid.240206.2McLean Hospital, 115 Mill St., Belmont, MA 02478 USA
| | - Sulagna Dia Ghosh
- grid.66859.34Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142 USA ,000000041936754Xgrid.38142.3cDepartment of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138 USA
| | - Alexandra Staskus
- 0000 0000 8795 072Xgrid.240206.2McLean Hospital, 115 Mill St., Belmont, MA 02478 USA
| | - Amy Kalinowski
- 0000 0000 8795 072Xgrid.240206.2McLean Hospital, 115 Mill St., Belmont, MA 02478 USA
| | - Rupinderjit Kaur
- 0000 0000 8795 072Xgrid.240206.2McLean Hospital, 115 Mill St., Belmont, MA 02478 USA
| | - Panagiotis Douvaras
- Blue Rock Therapeutics, Alexandria Center for Life Science, 450 E 29th Street, Suite 504, New York, NY 10016 USA
| | - Fei Du
- 000000041936754Xgrid.38142.3cHarvard Medical School, Boston, MA USA ,0000 0000 8795 072Xgrid.240206.2McLean Hospital, 115 Mill St., Belmont, MA 02478 USA
| | - Dost Ongur
- 000000041936754Xgrid.38142.3cHarvard Medical School, Boston, MA USA ,0000 0000 8795 072Xgrid.240206.2McLean Hospital, 115 Mill St., Belmont, MA 02478 USA
| | - Valentina Fossati
- 0000 0004 5906 3313grid.430819.7The New York Stem Cell Foundation Research Institute, 619 West 54th Street, 3rd Floor, New York, NY 10019 USA
| | - Kevin Eggan
- 000000041936754Xgrid.38142.3cDepartment of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138 USA ,grid.66859.34Stanley Center for Psychiatric Research and Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA 02142 USA
| | - Bruce M. Cohen
- 000000041936754Xgrid.38142.3cHarvard Medical School, Boston, MA USA ,0000 0000 8795 072Xgrid.240206.2McLean Hospital, 115 Mill St., Belmont, MA 02478 USA
| |
Collapse
|
21
|
Liu J, Chen J, Perrone-Bizzozero N, Calhoun VD. A Perspective of the Cross-Tissue Interplay of Genetics, Epigenetics, and Transcriptomics, and Their Relation to Brain Based Phenotypes in Schizophrenia. Front Genet 2018; 9:343. [PMID: 30190726 PMCID: PMC6115489 DOI: 10.3389/fgene.2018.00343] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 08/09/2018] [Indexed: 12/11/2022] Open
Abstract
Genetic association studies of psychiatric disorders have provided unprecedented insight into disease risk profiles with high confidence. Yet, the next research challenge is how to translate this rich information into mechanisms of disease, and further help interventions and treatments. Given other comprehensive reviews elsewhere, here we want to discuss the research approaches that integrate information across various tissue types. Taking schizophrenia as an example, the tissues, cells, or organisms being investigated include postmortem brain tissues or neurons, peripheral blood and saliva, in vivo brain imaging, and in vitro cell lines, particularly human induced pluripotent stem cells (iPSC) and iPSC derived neurons. There is a wealth of information on the molecular signatures including genetics, epigenetics, and transcriptomics of various tissues, along with neuronal phenotypic measurements including neuronal morphometry and function, together with brain imaging and other techniques that provide data from various spatial temporal points of disease development. Through consistent or complementary processes across tissues, such as cross-tissue methylation quantitative trait loci (QTL) and expression QTL effects, systemic integration of such information holds the promise to put the pieces of puzzle together for a more complete view of schizophrenia disease pathogenesis.
