251
|
Abstract
The remarkable advances in cellular reprogramming have made it possible to generate a renewable source of human neurons from fibroblasts obtained from skin samples of neonates and adults. As a result, we can now investigate the etiology of neurological diseases at the cellular level using neuronal populations derived from patients, which harbor the same genetic mutations thought to be relevant to the risk for pathology. Therapeutic implications include the ability to establish new humanized disease models for understanding mechanisms, conduct high-throughput screening for novel biogenic compounds to reverse or prevent the disease phenotype, identify and engineer genetic rescue of causal mutations, and develop patient-specific cellular replacement strategies. Although this field offers enormous potential for understanding and treating neurological disease, there are still many issues that must be addressed before we can fully exploit this technology. Here we summarize several recent studies presented at a symposium at the 2011 annual meeting of the Society for Neuroscience, which highlight innovative approaches to cellular reprogramming and how this revolutionary technique is being refined to model neurodevelopmental and neurodegenerative diseases, such as autism spectrum disorders, schizophrenia, familial dysautonomia, and Alzheimer's disease.
Collapse
|
252
|
Van de Kerkhof NW, Feenstra I, van der Heijden FM, de Leeuw N, Pfundt R, Stöber G, Egger JI, Verhoeven WM. Copy number variants in a sample of patients with psychotic disorders: is standard screening relevant for actual clinical practice? Neuropsychiatr Dis Treat 2012; 8:295-300. [PMID: 22848183 PMCID: PMC3404708 DOI: 10.2147/ndt.s32903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
With the introduction of new genetic techniques such as genome-wide array comparative genomic hybridization, studies on the putative genetic etiology of schizophrenia have focused on the detection of copy number variants (CNVs), ie, microdeletions and/or microduplications, that are estimated to be present in up to 3% of patients with schizophrenia. In this study, out of a sample of 100 patients with psychotic disorders, 80 were investigated by array for the presence of CNVs. The assessment of the severity of psychiatric symptoms was performed using standardized instruments and ICD-10 was applied for diagnostic classification. In three patients, a submicroscopic CNV was demonstrated, one with a loss in 1q21.1 and two with a gain in 1p13.3 and 7q11.2, respectively. The association between these or other CNVs and schizophrenia or schizophrenia-like psychoses and their clinical implications still remain equivocal. While the CNV affected genes may enhance the vulnerability for psychiatric disorders via effects on neuronal architecture, these insights have not resulted in major changes in clinical practice as yet. Therefore, genome-wide array analysis should presently be restricted to those patients in whom psychotic symptoms are paired with other signs, particularly dysmorphisms and intellectual impairment.
Collapse
Affiliation(s)
- Noortje Wa Van de Kerkhof
- Vincent van Gogh Institute for Psychiatry, Centre of Excellence for Neuropsychiatry, Venray, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
253
|
Yin DM, Chen YJ, Sathyamurthy A, Xiong WC, Mei L. Synaptic dysfunction in schizophrenia. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 970:493-516. [PMID: 22351070 DOI: 10.1007/978-3-7091-0932-8_22] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Schizophrenia alters basic brain processes of perception, emotion, and judgment to cause hallucinations, delusions, thought disorder, and cognitive deficits. Unlike neurodegeneration diseases that have irreversible neuronal degeneration and death, schizophrenia lacks agreeable pathological hallmarks, which makes it one of the least understood psychiatric disorders. With identification of schizophrenia susceptibility genes, recent studies have begun to shed light on underlying pathological mechanisms. Schizophrenia is believed to result from problems during neural development that lead to improper function of synaptic transmission and plasticity, and in agreement, many of the susceptibility genes encode proteins critical for neural development. Some, however, are also expressed at high levels in adult brain. Here, we will review evidence for altered neurotransmission at glutamatergic, GABAergic, dopaminergic, and cholinergic synapses in schizophrenia and discuss roles of susceptibility genes in neural development as well as in synaptic plasticity and how their malfunction may contribute to pathogenic mechanisms of schizophrenia. We propose that mouse models with precise temporal and spatial control of mutation or overexpression would be useful to delineate schizophrenia pathogenic mechanisms.
