51
|
DNA methylation differences in monozygotic twin pairs discordant for schizophrenia identifies psychosis related genes and networks. BMC Med Genomics 2015; 8:17. [PMID: 25943100 PMCID: PMC4494167 DOI: 10.1186/s12920-015-0093-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 04/24/2015] [Indexed: 01/28/2023] Open
Abstract
Background Despite their singular origin, monozygotic twin pairs often display discordance for complex disorders including schizophrenia. It is a common (1%) and often familial disease with a discordance rate of ~50% in monozygotic twins. This high discordance is often explained by the role of yet unknown environmental, random, and epigenetic factors. The involvement of DNA methylation in this disease appears logical, but remains to be established. Methods We have used blood DNA from two pairs of monozygotic twins discordant for schizophrenia and their parents in order to assess genome-wide methylation using a NimbleGen Methylation Promoter Microarray. Results The genome-wide results show that differentially methylated regions (DMRs) exist between members representing discordant monozygotic twins. Some DMRs are shared with parent(s) and others appear to be de novo. We found twenty-seven genes affected by DMR changes that were shared in the affected member of two discordant monozygotic pairs from unrelated families. Interestingly, the genes affected by pair specific DMRs share specific networks. Specifically, this study has identified two networks; “cell death and survival” and a “cellular movement and immune cell trafficking”. These two networks and the genes affected have been previously implicated in the aetiology of schizophrenia. Conclusions The results are compatible with the suggestion that DNA methylation may contribute to the discordance of monozygotic twins for schizophrenia. Also, this may be accomplished by the direct effect of gene specific methylation changes on specific biological networks rather than individual genes. It supports the extensive genetic, epigenetic and phenotypic heterogeneity implicated in schizophrenia. Electronic supplementary material The online version of this article (doi:10.1186/s12920-015-0093-1) contains supplementary material, which is available to authorized users.
Collapse
|
52
|
Affiliation(s)
- Brigham J. Hartley
- Departments of Psychiatry and Neuroscience, Mount Sinai School of
Medicine, 1425 Madison Avenue, New York, NY 10029
| | - Ngoc Tran
- Departments of Psychiatry and Neuroscience, Mount Sinai School of
Medicine, 1425 Madison Avenue, New York, NY 10029
| | - Ian Ladran
- Departments of Psychiatry and Neuroscience, Mount Sinai School of
Medicine, 1425 Madison Avenue, New York, NY 10029
| | - Kathryn Reggio
- Departments of Psychiatry and Neuroscience, Mount Sinai School of
Medicine, 1425 Madison Avenue, New York, NY 10029
| | - Kristen J. Brennand
- Departments of Psychiatry and Neuroscience, Mount Sinai School of
Medicine, 1425 Madison Avenue, New York, NY 10029
| |
Collapse
|
53
|
Translational potential of olfactory mucosa for the study of neuropsychiatric illness. Transl Psychiatry 2015; 5:e527. [PMID: 25781226 PMCID: PMC4354342 DOI: 10.1038/tp.2014.141] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Revised: 10/22/2014] [Accepted: 11/17/2014] [Indexed: 01/02/2023] Open
Abstract
The olfactory mucosa (OM) is a unique source of regenerative neural tissue that is readily obtainable from living human subjects and thus affords opportunities for the study of psychiatric illnesses. OM tissues can be used, either as ex vivo OM tissue or in vitro OM-derived neural cells, to explore parameters that have been difficult to assess in the brain of living individuals with psychiatric illness. As OM tissues are distinct from brain tissues, an understanding of the neurobiology of the OM is needed to relate findings in these tissues to those of the brain as well as to design and interpret ex vivo or in vitro OM studies. To that end, we discuss the molecular, cellular and functional characteristics of cell types within the olfactory mucosa, describe the organization of the OM and highlight its role in the olfactory neurocircuitry. In addition, we discuss various approaches to in vitro culture of OM-derived cells and their characterization, focusing on the extent to which they reflect the in vivo neurobiology of the OM. Finally, we review studies of ex vivo OM tissues and in vitro OM-derived cells from individuals with psychiatric, neurodegenerative and neurodevelopmental disorders. In particular, we discuss the concordance of this work with postmortem brain studies and highlight possible future approaches, which may offer distinct strengths in comparison to in vitro paradigms based on genomic reprogramming.
Collapse
|
54
|
Horváth S, Mirnics K. Schizophrenia as a disorder of molecular pathways. Biol Psychiatry 2015; 77:22-8. [PMID: 24507510 PMCID: PMC4092052 DOI: 10.1016/j.biopsych.2014.01.001] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Revised: 01/02/2014] [Accepted: 01/05/2014] [Indexed: 12/21/2022]
Abstract
Over the last decade, transcriptome studies of postmortem tissue from subjects with schizophrenia revealed that synaptic, mitochondrial, immune system, gamma-aminobutyric acidergic, and oligodendrocytic changes are all integral parts of the disease process. The combined genetic and transcriptomic studies argue that the molecular underpinnings of the disease are even more varied than the symptomatic diversity of schizophrenia. Ultimately, to decipher the pathophysiology of human disorders in general, we will need to understand the function of hundreds of genes and regulatory elements in our genome and the consequences of their overexpression and reduced expression in a developmental context. Furthermore, integration of knowledge from various data sources remains a monumental challenge that has to be systematically addressed in the upcoming decades. In the end, our success in interpreting the molecular changes in schizophrenia will depend on our ability to understand the biology using innovative ideas and cannot depend on the hope of developing novel, more powerful technologies.
