1
|
Pandamooz S, Salehi MS, Jurek B, Meinung CP, Azarpira N, Dianatpour M, Neumann ID. Oxytocin Receptor Expression in Hair Follicle Stem Cells: A Promising Model for Biological and Therapeutic Discovery in Neuropsychiatric Disorders. Stem Cell Rev Rep 2023; 19:2510-2524. [PMID: 37548806 DOI: 10.1007/s12015-023-10603-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/27/2023] [Indexed: 08/08/2023]
Abstract
The intricate nature of the human brain and the limitations of existing model systems to study molecular and cellular causes of neuropsychiatric disorders represent a major challenge for basic research. The promising progress in patient-derived stem cell technology and in our knowledge on the role of the brain oxytocin (OXT) system in health and disease offer new possibilities in that direction. In this study, the rat hair follicle stem cells (HFSCs) were isolated and expanded in vitro. The expression of oxytocin receptors (OXTR) was evaluated in these cells. The cellular viability was assessed 12 h post stimulation with OXT. The activation of OXTR-coupled intracellular signaling cascades, following OXT treatment was determined. Also, the influence of OXT on neurite outgrowth and cytoskeletal rearrangement were defined. The assessment of OXTR protein expression revealed this receptor is expressed abundantly in HFSCs. As evidenced by the cell viability assay, no adverse or cytotoxic effects were detected following 12 h treatment with different concentrations of OXT. Moreover, OXTR stimulation by OXT resulted in ERK1/2, CREB, and eEF2 activation, neurite length alterations, and cytoskeletal rearrangements that reveal the functionality of this receptor in HFSCs. Here, we introduced the rat HFSCs as an easy-to-obtain stem cell model that express functional OXTR. This cell-based model can contribute to our understanding of the progression and treatment of neuropsychiatric disorders with oxytocinergic system deficiency.
Collapse
Affiliation(s)
- Sareh Pandamooz
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Molecular and Behavioural Neurobiology, University of Regensburg, Regensburg, Germany
| | - Mohammad Saied Salehi
- Department of Molecular and Behavioural Neurobiology, University of Regensburg, Regensburg, Germany.
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Benjamin Jurek
- Department of Molecular and Behavioural Neurobiology, University of Regensburg, Regensburg, Germany
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, Munich, Germany
| | - Carl-Philipp Meinung
- Department of Molecular and Behavioural Neurobiology, University of Regensburg, Regensburg, Germany
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehdi Dianatpour
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Inga D Neumann
- Department of Molecular and Behavioural Neurobiology, University of Regensburg, Regensburg, Germany.
| |
Collapse
|
2
|
Yang L, Hung LY, Zhu Y, Ding S, Margolis KG, Leong KW. Material Engineering in Gut Microbiome and Human Health. RESEARCH (WASHINGTON, D.C.) 2022; 2022:9804014. [PMID: 35958108 PMCID: PMC9343081 DOI: 10.34133/2022/9804014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 06/10/2022] [Indexed: 12/11/2022]
Abstract
Tremendous progress has been made in the past decade regarding our understanding of the gut microbiome's role in human health. Currently, however, a comprehensive and focused review marrying the two distinct fields of gut microbiome and material research is lacking. To bridge the gap, the current paper discusses critical aspects of the rapidly emerging research topic of "material engineering in the gut microbiome and human health." By engaging scientists with diverse backgrounds in biomaterials, gut-microbiome axis, neuroscience, synthetic biology, tissue engineering, and biosensing in a dialogue, our goal is to accelerate the development of research tools for gut microbiome research and the development of therapeutics that target the gut microbiome. For this purpose, state-of-the-art knowledge is presented here on biomaterial technologies that facilitate the study, analysis, and manipulation of the gut microbiome, including intestinal organoids, gut-on-chip models, hydrogels for spatial mapping of gut microbiome compositions, microbiome biosensors, and oral bacteria delivery systems. In addition, a discussion is provided regarding the microbiome-gut-brain axis and the critical roles that biomaterials can play to investigate and regulate the axis. Lastly, perspectives are provided regarding future directions on how to develop and use novel biomaterials in gut microbiome research, as well as essential regulatory rules in clinical translation. In this way, we hope to inspire research into future biomaterial technologies to advance gut microbiome research and gut microbiome-based theragnostics.
Collapse
Affiliation(s)
- Letao Yang
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Lin Y. Hung
- Department of Pediatrics, Columbia University, New York, New York, USA
| | - Yuefei Zhu
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Suwan Ding
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Kara G. Margolis
- Department of Pediatrics, Columbia University, New York, New York, USA
| | - Kam W. Leong
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Department of Systems Biology, Columbia University, New York, NY, USA
| |
Collapse
|
3
|
Zhang W, Ross PJ, Ellis J, Salter MW. Targeting NMDA receptors in neuropsychiatric disorders by drug screening on human neurons derived from pluripotent stem cells. Transl Psychiatry 2022; 12:243. [PMID: 35680847 PMCID: PMC9184461 DOI: 10.1038/s41398-022-02010-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 05/25/2022] [Accepted: 05/27/2022] [Indexed: 01/04/2023] Open
Abstract
NMDA receptors (NMDARs), a prominent subtype of glutamatergic receptors, are implicated in the pathogenesis and development of neuropsychiatric disorders such as epilepsy, intellectual disability, autism spectrum disorder, and schizophrenia, and are therefore a potential therapeutic target in treating these disorders. Neurons derived from induced pluripotent stem cells (iPSCs) have provided the opportunity to investigate human NMDARs in their native environment. In this review, we describe the expression, function, and regulation of NMDARs in human iPSC-derived neurons and discuss approaches for utilizing human neurons for identifying potential drugs that target NMDARs in the treatment of neuropsychiatric disorders. A challenge in studying NMDARs in human iPSC-derived neurons is a predominance of those receptors containing the GluN2B subunit and low synaptic expression, suggesting a relatively immature phenotype of these neurons and delayed development of functional NMDARs. We outline potential approaches for improving neuronal maturation of human iPSC-derived neurons and accelerating the functional expression of NMDARs. Acceleration of functional expression of NMDARs in human iPSC-derived neurons will improve the modeling of neuropsychiatric disorders and facilitate the discovery and development of novel therapeutics targeting NMDARs for the treatment of these disorders.