Collapse
Affiliation(s)
- Dong-Min Yin
- Department of Neurology, Institute of Molecular Medicine and Genetics, Georgia Health Sciences University, Augusta, GA 30912, USA
| | | | | | | | | |
Collapse
|
254
|
Claes S, Tang YL, Gillespie CF, Cubells JF. Human genetics of schizophrenia. HANDBOOK OF CLINICAL NEUROLOGY 2012; 106:37-52. [DOI: 10.1016/b978-0-444-52002-9.00003-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
255
|
Arguello PA, Gogos JA. Genetic and cognitive windows into circuit mechanisms of psychiatric disease. Trends Neurosci 2012; 35:3-13. [PMID: 22177981 DOI: 10.1016/j.tins.2011.11.007] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2011] [Revised: 10/26/2011] [Accepted: 11/18/2011] [Indexed: 01/12/2023]
Affiliation(s)
- P Alexander Arguello
- Princeton Neuroscience Institute and Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | | |
Collapse
|
256
|
Abstract
Alterations in neurodevelopment are thought to modify risk of numerous psychiatric disorders, including schizophrenia, autism, ADHD, mood and anxiety disorders, and substance abuse. However, little is known about the cellular and molecular changes that guide these neurodevelopmental changes and how they contribute to mental illness. In this review, we suggest that elucidating this process in humans requires the use of model organisms. Furthermore, we advocate that such translational work should focus on the role that genes and/or environmental factors play in the development of circuits that regulate specific physiological and behavioral outcomes in adulthood. This emphasis on circuit development, as a fundamental unit for understanding behavior, is distinct from current approaches of modeling psychiatric illnesses in animals in two important ways. First, it proposes to replace the diagnostic and statistical manual of mental disorders (DSM) diagnostic system with measurable endophenotypes as the basis for modeling human psychopathology in animals. We argue that a major difficulty in establishing valid animal models lies in their reliance on the DSM/International Classification of Diseases conceptual framework, and suggest that the Research Domain Criteria project, recently proposed by the NIMH, provides a more suitable system to model human psychopathology in animals. Second, this proposal emphasizes the developmental origin of many (though clearly not all) psychiatric illnesses, an issue that is often glossed over in current animal models of mental illness. We suggest that animal models are essential to elucidate the mechanisms by which neurodevelopmental changes program complex behavior in adulthood. A better understanding of this issue, in animals, is the key for defining human psychopathology, and the development of earlier and more effective interventions for mental illness.
Collapse
|
257
|
Brandon NJ, Sawa A. Linking neurodevelopmental and synaptic theories of mental illness through DISC1. Nat Rev Neurosci 2011; 12:707-22. [PMID: 22095064 DOI: 10.1038/nrn3120] [Citation(s) in RCA: 337] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Recent advances in our understanding of the underlying genetic architecture of psychiatric disorders has blown away the diagnostic boundaries that are defined by currently used diagnostic manuals. The disrupted in schizophrenia 1 (DISC1) gene was originally discovered at the breakpoint of an inherited chromosomal translocation, which segregates with major mental illnesses. In addition, many biological studies have indicated a role for DISC1 in early neurodevelopment and synaptic regulation. Given that DISC1 is thought to drive a range of endophenotypes that underlie major mental conditions, elucidating the biology of DISC1 may enable the construction of new diagnostic categories for mental illnesses with a more meaningful biological foundation.
Collapse
|
258
|
Corvin AP. Two patients walk into a clinic...a genomics perspective on the future of schizophrenia. BMC Biol 2011; 9:77. [PMID: 22078159 PMCID: PMC3214150 DOI: 10.1186/1741-7007-9-77] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Accepted: 11/07/2011] [Indexed: 12/17/2022] Open
Abstract
Progress is being made in schizophrenia genomics, suggesting that this complex brain disorder involves rare, moderate to high-risk mutations and the cumulative impact of small genetic effects, coupled with environmental factors. The genetic heterogeneity underlying schizophrenia and the overlap with other neurodevelopmental disorders suggest that it will not continue to be viewed as a single disease. This has radical implications for clinical practice, as diagnosis and treatment will be guided by molecular etiology rather than clinical diagnostic criteria.