Collapse
Affiliation(s)
- Jingyu Liu
- Mind Research Network, Lovelace Biomedical and Environmental Research Institute, Albuquerque, NM, United States
- Department of Neurosciences, University of New Mexico, Albuquerque, NM, United States
| | - Jiayu Chen
- Mind Research Network, Lovelace Biomedical and Environmental Research Institute, Albuquerque, NM, United States
| | - Nora Perrone-Bizzozero
- Department of Electrical and Computer Engineering, University of New Mexico, Albuquerque, NM, United States
| | - Vince D. Calhoun
- Mind Research Network, Lovelace Biomedical and Environmental Research Institute, Albuquerque, NM, United States
- Department of Neurosciences, University of New Mexico, Albuquerque, NM, United States
| |
Collapse
|
22
|
Kucharska-Mazur J, Jabłoński M, Misiak B, Frydecka D, Rybakowski J, Ratajczak MZ, Samochowiec J. Adult stem cells in psychiatric disorders - New discoveries in peripheral blood. Prog Neuropsychopharmacol Biol Psychiatry 2018; 80:23-27. [PMID: 28392482 DOI: 10.1016/j.pnpbp.2017.04.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 03/28/2017] [Accepted: 04/05/2017] [Indexed: 12/15/2022]
Abstract
The new area of research in psychiatric disorders is concerned with abnormal regeneration processes. The role of brain neurogenesis has been studied for decades. New discoveries, concerned with the pluripotency of VSEL cells and the role of factors involved in stem cell trafficking in peripheral blood create hope that it will be possible to develop a better understanding of the processes of neuroregeneration/neurodegeneration. There is an ongoing research investigating concentrations of: sphingosine -1-phosphate, SDF-1, elements of complement cascade, and stem cells in peripheral blood, including their possible connection to psychiatric disorders. Collected data, suggesting an abnormal course of regeneration processes in psychiatric disorders, raises hope of finding new potential markers of psychosis and anxiety disorders.
Collapse
Affiliation(s)
- Jolanta Kucharska-Mazur
- Department of Psychiatry, Pomeranian University of Medicine, Broniewskiego 26, 71-460 Szczecin, Poland
| | - Marcin Jabłoński
- Department of Psychiatry, Pomeranian University of Medicine, Broniewskiego 26, 71-460 Szczecin, Poland
| | - Błażej Misiak
- Department of Genetics, Wroclaw Medical University, Marcinkowskiego 1, 50-368 Wrocław, Poland
| | - Dorota Frydecka
- Department of Psychiatry, Wroclaw Medical University, Pasteur 10, 50-367 Wroclaw, Poland
| | - Janusz Rybakowski
- Department of Adult Psychiatry, Poznan University of Medical Sciences, Szpitalna 27/33, 60-572 Poznan, Poland
| | | | - Jerzy Samochowiec
- Department of Psychiatry, Pomeranian University of Medicine, Broniewskiego 26, 71-460 Szczecin, Poland.
| |
Collapse
|
23
|
Iaconelli J, Lalonde J, Watmuff B, Liu B, Mazitschek R, Haggarty SJ, Karmacharya R. Lysine Deacetylation by HDAC6 Regulates the Kinase Activity of AKT in Human Neural Progenitor Cells. ACS Chem Biol 2017. [PMID: 28628306 DOI: 10.1021/acschembio.6b01014] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The AKT family of serine-threonine kinases functions downstream of phosphatidylinositol 3-kinase (PI3K) to transmit signals by direct phosphorylation of a number of targets, including the mammalian target of rapamycin (mTOR), glycogen synthase kinase 3β (GSK3β), and β-catenin. AKT binds to phosphatidylinositol (3,4,5)-triphosphate (PIP3) generated by PI3K activation, which results in its membrane localization and subsequent activation through phosphorylation by phosphoinositide-dependent protein kinase 1 (PDK1). Together, the PI3K-AKT signaling pathway plays pivotal roles in many cellular systems, including in the central nervous system where it governs both neurodevelopment and neuroplasticity. Recently, lysine residues (Lys14 and Lys20) on AKT, located within its pleckstrin homology (PH) domain that binds to membrane-bound PIP3, have been found to be acetylated under certain cellular contexts in various cancer cell lines. These acetylation modifications are removed by the enzymatic action of the class III lysine deacetylases, SIRT1 and SIRT2, of the sirtuin family. The extent to which reversible acetylation regulates AKT function in other cell types remains poorly understood. We report here that AKT kinase activity is modulated by a class IIb lysine deacetylase, histone deacetylase 6 (HDAC6), in human neural progenitor cells (NPCs). We find that HDAC6 and AKT physically interact with each other in the neuronal cells, and in the presence of selective HDAC6 inhibition, AKT is acetylated at Lys163 and Lys377 located in the kinase domain, two novel sites distinct from the acetylation sites in the PH-domain modulated by the sirtuins. Measurement of the functional effect of HDAC6 inhibition on AKT revealed decreased binding to PIP3, a correlated decrease in AKT kinase activity, decreased phosphorylation of Ser552 on β-catenin, and modulation of neuronal differentiation trajectories. Taken together, our studies implicate the deacetylase activity of HDAC6 as a novel regulator of AKT signaling and point to novel mechanisms for regulating AKT activity with small-molecule inhibitors of HDAC6 currently under clinical development.