Collapse
Affiliation(s)
- Wenbo Zhang
- Program in Neurosciences & Mental Health, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
- Department of Physiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - P Joel Ross
- Biology Department, University of Prince Edward Island, Charlottetown, PE, C1A 4P3, Canada
| | - James Ellis
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, M5S 1A8, Canada
| | - Michael W Salter
- Program in Neurosciences & Mental Health, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada.
- Department of Physiology, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
4
|
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: 47] [Impact Index Per Article: 11.8] [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
|
5
|
Fantuzzo JA, Robles DA, Mirabella VR, Hart RP, Pang ZP, Zahn JD. Development of a high-throughput arrayed neural circuitry platform using human induced neurons for drug screening applications. LAB ON A CHIP 2020; 20:1140-1152. [PMID: 32064487 PMCID: PMC7339603 DOI: 10.1039/c9lc01179j] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Proper brain function relies on the precise arrangement and flow of information between diverse neural subtypes. Developing improved human cell-based models which faithfully mimic biologically relevant connectivity patterns may improve drug screening efforts given the limited success of animal models to predict safety and efficacy of therapeutics in human clinical trials. To address this need, we have developed experimental models of defined neural circuitries through the compartmentalization of neuronal cell subtypes in a 96 well plate-based platform where each microwell is divided into two compartments connected by microchannels allowing high-throughput screening (HTS) of small molecules. We demonstrate that we can generate subtype-specific excitatory and inhibitory induced neuronal cells (iNs) from human stem cell lines and that these neurons form robust functional circuits with defined connectivity. Through the use of the genetically encoded calcium indicator GCaMP6f, we monitor calcium ion transients generated during neuronal firing between and within compartments. We further demonstrate functionality of the circuit by perturbing network activity through the addition of glutamate receptor blockers using automated liquid handling. Lastly, we show that we can stimulate network activity in defined neuronal subtypes through the expression of the designer receptor exclusively activated by designer drugs (DREADD) hM3Dq and application of the ligand clozapine-N-oxide (CNO). Our results demonstrate the formation of functional neural circuits in a high-throughput platform that is compatible with compound screening, representing an important step towards developing new screening platforms for studying and ultimately treating psychiatric brain disorders that arise from disordered neural circuit function.
Collapse
Affiliation(s)
- Joseph A Fantuzzo
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, USA. and Child Health Institute of New Jersey, Robert Wood Johnson Medical School, 89 French Street, New Brunswick, NJ 08901, USA
| | - Denise A Robles
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, USA.
| | - Vincent R Mirabella
- Child Health Institute of New Jersey, Robert Wood Johnson Medical School, 89 French Street, New Brunswick, NJ 08901, USA and Department of Neuroscience and Cell Biology, 675 Hoes Lane West, Research Tower, Third Floor, Piscataway, NJ 08854, USA and Pediatrics, Robert Wood Johnson Medical School, Rutgers University, One Robert Wood Johnson Place, MEB Third, PO Box 19, New Brunswick, NJ 08903, USA
| | - Ronald P Hart
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854, USA
| | - Zhiping P Pang
- Child Health Institute of New Jersey, Robert Wood Johnson Medical School, 89 French Street, New Brunswick, NJ 08901, USA and Department of Neuroscience and Cell Biology, 675 Hoes Lane West, Research Tower, Third Floor, Piscataway, NJ 08854, USA and Pediatrics, Robert Wood Johnson Medical School, Rutgers University, One Robert Wood Johnson Place, MEB Third, PO Box 19, New Brunswick, NJ 08903, USA
| | - Jeffrey D Zahn
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, USA.
| |
Collapse
|
6
|
Unprecedented Potential for Neural Drug Discovery Based on Self-Organizing hiPSC Platforms. Molecules 2020; 25:molecules25051150. [PMID: 32143423 PMCID: PMC7179160 DOI: 10.3390/molecules25051150] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/29/2020] [Accepted: 03/02/2020] [Indexed: 12/12/2022] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) have transformed conventional drug discovery pathways in recent years. In particular, recent advances in hiPSC biology, including organoid technologies, have highlighted a new potential for neural drug discovery with clear advantages over the use of primary tissues. This is important considering the financial and social burden of neurological health care worldwide, directly impacting the life expectancy of many populations. Patient-derived iPSCs-neurons are invaluable tools for novel drug-screening and precision medicine approaches directly aimed at reducing the burden imposed by the increasing prevalence of neurological disorders in an aging population. 3-Dimensional self-assembled or so-called ‘organoid’ hiPSCs cultures offer key advantages over traditional 2D ones and may well be gamechangers in the drug-discovery quest for neurological disorders in the coming years.
Collapse
|
7
|
Zhao WN, Tobe BTD, Udeshi ND, Xuan LL, Pernia CD, Zolg DP, Roberts AJ, Mani D, Blumenthal SR, Kurtser I, Patnaik D, Gaisina I, Bishop J, Sheridan SD, Lalonde J, Carr SA, Snyder EY, Haggarty SJ. Discovery of suppressors of CRMP2 phosphorylation reveals compounds that mimic the behavioral effects of lithium on amphetamine-induced hyperlocomotion. Transl Psychiatry 2020; 10:76. [PMID: 32094324 PMCID: PMC7039883 DOI: 10.1038/s41398-020-0753-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 11/08/2019] [Accepted: 11/21/2019] [Indexed: 12/12/2022] Open
Abstract
The effective treatment of bipolar disorder (BD) represents a significant unmet medical need. Although lithium remains a mainstay of treatment for BD, limited knowledge regarding how it modulates affective behavior has proven an obstacle to discovering more effective mood stabilizers with fewer adverse side effects. One potential mechanism of action of lithium is through inhibition of the serine/threonine protein kinase GSK3β, however, relevant substrates whose change in phosphorylation may mediate downstream changes in neuroplasticity remain poorly understood. Here, we used human induced pluripotent stem cell (hiPSC)-derived neuronal cells and stable isotope labeling by amino acids in cell culture (SILAC) along with quantitative mass spectrometry to identify global changes in the phosphoproteome upon inhibition of GSK3α/β with the highly selective, ATP-competitive inhibitor CHIR-99021. Comparison of phosphorylation changes to those induced by therapeutically relevant doses of lithium treatment led to the identification of collapsin response mediator protein 2 (CRMP2) as being highly sensitive to both treatments as well as an extended panel of structurally distinct GSK3α/β inhibitors. On this basis, a high-content image-based assay in hiPSC-derived neurons was developed to screen diverse compounds, including FDA-approved drugs, for their ability to mimic lithium's suppression of CRMP2 phosphorylation without directly inhibiting GSK3β kinase activity. Systemic administration of a subset of these CRMP2-phosphorylation suppressors were found to mimic lithium's attenuation of amphetamine-induced hyperlocomotion in mice. Taken together, these studies not only provide insights into the neural substrates regulated by lithium, but also provide novel human neuronal assays for supporting the development of mechanism-based therapeutics for BD and related neuropsychiatric disorders.