Collapse
Affiliation(s)
- Szatmár Horváth
- Department of Psychiatry; Department of Psychiatry, University of Szeged, Szeged, Hungary
| | - Károly Mirnics
- Department of Psychiatry; Vanderbilt Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, Tennessee; Department of Psychiatry, University of Szeged, Szeged, Hungary.
| |
Collapse
|
55
|
Narayan S, McLean C, Sawa A, Lin SY, Rai N, Hipolito MS, Cascella N, Nurnberger JJI, Ishizuka K, Nwulia EA. Olfactory neurons obtained through nasal biopsy combined with laser-capture microdissection: a potential approach to study treatment response in mental disorders. J Vis Exp 2014:51853. [PMID: 25549156 PMCID: PMC4396916 DOI: 10.3791/51853] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Bipolar disorder (BD) is a severe neuropsychiatric disorder with poorly understood pathophysiology and typically treated with the mood stabilizer, lithium carbonate. Animal studies as well as human genetic studies indicate that lithium affects molecular targets that are involved in neuronal growth, survival and maturation, and notably molecules involved in Wnt signaling. Given the ethical challenge to obtaining brain biopsies for investigating dynamic molecular changes associated with lithium-response in the central nervous system (CNS), one may consider the use of neurons obtained from olfactory tissues to achieve this goal.The olfactory epithelium contains olfactory receptor neurons at different stages of development and glial-like supporting cells. This provides a unique opportunity to study dynamic changes in the CNS of patients with neuropsychiatric diseases, using olfactory tissue safely obtained from nasal biopsies. To overcome the drawback posed by substantial contamination of biopsied olfactory tissue with non-neuronal cells, a novel approach to obtain enriched neuronal cell populations was developed by combining nasal biopsies with laser-capture microdissection. In this study, a system for investigating treatment-associated dynamic molecular changes in neuronal tissue was developed and validated, using a small pilot sample of BD patients recruited for the study of the molecular mechanisms of lithium treatment response.
Collapse
Affiliation(s)
| | - Charlee McLean
- Department of Psychiatry and Behavioral Sciences, Howard University
| | - Akira Sawa
- Department of Psychiatry, Johns Hopkins University
| | - Sandra Y Lin
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University
| | - Narayan Rai
- Department of Psychiatry and Behavioral Sciences, Howard University
| | | | | | | | | | | |
Collapse
|
56
|
Sawa A, Seidman LJ. Is prophylactic psychiatry around the corner? combating adolescent oxidative stress for adult psychosis and schizophrenia. Neuron 2014; 83:991-3. [PMID: 25189204 PMCID: PMC8924824 DOI: 10.1016/j.neuron.2014.08.028] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Early detection and intervention are key principles in clinical medicine and psychiatry. In this issue of Neuron, Cabungcal et al. (2014) demonstrate that prophylactic treatment with antioxidants in adolescence prevents adult deficits in a rat model relevant to schizophrenia.
Collapse
Affiliation(s)
- Akira Sawa
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| | - Larry J Seidman
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
57
|
Fournier M, Ferrari C, Baumann PS, Polari A, Monin A, Bellier-Teichmann T, Wulff J, Pappan KL, Cuenod M, Conus P, Do KQ. Impaired metabolic reactivity to oxidative stress in early psychosis patients. Schizophr Bull 2014; 40:973-83. [PMID: 24687046 PMCID: PMC4133680 DOI: 10.1093/schbul/sbu053] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Because increasing evidence point to the convergence of environmental and genetic risk factors to drive redox dysregulation in schizophrenia, we aim to clarify whether the metabolic anomalies associated with early psychosis reflect an adaptation to oxidative stress. Metabolomic profiling was performed to characterize the response to oxidative stress in fibroblasts from control individuals (n = 20) and early psychosis patients (n = 30), and in all, 282 metabolites were identified. In addition to the expected redox/antioxidant response, oxidative stress induced a decrease of lysolipid levels in fibroblasts from healthy controls that were largely muted in fibroblasts from patients. Most notably, fibroblasts from patients showed disrupted extracellular matrix- and arginine-related metabolism after oxidative stress, indicating impairments beyond the redox system. Plasma membrane and extracellular matrix, 2 regulators of neuronal activity and plasticity, appeared as particularly susceptible to oxidative stress and thus provide novel mechanistic insights for pathophysiological understanding of early stages of psychosis. Statistically, antipsychotic medication at the time of biopsy was not accounting for these anomalies in the metabolism of patients' fibroblasts, indicating that they might be intrinsic to the disease. Although these results are preliminary and should be confirmed in a larger group of patients, they nevertheless indicate that the metabolic signature of reactivity to oxidative stress may provide reliable early markers of psychosis. Developing protective measures aimed at normalizing the disrupted pathways should prevent the pathological consequences of environmental stressors.