Collapse
Affiliation(s)
- Jonathan Iaconelli
- Center for Experimental Drugs and Diagnostics, Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts 02114, United States
- Chemical Biology Program, Broad Institute of Harvard and MIT, Cambridge, Massachusetts 02142, United States
| | - Jasmin Lalonde
- Center for Experimental Drugs and Diagnostics, Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts 02114, United States
- Department of Neurology, Harvard Medical School, Boston, Massachusetts 02115, United States
- Chemical Neurobiology Laboratory, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
| | - Bradley Watmuff
- Center for Experimental Drugs and Diagnostics, Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts 02114, United States
- Chemical Biology Program, Broad Institute of Harvard and MIT, Cambridge, Massachusetts 02142, United States
| | - Bangyan Liu
- Center for Experimental Drugs and Diagnostics, Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts 02114, United States
| | - Ralph Mazitschek
- Center for Systems Biology, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts 02114, United States
- Infectious Diseases Program, Broad Institute of Harvard and MIT, Cambridge, Massachusetts 02142, United States
| | - Stephen J. Haggarty
- Center for Experimental Drugs and Diagnostics, Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts 02114, United States
- Department of Neurology, Harvard Medical School, Boston, Massachusetts 02115, United States
- Chemical Neurobiology Laboratory, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
| | - Rakesh Karmacharya
- Center for Experimental Drugs and Diagnostics, Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts 02114, United States
- Chemical Biology Program, Broad Institute of Harvard and MIT, Cambridge, Massachusetts 02142, United States
- Schizophrenia and Bipolar Disorder Program, McLean Hospital, Belmont, Massachusetts 02478, United States
| |
Collapse
|
24
|
Dimitrion P, Zhi Y, Clayton D, Apodaca GL, Wilcox MR, Johnson JW, Nimgaonkar V, D'Aiuto L. Low-Density Neuronal Cultures from Human Induced Pluripotent Stem Cells. MOLECULAR NEUROPSYCHIATRY 2017; 3:28-36. [PMID: 28879199 DOI: 10.1159/000476034] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 04/21/2017] [Indexed: 12/31/2022]
Abstract
Induced pluripotent stem cell (iPSC)-based technologies offer an unprecedented possibility to investigate defects occurring during neuronal differentiation in neuropsychiatric and neurodevelopmental disorders, but the density and intricacy of intercellular connections in neuronal cultures challenge currently available analytic methods. Low-density neuronal cultures facilitate the morphometric and functional analysis of neurons. We describe a differentiation protocol to generate low-density neuronal cultures (∼2,500 neurons/cm2) from human iPSC-derived neural stem cells/early neural progenitor cells. We generated low-density cultures using cells from 3 individuals. We also evaluated the morphometric features of neurons derived from 2 of these individuals, one harboring a microdeletion on chromosome 15q11.2 and the other without the microdeletion. An approximately 7.5-fold increase in the density of dendritic filopodia was observed in the neurons with the microdeletion, consistent with previous reports. Low-density neuronal cultures enable facile and unbiased comparisons of iPSC-derived neurons from different individuals or clones.