Collapse
Affiliation(s)
- Wen-Ning Zhao
- grid.32224.350000 0004 0386 9924Chemical Neurobiology Laboratory, Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114 USA ,grid.32224.350000 0004 0386 9924Departments of Psychiatry & Neurology, Massachusetts General Hospital & Harvard Medical School, Boston, MA 02114 USA
| | - Brian T. D. Tobe
- grid.479509.60000 0001 0163 8573Center for Stem Cells & Regenerative Medicine, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037 USA ,grid.266100.30000 0001 2107 4242Department of Psychiatry, University of California San Diego, La Jolla, CA 92037 USA ,grid.468218.1Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037 USA ,Present Address: Kaiser Health, San Diego, CA USA
| | - Namrata D. Udeshi
- grid.38142.3c000000041936754XProteomics Platform, Broad Institute of MIT and Harvard University, Cambridge, MA 02142 USA
| | - Lucius L. Xuan
- grid.32224.350000 0004 0386 9924Chemical Neurobiology Laboratory, Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114 USA ,grid.32224.350000 0004 0386 9924Departments of Psychiatry & Neurology, Massachusetts General Hospital & Harvard Medical School, Boston, MA 02114 USA
| | - Cameron D. Pernia
- grid.479509.60000 0001 0163 8573Center for Stem Cells & Regenerative Medicine, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037 USA ,grid.468218.1Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037 USA
| | - Daniel P. Zolg
- grid.32224.350000 0004 0386 9924Chemical Neurobiology Laboratory, Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114 USA ,grid.32224.350000 0004 0386 9924Departments of Psychiatry & Neurology, Massachusetts General Hospital & Harvard Medical School, Boston, MA 02114 USA ,grid.6936.a0000000123222966Present Address: TUM School of Life Sciences, Technical University of Munich, Munich, Germany
| | - Amanda J. Roberts
- grid.468218.1Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037 USA
| | - Deepak Mani
- grid.38142.3c000000041936754XProteomics Platform, Broad Institute of MIT and Harvard University, Cambridge, MA 02142 USA
| | - Sarah R. Blumenthal
- grid.32224.350000 0004 0386 9924Chemical Neurobiology Laboratory, Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114 USA ,grid.32224.350000 0004 0386 9924Departments of Psychiatry & Neurology, Massachusetts General Hospital & Harvard Medical School, Boston, MA 02114 USA
| | - Iren Kurtser
- grid.32224.350000 0004 0386 9924Chemical Neurobiology Laboratory, Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114 USA ,grid.32224.350000 0004 0386 9924Departments of Psychiatry & Neurology, Massachusetts General Hospital & Harvard Medical School, Boston, MA 02114 USA
| | - Debasis Patnaik
- grid.32224.350000 0004 0386 9924Chemical Neurobiology Laboratory, Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114 USA ,grid.32224.350000 0004 0386 9924Departments of Psychiatry & Neurology, Massachusetts General Hospital & Harvard Medical School, Boston, MA 02114 USA
| | - Irina Gaisina
- grid.185648.60000 0001 2175 0319Department of Medicinal Chemistry & Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - Joshua Bishop
- grid.32224.350000 0004 0386 9924Chemical Neurobiology Laboratory, Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114 USA ,grid.32224.350000 0004 0386 9924Departments of Psychiatry & Neurology, Massachusetts General Hospital & Harvard Medical School, Boston, MA 02114 USA ,grid.417993.10000 0001 2260 0793Present Address: Merck, Boston, MA USA
| | - Steven D. Sheridan
- grid.32224.350000 0004 0386 9924Chemical Neurobiology Laboratory, Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114 USA ,grid.32224.350000 0004 0386 9924Departments of Psychiatry & Neurology, Massachusetts General Hospital & Harvard Medical School, Boston, MA 02114 USA
| | - Jasmin Lalonde
- grid.34429.380000 0004 1936 8198Department of Molecular and Cellular Biology, University of Guelph, 50 Stone Road, East, Guelph, ON Canada N1G 2W1
| | - Steven A. Carr
- grid.38142.3c000000041936754XProteomics Platform, Broad Institute of MIT and Harvard University, Cambridge, MA 02142 USA
| | - Evan Y. Snyder
- grid.479509.60000 0001 0163 8573Center for Stem Cells & Regenerative Medicine, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037 USA ,grid.468218.1Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037 USA ,grid.266100.30000 0001 2107 4242Department of Pediatrics, University of California San Diego, La Jolla, CA 92037 USA
| | - Stephen J. Haggarty
- grid.32224.350000 0004 0386 9924Chemical Neurobiology Laboratory, Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114 USA ,grid.32224.350000 0004 0386 9924Departments of Psychiatry & Neurology, Massachusetts General Hospital & Harvard Medical School, Boston, MA 02114 USA
| |
Collapse
|
8
|
Farkhondeh A, Li R, Gorshkov K, Chen KG, Might M, Rodems S, Lo DC, Zheng W. Induced pluripotent stem cells for neural drug discovery. Drug Discov Today 2019; 24:992-999. [PMID: 30664937 DOI: 10.1016/j.drudis.2019.01.007] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 12/31/2018] [Accepted: 01/15/2019] [Indexed: 12/12/2022]
Abstract
Neurological diseases such as Alzheimer's disease and Parkinson's disease are growing problems, as average life expectancy is increasing globally. Drug discovery for neurological disease remains a major challenge. Poor understanding of disease pathophysiology and incomplete representation of human disease in animal models hinder therapeutic drug development. Recent advances with induced pluripotent stem cells (iPSCs) have enabled modeling of human diseases with patient-derived neural cells. Utilizing iPSC-derived neurons advances compound screening and evaluation of drug efficacy. These cells have the genetic backgrounds of patients that more precisely model disease-specific pathophysiology and phenotypes. Neural cells derived from iPSCs can be produced in a large quantity. Therefore, application of iPSC-derived human neurons is a new direction for neuronal drug discovery.