Collapse
Affiliation(s)
- Margot Fournier
- Department of Psychiatry, Unit for Research in Schizophrenia, Center for Psychiatric Neuroscience, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Carina Ferrari
- Department of Psychiatry, Unit for Research in Schizophrenia, Center for Psychiatric Neuroscience, Lausanne University Hospital (CHUV), Lausanne, Switzerland;,Department of Psychiatry, Service of General Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Philipp S. Baumann
- Department of Psychiatry, Unit for Research in Schizophrenia, Center for Psychiatric Neuroscience, Lausanne University Hospital (CHUV), Lausanne, Switzerland;,Department of Psychiatry, Service of General Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Andrea Polari
- Department of Psychiatry, Unit for Research in Schizophrenia, Center for Psychiatric Neuroscience, Lausanne University Hospital (CHUV), Lausanne, Switzerland;,Department of Psychiatry, Service of General Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Aline Monin
- Department of Psychiatry, Unit for Research in Schizophrenia, Center for Psychiatric Neuroscience, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Tanja Bellier-Teichmann
- Department of Psychiatry, Unit for Research in Schizophrenia, Center for Psychiatric Neuroscience, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | | | | | - Michel Cuenod
- Department of Psychiatry, Unit for Research in Schizophrenia, Center for Psychiatric Neuroscience, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Philippe Conus
- Department of Psychiatry, Service of General Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland;,These authors contributed equally to this work
| | - Kim Q. Do
- Department of Psychiatry, Unit for Research in Schizophrenia, Center for Psychiatric Neuroscience, Lausanne University Hospital (CHUV), Lausanne, Switzerland;,These authors contributed equally to this work
| |
Collapse
|
58
|
Mitchell AC, Bharadwaj R, Whittle C, Krueger W, Mirnics K, Hurd Y, Rasmussen T, Akbarian S. The genome in three dimensions: a new frontier in human brain research. Biol Psychiatry 2014; 75:961-9. [PMID: 23958183 PMCID: PMC3925763 DOI: 10.1016/j.biopsych.2013.07.015] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Revised: 06/16/2013] [Accepted: 07/10/2013] [Indexed: 12/21/2022]
Abstract
Less than 1.5% of the human genome encodes protein. However, vast portions of the human genome are subject to transcriptional and epigenetic regulation, and many noncoding regulatory DNA elements are thought to regulate the spatial organization of interphase chromosomes. For example, chromosomal "loopings" are pivotal for the orderly process of gene expression, by enabling distal regulatory enhancer or silencer elements to directly interact with proximal promoter and transcription start sites, potentially bypassing hundreds of kilobases of interspersed sequence on the linear genome. To date, however, epigenetic studies in the human brain are mostly limited to the exploration of DNA methylation and posttranslational modifications of the nucleosome core histones. In contrast, very little is known about the regulation of supranucleosomal structures. Here, we show that chromosome conformation capture, a widely used approach to study higher-order chromatin, is applicable to tissue collected postmortem, thereby informing about genome organization in the human brain. We introduce chromosome conformation capture protocols for brain and compare higher-order chromatin structures at the chromosome 6p22.2-22.1 schizophrenia and bipolar disorder susceptibility locus, and additional neurodevelopmental risk genes, (DPP10, MCPH1) in adult prefrontal cortex and various cell culture systems, including neurons derived from reprogrammed skin cells. We predict that the exploration of three-dimensional genome architectures and function will open up new frontiers in human brain research and psychiatric genetics and provide novel insights into the epigenetic risk architectures of regulatory noncoding DNA.
Collapse
Affiliation(s)
- Amanda C. Mitchell
- Departments of Psychiatry and Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Rahul Bharadwaj
- Departments of Psychiatry and Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY,Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester MA
| | - Catheryne Whittle
- Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester MA
| | - Winfried Krueger
- Center for Regenerative Biology and Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT
| | - Karoly Mirnics
- Department of Psychiatry, Vanderbilt University, Nashville, TN
| | - Yasmin Hurd
- Departments of Psychiatry and Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Theodore Rasmussen
- Center for Regenerative Biology and Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT
| | - Schahram Akbarian
- Departments of Psychiatry and Neuroscience , Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Brudnick Neuropsychiatric Research Institute , University of Massachusetts Medical School, Worcester, Massachusetts.
| |
Collapse
|
59
|
Abstract
PURPOSE OF REVIEW Oxidative stress has become an exciting area of schizophrenia research, and provides ample opportunities and hope for a better understanding of its pathophysiology, which may lead to novel treatment strategies. This review describes how recent methodological advances have allowed the study of oxidative stress to tackle fundamental questions and have provided several conceptual breakthroughs to the field. RECENT FINDINGS Recent human studies support the notion that intrinsic susceptibility to oxidative stress may underlie the pathophysiology of schizophrenia. More than one animal model that may be relevant to study the biology of schizophrenia also shows sign of oxidative stress in the brain. SUMMARY These advances have made this topic of paramount importance to the understanding of schizophrenia and will play a role in advancing the treatment options. This review covers topics from the classic biochemical studies of human biospecimens to the use of magnetic resonance spectroscopy and novel mouse models, and focuses on highlighting the promising areas of research.