Collapse
Affiliation(s)
- Aiden P Corvin
- Department of Psychiatry, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
259
|
Hochstenbach R, Buizer-Voskamp JE, Vorstman JAS, Ophoff RA. Genome arrays for the detection of copy number variations in idiopathic mental retardation, idiopathic generalized epilepsy and neuropsychiatric disorders: lessons for diagnostic workflow and research. Cytogenet Genome Res 2011; 135:174-202. [PMID: 22056632 DOI: 10.1159/000332928] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2022] Open
Abstract
We review the contributions and limitations of genome-wide array-based identification of copy number variants (CNVs) in the clinical diagnostic evaluation of patients with mental retardation (MR) and other brain-related disorders. In unselected MR referrals a causative genomic gain or loss is detected in 14-18% of cases. Usually, such CNVs arise de novo, are not found in healthy subjects, and have a major impact on the phenotype by altering the dosage of multiple genes. This high diagnostic yield justifies array-based segmental aneuploidy screening as the initial genetic test in these patients. This also pertains to patients with autism (expected yield about 5-10% in nonsyndromic and 10-20% in syndromic patients) and schizophrenia (at least 5% yield). CNV studies in idiopathic generalized epilepsy, attention-deficit hyperactivity disorder, major depressive disorder and Tourette syndrome indicate that patients have, on average, a larger CNV burden as compared to controls. Collectively, the CNV studies suggest that a wide spectrum of disease-susceptibility variants exists, most of which are rare (<0.1%) and of variable and usually small effect. Notwithstanding, a rare CNV can have a major impact on the phenotype. Exome sequencing in MR and autism patients revealed de novo mutations in protein coding genes in 60 and 20% of cases, respectively. Therefore, it is likely that arrays will be supplanted by next-generation sequencing methods as the initial and perhaps ultimate diagnostic tool in patients with brain-related disorders, revealing both CNVs and mutations in a single test.
Collapse
Affiliation(s)
- R Hochstenbach
- Division of Biomedical Genetics, Department of Medical Genetics, University Medical Centre Utrecht, Utrecht, The Netherlands.
| | | | | | | |
Collapse
|
260
|
Carless MA, Glahn DC, Johnson MP, Curran JE, Bozaoglu K, Dyer TD, Winkler AM, Cole SA, Almasy L, MacCluer JW, Duggirala R, Moses EK, Göring HHH, Blangero J. Impact of DISC1 variation on neuroanatomical and neurocognitive phenotypes. Mol Psychiatry 2011; 16:1096-104, 1063. [PMID: 21483430 PMCID: PMC3135724 DOI: 10.1038/mp.2011.37] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2010] [Revised: 01/19/2011] [Accepted: 02/17/2011] [Indexed: 02/03/2023]
Abstract
Although disrupted in schizophrenia 1 (DISC1) has been implicated in many psychiatric disorders, including schizophrenia, bipolar disorder, schizoaffective disorder and major depression, its biological role in these disorders is unclear. To better understand this gene and its role in psychiatric disease, we conducted transcriptional profiling and genome-wide association analysis in 1232 pedigreed Mexican-American individuals for whom we have neuroanatomic images, neurocognitive assessments and neuropsychiatric diagnoses. SOLAR was used to determine heritability, identify gene expression patterns and perform association analyses on 188 quantitative brain-related phenotypes. We found that the DISC1 transcript is highly heritable (h(2)=0.50; P=1.97 × 10(-22)), and that gene expression is strongly cis-regulated (cis-LOD=3.89) but is also influenced by trans-effects. We identified several DISC1 polymorphisms that were associated with cortical gray matter thickness within the parietal, temporal and frontal lobes. Associated regions affiliated with memory included the entorhinal cortex (rs821639, P=4.11 × 10(-5); rs2356606, P=4.71 × 10(-4)), cingulate cortex (rs16856322, P=2.88 × 10(-4)) and parahippocampal gyrus (rs821639, P=4.95 × 10(-4)); those affiliated with executive and other cognitive processing included the transverse temporal gyrus (rs9661837, P=5.21 × 10(-4); rs17773946, P=6.23 × 10(-4)), anterior cingulate cortex (rs2487453, P=4.79 × 10(-4); rs3738401, P=5.43 × 10(-4)) and medial orbitofrontal cortex (rs9661837; P=7.40 × 10(-4)). Cognitive measures of working memory (rs2793094, P=3.38 × 10(-4)), as well as lifetime history of depression (rs4658966, P=4.33 × 10(-4); rs12137417, P=4.93 × 10(-4)) and panic (rs12137417, P=7.41 × 10(-4)) were associated with DISC1 sequence variation. DISC1 has well-defined genetic regulation and clearly influences important phenotypes related to psychiatric disease.