Collapse
Affiliation(s)
- Atena Farkhondeh
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Rong Li
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Kirill Gorshkov
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Kevin G Chen
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Matthew Might
- University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Donald C Lo
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Wei Zheng
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
9
|
Gilbert TM, Zürcher NR, Wu CJ, Bhanot A, Hightower BG, Kim M, Albrecht DS, Wey HY, Schroeder FA, Rodriguez-Thompson A, Morin TM, Hart KL, Pellegrini AM, Riley MM, Wang C, Stufflebeam SM, Haggarty SJ, Holt DJ, Loggia ML, Perlis RH, Brown HE, Roffman JL, Hooker JM. PET neuroimaging reveals histone deacetylase dysregulation in schizophrenia. J Clin Invest 2018; 129:364-372. [PMID: 30530989 DOI: 10.1172/jci123743] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 11/02/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Patients with schizophrenia (SCZ) experience chronic cognitive deficits. Histone deacetylases (HDACs) are enzymes that regulate cognitive circuitry; however, the role of HDACs in cognitive disorders, including SCZ, remains unknown in humans. We previously determined that HDAC2 mRNA levels were lower in dorsolateral prefrontal cortex (DLPFC) tissue from donors with SCZ compared with controls. Here we investigated the relationship between in vivo HDAC expression and cognitive impairment in patients with SCZ and matched healthy controls using [11C]Martinostat positron emission tomography (PET). METHODS In a case-control study, relative [11C]Martinostat uptake was compared between 14 patients with SCZ or schizoaffective disorder (SCZ/SAD) and 17 controls using hypothesis-driven region-of-interest analysis and unbiased whole brain voxel-wise approaches. Clinical measures, including the MATRICS consensus cognitive battery, were administered. RESULTS Relative HDAC expression was lower in the DLPFC of patients with SCZ/SAD compared with controls, and HDAC expression positively correlated with cognitive performance scores across groups. Patients with SCZ/SAD also showed lower relative HDAC expression in the dorsomedial prefrontal cortex and orbitofrontal gyrus, and higher relative HDAC expression in the cerebral white matter, pons, and cerebellum compared with controls. CONCLUSIONS These findings provide in vivo evidence of HDAC dysregulation in patients with SCZ and suggest that altered HDAC expression may impact cognitive function in humans. FUNDING National Institute of Mental Health (NIMH), Brain and Behavior Foundation, Massachusetts General Hospital (MGH), Athinoula A. Martinos Center for Biomedical Imaging, National Institute of Biomedical Imaging and Bioengineering (NIBIB), NIH Shared Instrumentation Grant Program.
Collapse
Affiliation(s)
- Tonya M Gilbert
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Nicole R Zürcher
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Christine J Wu
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Anisha Bhanot
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Baileigh G Hightower
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Minhae Kim
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Daniel S Albrecht
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Hsiao-Ying Wey
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Frederick A Schroeder
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Anais Rodriguez-Thompson
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Thomas M Morin
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | | | | | - Misha M Riley
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Changning Wang
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Steven M Stufflebeam
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Stephen J Haggarty
- Center for Genomic Medicine.,Department of Neurology, and.,Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Daphne J Holt
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA.,Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Marco L Loggia
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Roy H Perlis
- Center for Genomic Medicine.,Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Hannah E Brown
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Joshua L Roffman
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA.,Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jacob M Hooker
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| |
Collapse
|
10
|
Cobb MM, Ravisankar A, Skibinski G, Finkbeiner S. iPS cells in the study of PD molecular pathogenesis. Cell Tissue Res 2018; 373:61-77. [PMID: 29234887 PMCID: PMC5997490 DOI: 10.1007/s00441-017-2749-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 11/21/2017] [Indexed: 12/12/2022]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease and its pathogenic mechanisms are poorly understood. The majority of PD cases are sporadic but a number of genes are associated with familial PD. Sporadic and familial PD have many molecular and cellular features in common, suggesting some shared pathogenic mechanisms. Induced pluripotent stem cells (iPSCs) have been derived from patients harboring a range of different mutations of PD-associated genes. PD patient-derived iPSCs have been differentiated into relevant cell types, in particular dopaminergic neurons and used as a model to study PD. In this review, we describe how iPSCs have been used to improve our understanding of the pathogenesis of PD. We describe what cellular and molecular phenotypes have been observed in neurons derived from iPSCs harboring known PD-associated mutations and what common pathways may be involved.
Collapse
Affiliation(s)
- Melanie M Cobb
- Gladstone Institutes, the Taube/Koret Center for Neurodegenerative Disease, San Francisco, CA, 94158, USA
| | - Abinaya Ravisankar
- Gladstone Institutes, the Taube/Koret Center for Neurodegenerative Disease, San Francisco, CA, 94158, USA
| | - Gaia Skibinski
- Gladstone Institutes, the Taube/Koret Center for Neurodegenerative Disease, San Francisco, CA, 94158, USA
| | - Steven Finkbeiner
- Gladstone Institutes, the Taube/Koret Center for Neurodegenerative Disease, San Francisco, CA, 94158, USA.
- Department of Neurology, University of California, San Francisco, CA, 94143, USA.
- Department Physiology, University of California, San Francisco, CA, 94143, USA.
- Graduate Programs in Neuroscience and Biomedical Sciences, University of California, San Francisco, CA, 94143, USA.
| |
Collapse
|
11
|
Mizee MR, Poel MVD, Huitinga I. Purification of cells from fresh human brain tissue: primary human glial cells. HANDBOOK OF CLINICAL NEUROLOGY 2018; 150:273-283. [PMID: 29496146 DOI: 10.1016/b978-0-444-63639-3.00019-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
In order to translate the findings obtained from postmortem brain tissue samples to functional biologic mechanisms of central nervous system disease, it will be necessary to understand how these findings affect the different cell populations in the brain. The acute isolation and analysis of pure glial cell populations are common practice in animal models for neurologic diseases, but are not yet regularly applied to human postmortem brain material. The development of novel cell isolation techniques and methods for transcriptomic and proteomic analysis have made it possible to isolate and phenotype primary human cell populations from the central nervous system. The psychiatric program of the Netherlands Brain Bank has considerable experience with the purification of glial cells. This chapter will review the rapid isolation and phenotyping procedures for two major glia cell populations in the human brain, microglia and astrocytes, and will also discuss the potential for biobanking these cells, as well as the possible alternatives to cell isolations. The acute isolation of glial cells without culture-based adherence steps allows the analysis of glial alterations that underlie, or are the result of, disease neuropathology of the donor.