Collapse
|
60
|
|
61
|
Darby MM, Sabunciyan S. Repetitive Elements and Epigenetic Marks in Behavior and Psychiatric Disease. ADVANCES IN GENETICS 2014; 86:185-252. [DOI: 10.1016/b978-0-12-800222-3.00009-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
62
|
Imaizumi Y, Okano H. Modeling human neurological disorders with induced pluripotent stem cells. J Neurochem 2013; 129:388-99. [PMID: 24286589 DOI: 10.1111/jnc.12625] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Revised: 11/18/2013] [Accepted: 11/22/2013] [Indexed: 02/06/2023]
Abstract
Human induced pluripotent stem (iPS) cells obtained by reprogramming technology are a source of great hope, not only in terms of applications in regenerative medicine, such as cell transplantation therapy, but also for modeling human diseases and new drug development. In particular, the production of iPS cells from the somatic cells of patients with intractable diseases and their subsequent differentiation into cells at affected sites (e.g., neurons, cardiomyocytes, hepatocytes, and myocytes) has permitted the in vitro construction of disease models that contain patient-specific genetic information. For example, disease-specific iPS cells have been established from patients with neuropsychiatric disorders, including schizophrenia and autism, as well as from those with neurodegenerative diseases, including Parkinson's disease and Alzheimer's disease. A multi-omics analysis of neural cells originating from patient-derived iPS cells may thus enable investigators to elucidate the pathogenic mechanisms of neurological diseases that have heretofore been unknown. In addition, large-scale screening of chemical libraries with disease-specific iPS cells is currently underway and is expected to lead to new drug discovery. Accordingly, this review outlines the progress made via the use of patient-derived iPS cells toward the modeling of neurological disorders, the testing of existing drugs, and the discovery of new drugs. The production of human induced pluripotent stem (iPS) cells from the patients' somatic cells and their subsequent differentiation into specific cells have permitted the in vitro construction of disease models that contain patient-specific genetic information. Furthermore, innovations of gene-editing technologies on iPS cells are enabling new approaches for illuminating the pathogenic mechanisms of human diseases. In this review article, we outlined the current status of neurological diseases-specific iPS cell research and described recently obtained knowledge in the form of actual examples.
Collapse
Affiliation(s)
- Yoichi Imaizumi
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan; Next Generation Systems CFU, Eisai Co. Ltd., Ibaraki, Japan
| | | |
Collapse
|
63
|
ROS and brain diseases: the good, the bad, and the ugly. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2013; 2013:963520. [PMID: 24381719 PMCID: PMC3871919 DOI: 10.1155/2013/963520] [Citation(s) in RCA: 243] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 11/04/2013] [Accepted: 11/05/2013] [Indexed: 12/31/2022]
Abstract
The brain is a major metabolizer of oxygen and yet has relatively feeble protective antioxidant mechanisms. This paper reviews the Janus-faced properties of reactive oxygen species. It will describe the positive aspects of moderately induced ROS but it will also outline recent research findings concerning the impact of oxidative and nitrooxidative stress on neuronal structure and function in neuropsychiatric diseases, including major depression. A common denominator of all neuropsychiatric diseases including schizophrenia and ADHD is an increased inflammatory response of the brain caused either by an exposure to proinflammatory agents during development or an accumulation of degenerated neurons, oxidized proteins, glycated products, or lipid peroxidation in the adult brain. Therefore, modulation of the prooxidant-antioxidant balance provides a therapeutic option which can be used to improve neuroprotection in response to oxidative stress. We also discuss the neuroprotective role of the nuclear factor erythroid 2-related factor (Nrf2) in the aged brain in response to oxidative stressors and nanoparticle-mediated delivery of ROS-scavenging drugs. The antioxidant therapy is a novel therapeutic strategy. However, the available drugs have pleiotropic actions and are not fully characterized in the clinic. Additional clinical trials are needed to assess the risks and benefits of antioxidant therapies for neuropsychiatric disorders.
Collapse
|
64
|
Horiuchi Y, Kano SI, Ishizuka K, Cascella NG, Ishii S, Talbot CC, Jaffe AE, Okano H, Pevsner J, Colantuoni C, Sawa A. Olfactory cells via nasal biopsy reflect the developing brain in gene expression profiles: utility and limitation of the surrogate tissues in research for brain disorders. Neurosci Res 2013; 77:247-50. [PMID: 24120685 PMCID: PMC4097863 DOI: 10.1016/j.neures.2013.09.010] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Revised: 09/23/2013] [Accepted: 09/24/2013] [Indexed: 12/22/2022]
Abstract
Human olfactory cells obtained by rapid nasal biopsy have been suggested to be a good surrogate system to address brain disease-associated molecular changes. Nonetheless, whether use of this experimental strategy is justified remains unclear. Here we compared expression profiles of olfactory cells systematically with those from the brain tissues and other cells. Principal component analysis indicated that the expression profiles of olfactory cells are very different from those of blood cells, but are closer to those of stem cells, in particular mesenchymal stem cells, that can be differentiated into the cells of the central nervous system.