Collapse
Affiliation(s)
- M A Carless
- Department of Genetics, Southwest Foundation for Biomedical Research, San Antonio, TX 78227, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
261
|
Buizer-Voskamp JE, Muntjewerff JW, Genetic Risk and Outcome in Psychosis (GROUP) Consortium, Strengman E, Sabatti C, Stefansson H, Vorstman JAS, Ophoff RA. Genome-wide analysis shows increased frequency of copy number variation deletions in Dutch schizophrenia patients. Biol Psychiatry 2011; 70:655-62. [PMID: 21489405 PMCID: PMC3137747 DOI: 10.1016/j.biopsych.2011.02.015] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Collaborators] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2010] [Revised: 02/09/2011] [Accepted: 02/11/2011] [Indexed: 01/30/2023]
Abstract
BACKGROUND Since 2008, multiple studies have reported on copy number variations (CNVs) in schizophrenia. However, many regions are unique events with minimal overlap between studies. This makes it difficult to gain a comprehensive overview of all CNVs involved in the etiology of schizophrenia. We performed a systematic CNV study on the basis of a homogeneous genome-wide dataset aiming at all CNVs ≥ 50 kilobase pair. We complemented this analysis with a review of cytogenetic and chromosomal abnormalities for schizophrenia reported in the literature with the purpose of combining classical genetic findings and our current understanding of genomic variation. METHODS We investigated 834 Dutch schizophrenia patients and 672 Dutch control subjects. The CNVs were included if they were detected by QuantiSNP (http://www.well.ox.ac.uk/QuantiSNP/) as well as PennCNV (http://www.neurogenome.org/cnv/penncnv/) and contain known protein coding genes. The integrated identification of CNV regions and cytogenetic loci indicates regions of interest (cytogenetic regions of interest [CROIs]). RESULTS In total, 2437 CNVs were identified with an average number of 2.1 CNVs/subject for both cases and control subjects. We observed significantly more deletions but not duplications in schizophrenia cases versus control subjects. The CNVs identified coincide with loci previously reported in the literature, confirming well-established schizophrenia CROIs 1q42 and 22q11.2 as well as indicating a potentially novel CROI on chromosome 5q35.1. CONCLUSIONS Chromosomal deletions are more prevalent in schizophrenia patients than in healthy subjects and therefore confer a risk factor for pathogenicity. The combination of our CNV data with previously reported cytogenetic abnormalities in schizophrenia provides an overview of potentially interesting regions for positional candidate genes.