Collapse
Affiliation(s)
- Mark Ronald Mizee
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands; Psychiatric Program, Netherlands Brain Bank, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Marlijn van der Poel
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Inge Huitinga
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands; Psychiatric Program, Netherlands Brain Bank, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands.
| |
Collapse
|
12
|
Gonzalez DM, Gregory J, Brennand KJ. The Importance of Non-neuronal Cell Types in hiPSC-Based Disease Modeling and Drug Screening. Front Cell Dev Biol 2017; 5:117. [PMID: 29312938 PMCID: PMC5742170 DOI: 10.3389/fcell.2017.00117] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 12/08/2017] [Indexed: 12/13/2022] Open
Abstract
Current applications of human induced pluripotent stem cell (hiPSC) technologies in patient-specific models of neurodegenerative and neuropsychiatric disorders tend to focus on neuronal phenotypes. Here, we review recent efforts toward advancing hiPSCs toward non-neuronal cell types of the central nervous system (CNS) and highlight their potential use for the development of more complex in vitro models of neurodevelopment and disease. We present evidence from previous works in both rodents and humans of the importance of these cell types (oligodendrocytes, microglia, astrocytes) in neurological disease and highlight new hiPSC-based models that have sought to explore these relationships in vitro. Lastly, we summarize efforts toward conducting high-throughput screening experiments with hiPSCs and propose methods by which new screening platforms could be designed to better capture complex relationships between neural cell populations in health and disease.
Collapse
Affiliation(s)
- David M Gonzalez
- Medical Scientist Training Program, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Department of Developmental and Stem Cell Biology, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Jill Gregory
- Instructional Technology Group, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Kristen J Brennand
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
13
|
RA Differentiation Enhances Dopaminergic Features, Changes Redox Parameters, and Increases Dopamine Transporter Dependency in 6-Hydroxydopamine-Induced Neurotoxicity in SH-SY5Y Cells. Neurotox Res 2017; 31:545-559. [DOI: 10.1007/s12640-016-9699-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 12/28/2016] [Accepted: 12/30/2016] [Indexed: 12/19/2022]
|
14
|
Combining Patient-Reprogrammed Neural Cells and Proteomics as a Model to Study Psychiatric Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 974:279-287. [DOI: 10.1007/978-3-319-52479-5_26] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
15
|
Watson LM, Wong MMK, Becker EBE. Induced pluripotent stem cell technology for modelling and therapy of cerebellar ataxia. Open Biol 2016; 5:150056. [PMID: 26136256 PMCID: PMC4632502 DOI: 10.1098/rsob.150056] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Induced pluripotent stem cell (iPSC) technology has emerged as an important tool in understanding, and potentially reversing, disease pathology. This is particularly true in the case of neurodegenerative diseases, in which the affected cell types are not readily accessible for study. Since the first descriptions of iPSC-based disease modelling, considerable advances have been made in understanding the aetiology and progression of a diverse array of neurodegenerative conditions, including Parkinson's disease and Alzheimer's disease. To date, however, relatively few studies have succeeded in using iPSCs to model the neurodegeneration observed in cerebellar ataxia. Given the distinct neurodevelopmental phenotypes associated with certain types of ataxia, iPSC-based models are likely to provide significant insights, not only into disease progression, but also to the development of early-intervention therapies. In this review, we describe the existing iPSC-based disease models of this heterogeneous group of conditions and explore the challenges associated with generating cerebellar neurons from iPSCs, which have thus far hindered the expansion of this research.
Collapse
Affiliation(s)
- Lauren M Watson
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Maggie M K Wong
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Esther B E Becker
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
16
|
Abstract
Despite a lack of recent progress in the treatment of schizophrenia, our understanding of its genetic and environmental causes has considerably improved, and their relationship to aberrant patterns of neurodevelopment has become clearer. This raises the possibility that 'disease-modifying' strategies could alter the course to - and of - this debilitating disorder, rather than simply alleviating symptoms. A promising window for course-altering intervention is around the time of the first episode of psychosis, especially in young people at risk of transition to schizophrenia. Indeed, studies performed in both individuals at risk of developing schizophrenia and rodent models for schizophrenia suggest that pre-diagnostic pharmacotherapy and psychosocial or cognitive-behavioural interventions can delay or moderate the emergence of psychosis. Of particular interest are 'hybrid' strategies that both relieve presenting symptoms and reduce the risk of transition to schizophrenia or another psychiatric disorder. This Review aims to provide a broad-based consideration of the challenges and opportunities inherent in efforts to alter the course of schizophrenia.
Collapse
|
17
|
Advancing drug discovery for neuropsychiatric disorders using patient-specific stem cell models. Mol Cell Neurosci 2016; 73:104-15. [PMID: 26826498 DOI: 10.1016/j.mcn.2016.01.011] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 01/22/2016] [Accepted: 01/25/2016] [Indexed: 12/17/2022] Open
Abstract
Compelling clinical, social, and economic reasons exist to innovate in the process of drug discovery for neuropsychiatric disorders. The use of patient-specific, induced pluripotent stem cells (iPSCs) now affords the ability to generate neuronal cell-based models that recapitulate key aspects of human disease. In the context of neuropsychiatric disorders, where access to physiologically active and relevant cell types of the central nervous system for research is extremely limiting, iPSC-derived in vitro culture of human neurons and glial cells is transformative. Potential applications relevant to early stage drug discovery, include support of quantitative biochemistry, functional genomics, proteomics, and perhaps most notably, high-throughput and high-content chemical screening. While many phenotypes in human iPSC-derived culture systems may prove adaptable to screening formats, addressing the question of which in vitro phenotypes are ultimately relevant to disease pathophysiology and therefore more likely to yield effective pharmacological agents that are disease-modifying treatments requires careful consideration. Here, we review recent examples of studies of neuropsychiatric disorders using human stem cell models where cellular phenotypes linked to disease and functional assays have been reported. We also highlight technical advances using genome-editing technologies in iPSCs to support drug discovery efforts, including the interpretation of the functional significance of rare genetic variants of unknown significance and for the purpose of creating cell type- and pathway-selective functional reporter assays. Additionally, we evaluate the potential of in vitro stem cell models to investigate early events of disease pathogenesis, in an effort to understand the underlying molecular mechanism, including the basis of selective cell-type vulnerability, and the potential to create new cell-based diagnostics to aid in the classification of patients and subsequent selection for clinical trials. A number of key challenges remain, including the scaling of iPSC models to larger cohorts and integration with rich clinicopathological information and translation of phenotypes. Still, the overall use of iPSC-based human cell models with functional cellular and biochemical assays holds promise for supporting the discovery of next-generation neuropharmacological agents for the treatment and ultimately prevention of a range of severe mental illnesses.