Collapse
Affiliation(s)
- Yasue Horiuchi
- Department of Psychiatry Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Shin-ichi Kano
- Department of Psychiatry Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Koko Ishizuka
- Department of Psychiatry Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nicola G. Cascella
- Department of Psychiatry Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Seiji Ishii
- Department of Physiology, Keio University School of Medicine, Japan
| | - C. Conover Talbot
- Institute for Basic Biomedical Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Japan
| | - Jonathan Pevsner
- Department of Psychiatry Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Hugo W Moser Research Institute at Kennedy Krieger, Baltimore, MD, USA
| | | | - Akira Sawa
- Department of Psychiatry Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
65
|
Bentsen H, Osnes K, Refsum H, Solberg DK, Bøhmer T. A randomized placebo-controlled trial of an omega-3 fatty acid and vitamins E+C in schizophrenia. Transl Psychiatry 2013; 3:e335. [PMID: 24346133 PMCID: PMC3906471 DOI: 10.1038/tp.2013.110] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 10/09/2013] [Accepted: 10/21/2013] [Indexed: 02/07/2023] Open
Abstract
Membrane lipid metabolism and redox regulation may be disturbed in schizophrenia. We examined the clinical effect of adding an omega-3 fatty acid and/or vitamins E+C to antipsychotics. It was hypothesized that lower baseline levels of polyunsaturated fatty acids (PUFAs) would predict more benefit from the add-on treatment. The trial had a multicenter, randomized, double-blind, placebo-controlled 2 × 2 factorial design. Patients aged 18-39 years with schizophrenia or related psychoses were consecutively included at admission to psychiatric departments in Norway. They received active or placebo ethyl-eicosapentaenoate (EPA) 2 g day⁻¹ and active or placebo vitamin E 364 mg day⁻¹+vitamin C 1000 mg day⁻¹ (vitamins) for 16 weeks. The main outcome measures were Positive and Negative Syndrome Scale (PANSS) total and subscales scores, analyzed by linear mixed models. Ninety-nine patients were included. At baseline, erythrocyte PUFA were measured in 97 subjects. Given separately, EPA and vitamins increased drop-out rates, whereas when combined they did not differ from placebo. In low PUFA patients, EPA alone impaired the course of total PANSS (Cohen's d=0.29; P=0.03) and psychotic symptoms (d=0.40; P=0.003), especially persecutory delusions (d=0.48; P=0.0004). Vitamins alone impaired the course of psychotic symptoms (d= 0.37; P=0.005), especially persecutory delusions (d=0.47; P=0.0005). Adding vitamins to EPA neutralized the detrimental effect on psychosis (interaction d=0.31; P=0.02). In high PUFA patients, there were no significant effects of trial drugs on PANSS scales. In conclusion, given separately during an acute episode, EPA and vitamins E+C induce psychotic symptoms in patients with low levels of PUFA. Combined, these agents seem safe.
Collapse
Affiliation(s)
- H Bentsen
- Center for Psychopharmacology, Diakonhjemmet Hospital, Oslo, Norway,Division of Psychiatry, Oslo University Hospital, Aker, Oslo, Norway,Center for Psychopharmacology, Diakonhjemmet Hospital, P.o.b. 85, Vinderen, Oslo 0319, Norway. E-mail:
| | - K Osnes
- Department of Psychosomatic Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - H Refsum
- Center for Psychopharmacology, Diakonhjemmet Hospital, Oslo, Norway
| | - D K Solberg
- Center for Psychopharmacology, Diakonhjemmet Hospital, Oslo, Norway
| | - T Bøhmer
- Nutritional Laboratory, Department of Medical Biochemistry, Oslo University Hospital, Aker, Oslo, Norway
| |
Collapse
|
66
|
Bharadwaj R, Jiang Y, Mao W, Jakovcevski M, Dincer A, Krueger W, Garbett K, Whittle C, Tushir JS, Liu J, Sequeira A, Vawter MP, Gardner PD, Casaccia P, Rasmussen T, Bunney WE, Mirnics K, Futai K, Akbarian S. Conserved chromosome 2q31 conformations are associated with transcriptional regulation of GAD1 GABA synthesis enzyme and altered in prefrontal cortex of subjects with schizophrenia. J Neurosci 2013; 33:11839-51. [PMID: 23864674 PMCID: PMC3713726 DOI: 10.1523/jneurosci.1252-13.2013] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Revised: 05/16/2013] [Accepted: 06/12/2013] [Indexed: 01/07/2023] Open
Abstract
Little is known about chromosomal loopings involving proximal promoter and distal enhancer elements regulating GABAergic gene expression, including changes in schizophrenia and other psychiatric conditions linked to altered inhibition. Here, we map in human chromosome 2q31 the 3D configuration of 200 kb of linear sequence encompassing the GAD1 GABA synthesis enzyme gene locus, and we describe a loop formation involving the GAD1 transcription start site and intergenic noncoding DNA elements facilitating reporter gene expression. The GAD1-TSS(-50kbLoop) was enriched with nucleosomes epigenetically decorated with the transcriptional mark, histone H3 trimethylated at lysine 4, and was weak or absent in skin fibroblasts and pluripotent stem cells compared with neuronal cultures differentiated from them. In the prefrontal cortex of subjects with schizophrenia, GAD1-TSS(-50kbLoop) was decreased compared with controls, in conjunction with downregulated GAD1 expression. We generated transgenic mice expressing Gad2 promoter-driven green fluorescent protein-conjugated histone H2B and confirmed that Gad1-TSS(-55kbLoop), the murine homolog to GAD1-TSS(-50kbLoop), is a chromosomal conformation specific for GABAergic neurons. In primary neuronal culture, Gad1-TSS(-55kbLoop) and Gad1 expression became upregulated when neuronal activity was increased. We conclude that 3D genome architectures, including chromosomal loopings for promoter-enhancer interactions involved in the regulation of GABAergic gene expression, are conserved between the rodent and primate brain, and subject to developmental and activity-dependent regulation, and disordered in some cases with schizophrenia. More broadly, the findings presented here draw a connection between noncoding DNA, spatial genome architecture, and neuronal plasticity in development and disease.