Collapse
Affiliation(s)
- Jacobine E Buizer-Voskamp
- Rudolf Magnus Institute of Neuroscience, Department of Psychiatry, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
- Department of Medical Genetics, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG Utrecht, The Netherlands
| | - Jan-Willem Muntjewerff
- Department of Psychiatry, University Medical Centre St Radboud, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| | | | - Eric Strengman
- Department of Medical Genetics, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG Utrecht, The Netherlands
| | - Chiara Sabatti
- Department of Health Research and Policy, Stanford University School of Medicine, HRP Redwood Building, Stanford, CA 94305-5405, USA
| | - Hreinn Stefansson
- CNS Division, deCODE genetics, Sturlugata 8, IS-101 Reykjavik, Iceland
| | - Jacob AS Vorstman
- Rudolf Magnus Institute of Neuroscience, Department of Psychiatry, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Roel A Ophoff
- Rudolf Magnus Institute of Neuroscience, Department of Psychiatry, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
- Department of Medical Genetics, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG Utrecht, The Netherlands
- Center for Neurobehavioral Genetics, University of California, 695 Charles E Young Drive South, Los Angeles, CA 90095, USA
| |
Collapse
Collaborators
René S Kahn, Don H Linszen, Jim van Os, Durk Wiersma, Richard Bruggeman, Wiepke Cahn, Lieuwe de Haan, Lydia Krabbendam, Inez Myin-Germeys,
Collapse
|
262
|
Wang Q, Charych EI, Pulito VL, Lee JB, Graziane NM, Crozier RA, Revilla-Sanchez R, Kelly MP, Dunlop AJ, Murdoch H, Taylor N, Xie Y, Pausch M, Hayashi-Takagi A, Ishizuka K, Seshadri S, Bates B, Kariya K, Sawa A, Weinberg RJ, Moss SJ, Houslay MD, Yan Z, Brandon NJ. The psychiatric disease risk factors DISC1 and TNIK interact to regulate synapse composition and function. Mol Psychiatry 2011; 16:1006-23. [PMID: 20838393 PMCID: PMC3176992 DOI: 10.1038/mp.2010.87] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2010] [Revised: 07/18/2010] [Accepted: 07/26/2010] [Indexed: 02/08/2023]
Abstract
Disrupted in schizophrenia 1 (DISC1), a genetic risk factor for multiple serious psychiatric diseases including schizophrenia, bipolar disorder and autism, is a key regulator of multiple neuronal functions linked to both normal development and disease processes. As these diseases are thought to share a common deficit in synaptic function and architecture, we have analyzed the role of DISC1 using an approach that focuses on understanding the protein-protein interactions of DISC1 specifically at synapses. We identify the Traf2 and Nck-interacting kinase (TNIK), an emerging risk factor itself for disease, as a key synaptic partner for DISC1, and provide evidence that the DISC1-TNIK interaction regulates synaptic composition and activity by stabilizing the levels of key postsynaptic density proteins. Understanding the novel DISC1-TNIK interaction is likely to provide insights into the etiology and underlying synaptic deficits found in major psychiatric diseases.
Collapse
Affiliation(s)
- Q Wang
- Pfizer Neuroscience Research Unit, Princeton, NJ, USA
- Pfizer Neuroscience Research Unit, Pfizer, Groton, CT, USA
| | - EI Charych
- Pfizer Neuroscience Research Unit, Princeton, NJ, USA
- Pfizer Neuroscience Research Unit, Pfizer, Groton, CT, USA
| | - VL Pulito
- Pfizer Neuroscience Research Unit, Princeton, NJ, USA
| | - JB Lee
- Department of Physiology and Biophysics, University at Buffalo, SUNY, Buffalo, NY, USA
| | - NM Graziane
- Department of Physiology and Biophysics, University at Buffalo, SUNY, Buffalo, NY, USA
| | - RA Crozier
- Pfizer Neuroscience Research Unit, Princeton, NJ, USA
| | - R Revilla-Sanchez
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| | - MP Kelly
- Pfizer Neuroscience Research Unit, Princeton, NJ, USA
- Pfizer Neuroscience Research Unit, Pfizer, Groton, CT, USA
| | - AJ Dunlop
- Neuroscience and Molecular Pharmacology, Faculty of Biomedical and Life Sciences, University of Glasgow, Scotland, UK
| | - H Murdoch
- Neuroscience and Molecular Pharmacology, Faculty of Biomedical and Life Sciences, University of Glasgow, Scotland, UK
| | - N Taylor
- Pfizer Global Biotherapeutic Technologies, Cambridge, MA, USA
| | - Y Xie
- Pfizer Global Biotherapeutic Technologies, Cambridge, MA, USA
| | - M Pausch
- Pfizer Neuroscience Research Unit, Princeton, NJ, USA
| | - A Hayashi-Takagi