Collapse
|
18
|
Disease signatures for schizophrenia and bipolar disorder using patient-derived induced pluripotent stem cells. Mol Cell Neurosci 2016; 73:96-103. [PMID: 26777134 DOI: 10.1016/j.mcn.2016.01.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 01/05/2016] [Accepted: 01/11/2016] [Indexed: 12/12/2022] Open
Abstract
Schizophrenia and bipolar disorder are complex psychiatric disorders that present unique challenges in the study of disease biology. There are no objective biological phenotypes for these disorders, which are characterized by complex genetics and prominent roles for gene-environment interactions. The study of the neurobiology underlying these severe psychiatric disorders has been hindered by the lack of access to the tissue of interest - neurons from patients. The advent of reprogramming methods that enable generation of induced pluripotent stem cells (iPSCs) from patient fibroblasts and peripheral blood mononuclear cells has opened possibilities for new approaches to study relevant disease biology using iPSC-derived neurons. While early studies with patient iPSCs have led to promising and intriguing leads, significant hurdles remain in our attempts to capture the complexity of these disorders in vitro. We present here an overview of studies to date of schizophrenia and bipolar disorder using iPSC-derived neuronal cells and discuss potential future directions that can result in the identification of robust and valid cellular phenotypes that in turn can lay the groundwork for meaningful clinical advances.
Collapse
|
19
|
Bradford AB, McNutt PM. Importance of being Nernst: Synaptic activity and functional relevance in stem cell-derived neurons. World J Stem Cells 2015; 7:899-921. [PMID: 26240679 PMCID: PMC4515435 DOI: 10.4252/wjsc.v7.i6.899] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 02/28/2015] [Accepted: 05/11/2015] [Indexed: 02/06/2023] Open
Abstract
Functional synaptogenesis and network emergence are signature endpoints of neurogenesis. These behaviors provide higher-order confirmation that biochemical and cellular processes necessary for neurotransmitter release, post-synaptic detection and network propagation of neuronal activity have been properly expressed and coordinated among cells. The development of synaptic neurotransmission can therefore be considered a defining property of neurons. Although dissociated primary neuron cultures readily form functioning synapses and network behaviors in vitro, continuously cultured neurogenic cell lines have historically failed to meet these criteria. Therefore, in vitro-derived neuron models that develop synaptic transmission are critically needed for a wide array of studies, including molecular neuroscience, developmental neurogenesis, disease research and neurotoxicology. Over the last decade, neurons derived from various stem cell lines have shown varying ability to develop into functionally mature neurons. In this review, we will discuss the neurogenic potential of various stem cells populations, addressing strengths and weaknesses of each, with particular attention to the emergence of functional behaviors. We will propose methods to functionally characterize new stem cell-derived neuron (SCN) platforms to improve their reliability as physiological relevant models. Finally, we will review how synaptically active SCNs can be applied to accelerate research in a variety of areas. Ultimately, emphasizing the critical importance of synaptic activity and network responses as a marker of neuronal maturation is anticipated to result in in vitro findings that better translate to efficacious clinical treatments.
Collapse
|
20
|
Madison JM, Zhou F, Nigam A, Hussain A, Barker DD, Nehme R, van der Ven K, Hsu J, Wolf P, Fleishman M, O’Dushlaine C, Rose S, Chambert K, Lau FH, Ahfeldt T, Rueckert EH, Sheridan SD, Fass DM, Nemesh J, Mullen TE, Daheron L, McCarroll S, Sklar P, Perlis RH, Haggarty SJ. Characterization of bipolar disorder patient-specific induced pluripotent stem cells from a family reveals neurodevelopmental and mRNA expression abnormalities. Mol Psychiatry 2015; 20:703-17. [PMID: 25733313 PMCID: PMC4440839 DOI: 10.1038/mp.2015.7] [Citation(s) in RCA: 140] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 10/29/2014] [Accepted: 12/19/2014] [Indexed: 02/07/2023]
Abstract
Bipolar disorder (BD) is a common neuropsychiatric disorder characterized by chronic recurrent episodes of depression and mania. Despite evidence for high heritability of BD, little is known about its underlying pathophysiology. To develop new tools for investigating the molecular and cellular basis of BD, we applied a family-based paradigm to derive and characterize a set of 12 induced pluripotent stem cell (iPSC) lines from a quartet consisting of two BD-affected brothers and their two unaffected parents. Initially, no significant phenotypic differences were observed between iPSCs derived from the different family members. However, upon directed neural differentiation, we observed that CXCR4 (CXC chemokine receptor-4) expressing central nervous system (CNS) neural progenitor cells (NPCs) from both BD patients compared with their unaffected parents exhibited multiple phenotypic differences at the level of neurogenesis and expression of genes critical for neuroplasticity, including WNT pathway components and ion channel subunits. Treatment of the CXCR4(+) NPCs with a pharmacological inhibitor of glycogen synthase kinase 3, a known regulator of WNT signaling, was found to rescue a progenitor proliferation deficit in the BD patient NPCs. Taken together, these studies provide new cellular tools for dissecting the pathophysiology of BD and evidence for dysregulation of key pathways involved in neurodevelopment and neuroplasticity. Future generation of additional iPSCs following a family-based paradigm for modeling complex neuropsychiatric disorders in conjunction with in-depth phenotyping holds promise for providing insights into the pathophysiological substrates of BD and is likely to inform the development of targeted therapeutics for its treatment and ideally prevention.