Collapse
Affiliation(s)
- Rahul Bharadwaj
- Graduate School of Biomedical Sciences and
- Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester, Massachusetts 01655
| | - Yan Jiang
- Departments of Psychiatry and Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Wenjie Mao
- Graduate School of Biomedical Sciences and
- Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester, Massachusetts 01655
| | | | - Aslihan Dincer
- Departments of Psychiatry and Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Winfried Krueger
- Center for Regenerative Biology and Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut 06269
| | - Krassimira Garbett
- Department of Psychiatry, Vanderbilt University, Nashville, Tennessee 37232, and
| | - Catheryne Whittle
- Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester, Massachusetts 01655
| | - Jogender Singh Tushir
- Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester, Massachusetts 01655
| | - Jia Liu
- Departments of Psychiatry and Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Adolfo Sequeira
- Department of Psychiatry and Human Behavior, University of California, Irvine, California 92697
| | - Marquis P. Vawter
- Department of Psychiatry and Human Behavior, University of California, Irvine, California 92697
| | - Paul D. Gardner
- Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester, Massachusetts 01655
| | - Patrizia Casaccia
- Departments of Psychiatry and Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Theodore Rasmussen
- Center for Regenerative Biology and Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut 06269
| | - William E. Bunney
- Department of Psychiatry and Human Behavior, University of California, Irvine, California 92697
| | - Karoly Mirnics
- Department of Psychiatry, Vanderbilt University, Nashville, Tennessee 37232, and
| | - Kensuke Futai
- Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester, Massachusetts 01655
| | - Schahram Akbarian
- Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester, Massachusetts 01655
- Departments of Psychiatry and Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| |
Collapse
|
67
|
Hasan A, Mitchell A, Schneider A, Halene T, Akbarian S. Epigenetic dysregulation in schizophrenia: molecular and clinical aspects of histone deacetylase inhibitors. Eur Arch Psychiatry Clin Neurosci 2013; 263:273-84. [PMID: 23381549 DOI: 10.1007/s00406-013-0395-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2012] [Accepted: 01/18/2013] [Indexed: 12/22/2022]
Abstract
Notwithstanding the considerable advances in the treatment options for schizophrenia, the cognitive symptoms in particular are not receptive to antipsychotic treatment and considered one of the main predictors for poor social and functional outcome of the disease. Recent findings in preclinical model systems indicate that epigenetic modulation might emerge as a promising target for the treatment of cognitive disorders. The aim of this review is to introduce some of the principles of chromatin biology to the reader and to discuss a possible role in the neurobiology and pathophysiology of schizophrenia. We will discuss potential epigenetic targets for drug therapy, including histone deacetylase inhibitors (HDACi). In a second part, conceptual and practical challenges associated with clinical trials of chromatin-modifying drugs in psychiatric patient populations are discussed, including safety profiles, the potential for adverse effects and general issues revolving around pharmacokinetics and pharmacodynamics. Additional investigations are required in order to fully evaluate the potential of HDACi and similar "epigenetic therapies" as novel treatment options for schizophrenia and other psychotic disease.
Collapse
Affiliation(s)
- Alkomiet Hasan
- Deparment of Psychiatry and Psychotherapy, Ludwig-Maximilians-University, Nussbaumstr. 7, 80336 Munich, Germany.