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - K Ishizuka
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - S Seshadri
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - B Bates
- Pfizer Global Biotherapeutic Technologies, Cambridge, MA, USA
| | - K Kariya
- Department of Medical Biochemistry, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - A Sawa
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - RJ Weinberg
- Department of Cell and Developmental Biology, University of North Carolina, Chapel Hill, NC, USA
| | - SJ Moss
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| | - MD Houslay
- Neuroscience and Molecular Pharmacology, Faculty of Biomedical and Life Sciences, University of Glasgow, Scotland, UK
| | - Z Yan
- Department of Physiology and Biophysics, University at Buffalo, SUNY, Buffalo, NY, USA
| | - NJ Brandon
- Pfizer Neuroscience Research Unit, Princeton, NJ, USA
- Pfizer Neuroscience Research Unit, Pfizer, Groton, CT, USA
| |
Collapse
|
263
|
A Novel Microduplication in the Neurodevelopmental Gene SRGAP3 That Segregates with Psychotic Illness in the Family of a COS Proband. Case Rep Genet 2011; 2011:585893. [PMID: 23074677 PMCID: PMC3447216 DOI: 10.1155/2011/585893] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Accepted: 07/14/2011] [Indexed: 01/27/2023] Open
Abstract
Schizophrenia is a debilitating mental disorder affecting approximately 1% of the world's population. Childhood onset schizophrenia (COS), defined as onset before age 13, is a rare and severe form of the illness that may have more salient genetic influence. We identified a ~134 kb duplication spanning exons 2–4 of the Slit-Robo GTPase-activating protein 3 (SRGAP3) gene on chromosome 3p25.3 that tracks with psychotic illness in the family of a COS proband. Cloning and sequencing of the duplication junction confirmed that the duplication is tandem, and analysis of the resulting mRNA transcript suggests that the duplication would result in a frame shift mutation. This is the first family report of a SRGAP3 copy number variant (CNV) in schizophrenia. Considering that SRGAP3 is important in neural development, we conclude that this SRGAP3 duplication may be an important factor contributing to the psychotic phenotype in this family.
Collapse
|
264
|
Kuroda K, Yamada S, Tanaka M, Iizuka M, Yano H, Mori D, Tsuboi D, Nishioka T, Namba T, Iizuka Y, Kubota S, Nagai T, Ibi D, Wang R, Enomoto A, Isotani-Sakakibara M, Asai N, Kimura K, Kiyonari H, Abe T, Mizoguchi A, Sokabe M, Takahashi M, Yamada K, Kaibuchi K. Behavioral alterations associated with targeted disruption of exons 2 and 3 of the Disc1 gene in the mouse. Hum Mol Genet 2011; 20:4666-83. [PMID: 21903668 DOI: 10.1093/hmg/ddr400] [Citation(s) in RCA: 118] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Disrupted-In-Schizophrenia 1 (DISC1) is a promising candidate gene for susceptibility to psychiatric disorders, including schizophrenia. DISC1 appears to be involved in neurogenesis, neuronal migration, axon/dendrite formation and synapse formation; during these processes, DISC1 acts as a scaffold protein by interacting with various partners. However, the lack of Disc1 knockout mice and a well-characterized antibody to DISC1 has made it difficult to determine the exact role of DISC1 in vivo. In this study, we generated mice lacking exons 2 and 3 of the Disc1 gene and prepared specific antibodies to the N- and C-termini of DISC1. The Disc1 mutant mice are viable and fertile, and no gross phenotypes, such as disorganization of the brain's cytoarchitecture, were observed. Western blot analysis revealed that the DISC1-specific antibodies recognize a protein with an apparent molecular mass of ~100 kDa in brain extracts from wild-type mice but not in brain extracts from DISC1 mutant mice. Immunochemical studies demonstrated that DISC1 is mainly localized to the vicinity of the Golgi apparatus in hippocampal neurons and astrocytes. A deficiency of full-length Disc1 induced a threshold shift in the induction of long-term potentiation in the dentate gyrus. The Disc1 mutant mice displayed abnormal emotional behavior as assessed by the elevated plus-maze and cliff-avoidance tests, thereby suggesting that a deficiency of full-length DISC1 may result in lower anxiety and/or higher impulsivity. Based on these results, we suggest that full-length Disc1-deficient mice and DISC1-specific antibodies are powerful tools for dissecting the pathophysiological functions of DISC1.