Collapse
Affiliation(s)
- Peter Dimitrion
- Department of Psychiatry, Western Psychiatric Institute and Clinic, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Yun Zhi
- Department of Pharmacology and Pharmaceutical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Dennis Clayton
- Division of Renal-Electrolyte, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Gerard L Apodaca
- Division of Renal-Electrolyte, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Madeleine R Wilcox
- Department of Neuroscience and Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jon W Johnson
- Department of Neuroscience and Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Vishwajit Nimgaonkar
- Department of Psychiatry, Western Psychiatric Institute and Clinic, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Leonardo D'Aiuto
- Department of Psychiatry, Western Psychiatric Institute and Clinic, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
25
|
Watmuff B, Liu B, Karmacharya R. Stem cell-derived neurons in the development of targeted treatment for schizophrenia and bipolar disorder. Pharmacogenomics 2017; 18:471-479. [PMID: 28346060 DOI: 10.2217/pgs-2016-0187] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The recent advent of induced pluripotent stem cells has enabled the study of patient-specific and disease-related neurons in vitro and has facilitated new directions of inquiry into disease mechanisms. With these approaches, we now have the possibility of correlating ex vivo cellular phenotypes with individual patient response to treatment and/or side effects, which makes targeted treatments for schizophrenia and bipolar disorder a distinct prospect in the coming years. Here, we briefly review the current state of stem cell-based models and explore studies that are providing new insights into the disease biology of schizophrenia and bipolar disorder, which are laying the foundations for the development of novel targeted therapies.
Collapse
Affiliation(s)
- Bradley Watmuff
- Center for Experimental Drugs & Diagnostics, Psychiatric & Neurodevelopmental Genetics Unit, Center for Human Genetic Research, Harvard Medical School & Massachusetts General Hospital, Boston, MA 02114, USA.,Chemical Biology Program, Broad Institute of Harvard & MIT, Cambridge, MA 02142, USA
| | - Bangyan Liu
- Center for Experimental Drugs & Diagnostics, Psychiatric & Neurodevelopmental Genetics Unit, Center for Human Genetic Research, Harvard Medical School & Massachusetts General Hospital, Boston, MA 02114, USA
| | - Rakesh Karmacharya
- Center for Experimental Drugs & Diagnostics, Psychiatric & Neurodevelopmental Genetics Unit, Center for Human Genetic Research, Harvard Medical School & Massachusetts General Hospital, Boston, MA 02114, USA.,Chemical Biology Program, Broad Institute of Harvard & MIT, Cambridge, MA 02142, USA.,Schizophrenia & Bipolar Disorder Program, McLean Hospital, Belmont, MA 02478, USA
| |
Collapse
|
26
|
Huang JH, Park H, Iaconelli J, Berkovitch SS, Watmuff B, McPhie D, Öngür D, Cohen BM, Clish CB, Karmacharya R. Unbiased Metabolite Profiling of Schizophrenia Fibroblasts under Stressful Perturbations Reveals Dysregulation of Plasmalogens and Phosphatidylcholines. J Proteome Res 2016; 16:481-493. [DOI: 10.1021/acs.jproteome.6b00628] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Joanne H. Huang
- Center
for Experimental Drugs and Diagnostics, Psychiatric and Neurodevelopmental
Genetics Unit, Center for Human Genetic Research, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts 02114, United States
- Chemical
Biology Program, Broad Institute of Harvard and MIT, Cambridge, Massachusetts 02142, United States
| | - Hyoungjun Park
- Institute
of Neuroinformatics, ETH Zurich and University of Zurich, CH-8057, Zurich, Switzerland
| | - Jonathan Iaconelli
- Center
for Experimental Drugs and Diagnostics, Psychiatric and Neurodevelopmental
Genetics Unit, Center for Human Genetic Research, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts 02114, United States
- Chemical
Biology Program, Broad Institute of Harvard and MIT, Cambridge, Massachusetts 02142, United States
| | - Shaunna S. Berkovitch
- Center
for Experimental Drugs and Diagnostics, Psychiatric and Neurodevelopmental
Genetics Unit, Center for Human Genetic Research, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts 02114, United States
- Chemical
Biology Program, Broad Institute of Harvard and MIT, Cambridge, Massachusetts 02142, United States
| | - Bradley Watmuff
- Center
for Experimental Drugs and Diagnostics, Psychiatric and Neurodevelopmental
Genetics Unit, Center for Human Genetic Research, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts 02114, United States
- Chemical