| | | | | | | | | |
Collapse
|
68
|
Ballesteros A, Jiang P, Summerfelt A, Du X, Chiappelli J, O’Donnell P, Kochunov P, Hong LE. No evidence of exogenous origin for the abnormal glutathione redox state in schizophrenia. Schizophr Res 2013; 146:184-9. [PMID: 23466187 PMCID: PMC3622807 DOI: 10.1016/j.schres.2013.02.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2013] [Accepted: 02/04/2013] [Indexed: 11/29/2022]
Abstract
Schizophrenia has been associated with low glutathione (GSH), one of the most important substrates for natural defense against oxidative stress. This abnormality is often attributed to genetic or other pathological causes. However, low GSH in schizophrenia could also be due to insufficient antioxidant consumption or other exogenous factors. We evaluated GSH in relation to diet, smoking, and medication status in schizophrenia patients. We recruited 54 participants (29 schizophrenia patients and 25 normal controls). The Antioxidant Dietary Source Questions was used to estimate the total antioxidant capacity (TAC) from participants' diet. GSH and the oxidized form of glutathione (GSSG) were assayed. We found that GSH was significantly lower (p<0.001) while %GSSG was 2 to 5 fold higher (p = 0.023) in patients compared with controls. No evidence for lower TAC dietary intake was found in schizophrenia patients compared with controls; rather nominally higher TAC level was found in the patients diet (p = 0.02). Analysis of consumption of individual food categories also failed to find evidence of reduced dietary antioxidant intake in schizophrenia patients. Smoking and medications did not significantly predict the GSH deficit either. However, there was a significant smoking by diagnosis interaction on GSH (p = 0.026) such that smoking was associated with higher GSH level in controls while smoking in patients was not associated with this effect. Schizophrenia patients may have an impaired upregulation of GSH synthesis that normally occurs due to smoking-induced antioxidative response. Lower GSH was independently present in patients on clozapine (p = 0.005) and patients on other antipsychotics (p<0.001) compared with controls. In conclusion, none of the exogenous sources played a major role in explaining abnormalities in the glutathione pathway in patients. The state of abnormal glutathione redox may therefore be a part of schizophrenia pathophysiology.
Collapse
Affiliation(s)
- Alejandro Ballesteros
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine. 55 Wade Avenue, 21228, Catonsville, MD, USA,Hospital Clínico de Salamanca, Department of Psychiatry. Calle de San Vicente, 182. 37007, Salamanca, Spain
| | - Pan Jiang
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine. 55 Wade Avenue, 21228, Catonsville, MD, USA
| | - Ann Summerfelt
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine. 55 Wade Avenue, 21228, Catonsville, MD, USA
| | - Xiaoming Du
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine. 55 Wade Avenue, 21228, Catonsville, MD, USA
| | - Joshua Chiappelli
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine. 55 Wade Avenue, 21228, Catonsville, MD, USA
| | - Patricio O’Donnell
- Department of Anatomy & Neurobiology and Psychiatry, University of Maryland School of Medicine. 685 West Baltimore Street, 21201, Baltimore, MD, USA
| | - Peter Kochunov
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine. 55 Wade Avenue, 21228, Catonsville, MD, USA
| | - L. Elliot Hong
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine. 55 Wade Avenue, 21228, Catonsville, MD, USA,Address reprint requests to Dr. Hong, Maryland Psychiatric Research Center, P.O. Box 21247, Baltimore, MD 21228. Tel: 410 402 6828. Fax: 410 402 6023.
| |
Collapse
|
69
|
Shulha HP, Cheung I, Guo Y, Akbarian S, Weng Z. Coordinated cell type-specific epigenetic remodeling in prefrontal cortex begins before birth and continues into early adulthood. PLoS Genet 2013; 9:e1003433. [PMID: 23593028 PMCID: PMC3623761 DOI: 10.1371/journal.pgen.1003433] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Accepted: 02/20/2013] [Indexed: 11/21/2022] Open
Abstract
Development of prefrontal and other higher-order association cortices is associated with widespread changes in the cortical transcriptome, particularly during the transitions from prenatal to postnatal development, and from early infancy to later stages of childhood and early adulthood. However, the timing and longitudinal trajectories of neuronal gene expression programs during these periods remain unclear in part because of confounding effects of concomitantly occurring shifts in neuron-to-glia ratios. Here, we used cell type–specific chromatin sorting techniques for genome-wide profiling of a histone mark associated with transcriptional regulation—H3 with trimethylated lysine 4 (H3K4me3)—in neuronal chromatin from 31 subjects from the late gestational period to 80 years of age. H3K4me3 landscapes of prefrontal neurons were developmentally regulated at 1,157 loci, including 768 loci that were proximal to transcription start sites. Multiple algorithms consistently revealed that the overwhelming majority and perhaps all of developmentally regulated H3K4me3 peaks were on a unidirectional trajectory defined by either rapid gain or loss of histone methylation during the late prenatal period and the first year after birth, followed by similar changes but with progressively slower kinetics during early and later childhood and only minimal changes later in life. Developmentally downregulated H3K4me3 peaks in prefrontal neurons were enriched for Paired box (Pax) and multiple Signal Transducer and Activator of Transcription (STAT) motifs, which are known to promote glial differentiation. In contrast, H3K4me3 peaks subject to a progressive increase in maturing prefrontal neurons were enriched for activating protein-1 (AP-1) recognition elements that are commonly associated with activity-dependent regulation of neuronal gene expression. We uncovered a developmental program governing the remodeling of neuronal histone methylation landscapes in the prefrontal cortex from the late prenatal period to early adolescence, which is linked to cis-regulatory sequences around transcription start sites. Prolonged maturation of the human cerebral cortex, which extends into the third decade of life, is critical for proper development of executive functions such as higher-order problem-solving and complex cognition. Little is known about changes of post-mitotic neurons during this prolonged maturation period, including changes in epigenetic regulation, and more broadly, in genome organization and function. Such knowledge is critical for a deeper understanding of human development, cognitive abilities, and psychiatric diseases. Here, we identify 1,157 genomic loci in neuronal cells from the prefrontal cortex that show developmental changes in a chromatin mark, histone H3 trimethylated at lysine 4 (H3K4me3), which has been associated with regulation of gene expression. Interestingly, the overwhelming majority of these developmentally regulated H3K4me3 peaks were defined by rapid gain or loss of histone methylation during the late prenatal period and the first year after birth, followed by slower changes during early and later childhood and minimal changes thereafter. The genomic sequences showing these dynamic changes in H3K4me3 were enriched with distinct transcription factor motifs. Our findings suggest that there is highly regulated, pre-programmed remodeling of neuronal histone methylation landscapes in the human brain that begins before birth and continues into adolescence.