Collapse
Affiliation(s)
- Jon M. Madison
- Stanley Center for Psychiatric Research, Broad Institute of MIT & Harvard, Cambridge, MA 02142, USA,Psychiatric & Neurodevelopmental Genetics Unit, Center for Human Genetics Research, Massachusetts General Hospital, Boston, MA 02114, USA,Department of Psychiatry, Massachusetts General Hospital, Boston, MA 02114, USA,Correspondence: (JM), (SJH)
| | - Fen Zhou
- Stanley Center for Psychiatric Research, Broad Institute of MIT & Harvard, Cambridge, MA 02142, USA,Psychiatric & Neurodevelopmental Genetics Unit, Center for Human Genetics Research, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Aparna Nigam
- Stanley Center for Psychiatric Research, Broad Institute of MIT & Harvard, Cambridge, MA 02142, USA,Department of Psychiatry, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Ali Hussain
- Stanley Center for Psychiatric Research, Broad Institute of MIT & Harvard, Cambridge, MA 02142, USA,Psychiatric & Neurodevelopmental Genetics Unit, Center for Human Genetics Research, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Douglas D. Barker
- Stanley Center for Psychiatric Research, Broad Institute of MIT & Harvard, Cambridge, MA 02142, USA
| | - Ralda Nehme
- Stanley Center for Psychiatric Research, Broad Institute of MIT & Harvard, Cambridge, MA 02142, USA,Department of Stem Cell & Regenerative Biology, Harvard University, Cambridge, MA,Department of Neurology, Massachusetts General Hospital & Harvard Medical School, Boston, MA 02114, USA
| | - Karlijn van der Ven
- Psychiatric & Neurodevelopmental Genetics Unit, Center for Human Genetics Research, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Jenny Hsu
- Stanley Center for Psychiatric Research, Broad Institute of MIT & Harvard, Cambridge, MA 02142, USA,Psychiatric & Neurodevelopmental Genetics Unit, Center for Human Genetics Research, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Pavlina Wolf
- Stanley Center for Psychiatric Research, Broad Institute of MIT & Harvard, Cambridge, MA 02142, USA,Psychiatric & Neurodevelopmental Genetics Unit, Center for Human Genetics Research, Massachusetts General Hospital, Boston, MA 02114, USA,Department of Neurology, Massachusetts General Hospital & Harvard Medical School, Boston, MA 02114, USA
| | - Morgan Fleishman
- Stanley Center for Psychiatric Research, Broad Institute of MIT & Harvard, Cambridge, MA 02142, USA,Psychiatric & Neurodevelopmental Genetics Unit, Center for Human Genetics Research, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Colm O’Dushlaine
- Stanley Center for Psychiatric Research, Broad Institute of MIT & Harvard, Cambridge, MA 02142, USA
| | - Sam Rose
- Stanley Center for Psychiatric Research, Broad Institute of MIT & Harvard, Cambridge, MA 02142, USA
| | - Kimberly Chambert
- Stanley Center for Psychiatric Research, Broad Institute of MIT & Harvard, Cambridge, MA 02142, USA
| | - Frank H. Lau
- Department of Stem Cell & Regenerative Biology, Harvard University, Cambridge, MA
| | - Tim Ahfeldt
- Department of Stem Cell & Regenerative Biology, Harvard University, Cambridge, MA
| | - Erroll H. Rueckert
- Stanley Center for Psychiatric Research, Broad Institute of MIT & Harvard, Cambridge, MA 02142, USA,Psychiatric & Neurodevelopmental Genetics Unit, Center for Human Genetics Research, Massachusetts General Hospital, Boston, MA 02114, USA,Chemical Neurobiology Laboratory, Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Steven D. Sheridan
- Chemical Neurobiology Laboratory, Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Daniel M. Fass
- Stanley Center for Psychiatric Research, Broad Institute of MIT & Harvard, Cambridge, MA 02142, USA,Department of Neurology, Massachusetts General Hospital & Harvard Medical School, Boston, MA 02114, USA,Chemical Neurobiology Laboratory, Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA 02114, USA
| | - James Nemesh
- Stanley Center for Psychiatric Research, Broad Institute of MIT & Harvard, Cambridge, MA 02142, USA,Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Thomas E. Mullen
- Stanley Center for Psychiatric Research, Broad Institute of MIT & Harvard, Cambridge, MA 02142, USA,Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Laurence Daheron
- Department of Stem Cell & Regenerative Biology, Harvard University, Cambridge, MA
| | - Steve McCarroll
- Stanley Center for Psychiatric Research, Broad Institute of MIT & Harvard, Cambridge, MA 02142, USA,Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Pamela Sklar
- Department of Psychiatry, Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Roy H. Perlis
- Stanley Center for Psychiatric Research, Broad Institute of MIT & Harvard, Cambridge, MA 02142, USA,Psychiatric & Neurodevelopmental Genetics Unit, Center for Human Genetics Research, Massachusetts General Hospital, Boston, MA 02114, USA,Department of Psychiatry, Massachusetts General Hospital, Boston, MA 02114, USA,Chemical Neurobiology Laboratory, Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Stephen J. Haggarty
- Stanley Center for Psychiatric Research, Broad Institute of MIT & Harvard, Cambridge, MA 02142, USA,Psychiatric & Neurodevelopmental Genetics Unit, Center for Human Genetics Research, Massachusetts General Hospital, Boston, MA 02114, USA,Department of Psychiatry, Massachusetts General Hospital, Boston, MA 02114, USA,Department of Neurology, Massachusetts General Hospital & Harvard Medical School, Boston, MA 02114, USA,Chemical Neurobiology Laboratory, Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA 02114, USA,Correspondence: (JM), (SJH)
| |
Collapse
|
21
|
Hohmann S, Adamo N, Lahey BB, Faraone SV, Banaschewski T. Genetics in child and adolescent psychiatry: methodological advances and conceptual issues. Eur Child Adolesc Psychiatry 2015; 24:619-34. [PMID: 25850999 DOI: 10.1007/s00787-015-0702-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 03/06/2015] [Indexed: 10/23/2022]
Abstract
Discovering the genetic basis of early-onset psychiatric disorders has been the aim of intensive research during the last decade. We will first selectively summarize results of genetic research in child and adolescent psychiatry by using examples from different disorders and discuss methodological issues, emerging questions and future directions. In the second part of this review, we will focus on how to link genetic causes of disorders with physiological pathways, discuss the impact of genetic findings on diagnostic systems, prevention and therapeutic interventions. Finally we will highlight some ethical aspects connected to genetic research in child and adolescent psychiatry. Advances in molecular genetic methods have led to insights into the genetic architecture of psychiatric disorders, but not yet provided definite pathways to pathophysiology. If replicated, promising findings from genetic studies might in some cases lead to personalized treatments. On the one hand, knowledge of the genetic basis of disorders may influence diagnostic categories. On the other hand, models also suggest studying the genetic architecture of psychiatric disorders across diagnoses and clinical groups.