Biology Program, Broad Institute of Harvard and MIT, Cambridge, Massachusetts 02142, United States
| | - Donna McPhie
- Schizophrenia
and Bipolar Disorder Program, Harvard Medical School and McLean Hospital, Belmont, Massachusetts 02478, United States
| | - Dost Öngür
- Schizophrenia
and Bipolar Disorder Program, Harvard Medical School and McLean Hospital, Belmont, Massachusetts 02478, United States
| | - Bruce M. Cohen
- Schizophrenia
and Bipolar Disorder Program, Harvard Medical School and McLean Hospital, Belmont, Massachusetts 02478, United States
| | - Clary B. Clish
- Chemical
Biology Program, Broad Institute of Harvard and MIT, Cambridge, Massachusetts 02142, United States
| | - Rakesh Karmacharya
- Center
for Experimental Drugs and Diagnostics, Psychiatric and Neurodevelopmental
Genetics Unit, Center for Human Genetic Research, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts 02114, United States
- Chemical
Biology Program, Broad Institute of Harvard and MIT, Cambridge, Massachusetts 02142, United States
- Schizophrenia
and Bipolar Disorder Program, Harvard Medical School and McLean Hospital, Belmont, Massachusetts 02478, United States
| |
Collapse
|
27
|
Wimalasena NK, Le VQ, Wimalasena K, Schreiber SL, Karmacharya R. Gene Expression-Based Screen for Parkinson's Disease Identifies GW8510 as a Neuroprotective Agent. ACS Chem Neurosci 2016; 7:857-63. [PMID: 27270122 DOI: 10.1021/acschemneuro.6b00076] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
We carried out a gene expression-based in silico screen in order to identify small molecules with gene-expression profiles that are anticorrelated with a gene-expression profile for Parkinson's disease (PD). We identified the cyclin-dependent kinase 2/5 (CDK2/5) inhibitor GW8510 as our most significant hit and characterized its effects in rodent MN9D cells and in human neuronal cells derived from induced pluripotent stem cells. GW8510 demonstrated neuroprotective ability in MN9D cells in the presence of 1-methyl-4-phenylpyridium (MPP(+)), a widely used neurotoxin model for Parkinson's disease. In order to delineate the nature and extent of GW8510's neuroprotective properties, we studied GW8510 in human neuronal cells in the context of various mechanisms of cellular stress. We found that GW8510 was protective against small-molecule mitochondrial and endoplasmic reticulum stressors. Our findings illustrate an approach to using small-molecule gene expression libraries to identify compounds with therapeutic potential in human diseases.
Collapse
Affiliation(s)
- Nivanthika K. Wimalasena
- Center for the Science of Therapeutics, Broad Institute of Harvard and MIT, Cambridge, Massachusetts 02142, United States
- Center
for Experimental Drugs and Diagnostics, Psychiatric and Neurodevelopmental
Genetics Unit, Center for Human Genetic Research, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts 02114, United States
- Program in Neuroscience, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Viet Q. Le
- Department of Science and Mathematics,
National Technical Institute for the Deaf, Rochester Institute of Technology, Rochester, New York 14623, United States
| | - Kandatege Wimalasena
- Department of Chemistry, Wichita State University, Wichita, Kansas 67260, United States
| | - Stuart L. Schreiber
- Center for the Science of Therapeutics, Broad Institute of Harvard and MIT, Cambridge, Massachusetts 02142, United States
- Howard Hughes Medical Institute, Department of Chemistry and Chemical
Biology, Harvard University, Cambridge, Massachusetts 02138, United States
| | - Rakesh Karmacharya
- Center for the Science of Therapeutics, Broad Institute of Harvard and MIT, Cambridge, Massachusetts 02142, United States
- Center
for Experimental Drugs and Diagnostics, Psychiatric and Neurodevelopmental
Genetics Unit, Center for Human Genetic Research, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts 02114, United States
- Schizophrenia and Bipolar Disorder Program, Harvard Medical School and McLean Hospital, Belmont, Massachusetts 02478, United States
| |
Collapse
|
28
|
Affiliation(s)
- Rakesh Karmacharya
- Center for Human Genetic Research, Harvard Medical School and Massachusetts General Hospital, Boston, MA 02114, United States; Center for the Science of Therapeutics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, United States; Schizophrenia and Bipolar Disorder Program, McLean Hospital, Belmont, MA 02478, United States.
| | - Stephen J Haggarty
- Center for Human Genetic Research, Harvard Medical School and Massachusetts General Hospital, Boston, MA 02114, United States; Center for Experimental Drugs & Diagnostics, Departments of Psychiatry & Neurology, Massachusetts General Hospital, Boston, MA 02114, United States
| |
Collapse
|