Collapse
Affiliation(s)
- Hennady P. Shulha
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Iris Cheung
- Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Yin Guo
- Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Schahram Akbarian
- Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- Departments of Psychiatry and Neuroscience, Mount Sinai School of Medicine, New York, New York, United States of America
- * E-mail: (SA); (ZW)
| | - Zhiping Weng
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- * E-mail: (SA); (ZW)
| |
Collapse
|
70
|
Mor E, Kano SI, Colantuoni C, Sawa A, Navon R, Shomron N. MicroRNA-382 expression is elevated in the olfactory neuroepithelium of schizophrenia patients. Neurobiol Dis 2013; 55:1-10. [PMID: 23542694 DOI: 10.1016/j.nbd.2013.03.011] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2012] [Revised: 03/17/2013] [Accepted: 03/20/2013] [Indexed: 02/06/2023] Open
Abstract
Schizophrenia is a common neuropsychiatric disorder that has a strong genetic component. MicroRNAs (miRNAs) have been implicated in neurodevelopmental and psychiatric disorders including schizophrenia, as indicated by their dysregulation in post-mortem brain tissues and in peripheral blood of schizophrenia patients. The olfactory epithelium (OE) is one of the few accessible neural tissues that contain neurons and their stem cells. Previous studies showed that OE-derived tissues and cells can be safely and easily collected from live human subjects and may provide a "window" into neuronal processes involved in disorders such as schizophrenia, while avoiding the limitations of using postmortem brain samples or non-neuronal tissues. In this study, we found that the brain-enriched miR-382 (miR-382-5p) expression was elevated in in vitro cultured olfactory cells, in a cohort of seven schizophrenia patients compared with seven non-schizophrenic controls. MiR-382 elevation was further confirmed in laser-capture microdissected OE neuronal tissue (LCM-OE), enriched for mature olfactory neurons, in a cohort of 18 schizophrenia patients and 18 non-schizophrenic controls. In sharp contrast, miR-382 expression could not be detected in lymphoblastoid cell lines generated from schizophrenic or non-schizophrenic individuals. We further found that miR-382 directly regulates the expression of two genes, FGFR1 and SPRY4, which are downregulated in both the cultured olfactory cells and LCM-OE derived from schizophrenia patients. These genes are involved in the fibroblast growth factor (FGF) signaling pathway, while impairment of this pathway may underlie abnormal brain development and function associated with schizophrenia. Our data suggest that miR-382 elevation detected in patients' OE-derived samples might serve to strengthen current biomarker studies in schizophrenia. This study also illustrates the potential utility of OE-derived tissues and cells as surrogate samples for the brain.
Collapse
Affiliation(s)
- Eyal Mor
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.
| | | | | | | | | | | |
Collapse
|
71
|
Nishioka M, Bundo M, Kasai K, Iwamoto K. DNA methylation in schizophrenia: progress and challenges of epigenetic studies. Genome Med 2012; 4:96. [PMID: 23234572 PMCID: PMC3580436 DOI: 10.1186/gm397] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Schizophrenia is a severe psychiatric disease affecting about 1% of the world's population, with significant effects on patients and society. Genetic studies have identified several candidate risk genes or genomic regions for schizophrenia, and epidemiological studies have revealed several environmental risk factors. However, the etiology of schizophrenia still remains largely unknown. Epigenetic mechanisms such as DNA methylation and histone modifications can explain the interaction between genetic and environmental factors at the molecular level, and accumulating evidence suggests that such epigenetic alterations are involved in the pathophysiology of schizophrenia. However, replication studies to validate previous findings and investigations of the causality of epigenetic alterations in schizophrenia are needed. Here, we review epigenetic studies of schizophrenia patients using postmortem brains or peripheral tissues, focusing mainly on DNA methylation. We also highlight the recent progress and challenges in characterizing the potentially complex and dynamic patterns of epigenomic variations. Such studies are expected to contribute to our understanding of schizophrenia etiology and should provide novel opportunities for the development of therapeutic drugs.
Collapse
Affiliation(s)
- Masaki Nishioka
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 1138655, Japan
- Department of Molecular Psychiatry, Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 1138655, Japan
| | - Miki Bundo
- Department of Molecular Psychiatry, Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 1138655, Japan
| | - Kiyoto Kasai
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 1138655, Japan
| | - Kazuya Iwamoto
- Department of Molecular Psychiatry, Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 1138655, Japan
- PRESTO, Japan Science and Technology Agency, 4-1-8, Honcho, Kawaguchi-shi, Saitama 332-0012, Japan
| |
Collapse
|