Collapse
Affiliation(s)
- Sarah Hohmann
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, Mannheim, Germany
| | | | | | | | | |
Collapse
|
22
|
McCoy TH, Perlis RH. A tool to utilize adverse effect profiles to identify brain-active medications for repurposing. Int J Neuropsychopharmacol 2015; 18:pyu078. [PMID: 25673184 PMCID: PMC4360243 DOI: 10.1093/ijnp/pyu078] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND To shorten the time required to bring new treatments to clinics, recent efforts have focused on repurposing existing Food and Drug Administration (FDA)-approved drugs with established safety data for new indications. We hypothesized that adverse effect profiles might aid in prioritizing compounds for investigation in central nervous system (CNS) applications by providing an indication of their abilities to cross the blood-brain barrier. METHODS Data were drawn from an investigation of similarity of adverse effect profiles, utilizing pre- and post-marketing data. A panel of known CNS-active drugs was utilized to estimate aggregate similarity profiles for all other FDA drugs in the database. Permutations were used to test whether similarities for any given drug exceeded that expected under the null hypothesis. To estimate the performance of algorithms using such profiles, manually-curated lists of known CNS-active and -inactive medications were classified using logistic regression. Algorithms with and without this similarity data were compared for prediction of CNS penetrance. RESULTS Models incorporating adverse effect similarity data exhibited greater discrimination of brain-penetrant and non-penetrant drugs than models without this data. A visualization tool was developed to allow any medication to be evaluated for adverse effect similarity to the CNS panel or a custom panel. CONCLUSIONS Consideration of adverse effect profiles allows in silico prioritization of compounds for follow-up investigation for CNS indications. In concert with chemical screening approaches, this may accelerate repurposing efforts for putative CNS-active medications.
Collapse
Affiliation(s)
- Thomas H McCoy
- Center for Experimental Drugs and Diagnostics, Massachusetts General Hospital Department of Psychiatry and Harvard Medical School, Boston, MA (Drs McCoy and Perlis)
| | - Roy H Perlis
- Center for Experimental Drugs and Diagnostics, Massachusetts General Hospital Department of Psychiatry and Harvard Medical School, Boston, MA (Drs McCoy and Perlis).
| |
Collapse
|
23
|
Zeng X, Hunsberger JG, Simeonov A, Malik N, Pei Y, Rao M. Concise review: modeling central nervous system diseases using induced pluripotent stem cells. Stem Cells Transl Med 2014; 3:1418-28. [PMID: 25368377 DOI: 10.5966/sctm.2014-0102] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) offer an opportunity to delve into the mechanisms underlying development while also affording the potential to take advantage of a number of naturally occurring mutations that contribute to either disease susceptibility or resistance. Just as with any new field, several models of screening are being explored, and innovators are working on the most efficient methods to overcome the inherent limitations of primary cell screens using iPSCs. In the present review, we provide a background regarding why iPSCs represent a paradigm shift for central nervous system (CNS) disease modeling. We describe the efforts in the field to develop more biologically relevant CNS disease models, which should provide screening assays useful for the pharmaceutical industry. We also provide some examples of successful uses for iPSC-based screens and suggest that additional development could revolutionize the field of drug discovery. The development and implementation of these advanced iPSC-based screens will create a more efficient disease-specific process underpinned by the biological mechanism in a patient- and disease-specific manner rather than by trial-and-error. Moreover, with careful and strategic planning, shared resources can be developed that will enable exponential advances in the field. This will undoubtedly lead to more sensitive and accurate screens for early diagnosis and allow the identification of patient-specific therapies, thus, paving the way to personalized medicine.
Collapse
Affiliation(s)
- Xianmin Zeng
- XCell Science Inc., Novato, California, USA; Buck Institute for Research on Aging, Novato, California, USA; Laboratory of Stem Cell Biology, NIH Center for Regenerative Medicine, Bethesda, Maryland, USA; National Center for Advancing Translational Sciences, NIH, Bethesda, Maryland, USA; New York Stem Cell Foundation, New York, New York, USA
| | - Joshua G Hunsberger
- XCell Science Inc., Novato, California, USA; Buck Institute for Research on Aging, Novato, California, USA; Laboratory of Stem Cell Biology, NIH Center for Regenerative Medicine, Bethesda, Maryland, USA; National Center for Advancing Translational Sciences, NIH, Bethesda, Maryland, USA; New York Stem Cell Foundation, New York, New York, USA
| | - Anton Simeonov
- XCell Science Inc., Novato, California, USA; Buck Institute for Research on Aging, Novato, California, USA; Laboratory of Stem Cell Biology, NIH Center for Regenerative Medicine, Bethesda, Maryland, USA; National Center for Advancing Translational Sciences, NIH, Bethesda, Maryland, USA; New York Stem Cell Foundation, New York, New York, USA
| | - Nasir Malik
- XCell Science Inc., Novato, California, USA; Buck Institute for Research on Aging, Novato, California, USA; Laboratory of Stem Cell Biology, NIH Center for Regenerative Medicine, Bethesda, Maryland, USA; National Center for Advancing Translational Sciences, NIH, Bethesda, Maryland, USA; New York Stem Cell Foundation, New York, New York, USA
| | - Ying Pei
- XCell Science Inc., Novato, California, USA; Buck Institute for Research on Aging, Novato, California, USA; Laboratory of Stem Cell Biology, NIH Center for Regenerative Medicine, Bethesda, Maryland, USA; National Center for Advancing Translational Sciences, NIH, Bethesda, Maryland, USA; New York Stem Cell Foundation, New York, New York, USA
| | - Mahendra Rao
- XCell Science Inc., Novato, California, USA; Buck Institute for Research on Aging, Novato, California, USA; Laboratory of Stem Cell Biology, NIH Center for Regenerative Medicine, Bethesda, Maryland, USA; National Center for Advancing Translational Sciences, NIH, Bethesda, Maryland, USA; New York Stem Cell Foundation, New York, New York, USA
| |
Collapse
|
24
|
Gamo NJ, Sawa A. Human stem cells and surrogate tissues for basic and translational study of mental disorders. Biol Psychiatry 2014; 75:918-9. [PMID: 24862565 PMCID: PMC4302385 DOI: 10.1016/j.biopsych.2014.03.025] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 03/14/2014] [Indexed: 12/28/2022]
Affiliation(s)
| | - Akira Sawa
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|