Collapse
Affiliation(s)
- Keisuke Kuroda
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
265
|
Raznahan A, Lee Y, Long R, Greenstein D, Clasen L, Addington A, Rapoport JL, Giedd JN. Common functional polymorphisms of DISC1 and cortical maturation in typically developing children and adolescents. Mol Psychiatry 2011; 16:917-26. [PMID: 20628343 PMCID: PMC3162084 DOI: 10.1038/mp.2010.72] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2009] [Revised: 04/10/2010] [Accepted: 04/12/2010] [Indexed: 12/17/2022]
Abstract
Disrupted-in-schizophrenia-1 (DISC1), contains two common non-synonymous single-nucleotide polymorphisms (SNPs)--Leu607Phe and Ser704Cys--that modulate (i) facets of DISC1 molecular functioning important for cortical development, (ii) fronto-temporal cortical anatomy in adults and (iii) risk for diverse psychiatric phenotypes that often emerge during childhood and adolescence, and are associated with altered fronto-temporal cortical development. It remains unknown, however, if Leu607Phe and Ser704Cys influence cortical maturation before adulthood, and whether each SNP shows unique or overlapping effects. Therefore, we related genotype at Leu607Phe and Ser704Cys to cortical thickness (CT) in 255 typically developing individuals aged 9-22 years on whom 598 magnetic resonance imaging brain scans had been acquired longitudinally. Rate of cortical thinning varied with DISC1 genotype. Specifically, the rate of cortical thinning was attenuated in Phe-carrier compared with Leu-homozygous groups (in bilateral superior frontal and left angular gyri) and accelerated in Ser-homozygous compared with Cys-carrier groups (in left anterior cingulate and temporal cortices). Both SNPs additively predicted fixed differences in right lateral temporal CT, which were maximal between Phe-carrier/Ser-homozygous (thinnest) vs Leu-homozygous/Cys-carrier (thickest) groups. Leu607Phe and Ser704Cys genotype interacted to predict the rate of cortical thinning in right orbitofrontal, middle temporal and superior parietal cortices, wherein a significantly reduced rate of CT loss was observed in Phe-carrier/Cys-carrier participants only. Our findings argue for further examination of Leu607Phe and Ser704Cys interactions at a molecular level, and suggest that these SNPs might operate (in concert with other genetic and environmental factors) to shape risk for diverse phenotypes by impacting on the early maturation of fronto-temporal cortices.
Collapse
Affiliation(s)
- A Raznahan
- Child Psychiatry Branch, National Institute of Mental Health, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | | | |
Collapse
|
266
|
Sanchez-Pulido L, Ponting CP. Structure and evolutionary history of DISC1. Hum Mol Genet 2011; 20:R175-81. [DOI: 10.1093/hmg/ddr374] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
267
|
Moens LN, De Rijk P, Reumers J, Van Den Bossche MJA, Glassee W, De Zutter S, Lenaerts AS, Nordin A, Nilsson LG, Medina Castello I, Norrback KF, Goossens D, Van Steen K, Adolfsson R, Del-Favero J. Sequencing of DISC1 pathway genes reveals increased burden of rare missense variants in schizophrenia patients from a northern Swedish population. PLoS One 2011; 6:e23450. [PMID: 21853134 PMCID: PMC3154939 DOI: 10.1371/journal.pone.0023450] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2011] [Accepted: 07/18/2011] [Indexed: 01/24/2023] Open
Abstract
In recent years, DISC1 has emerged as one of the most credible and best supported candidate genes for schizophrenia and related neuropsychiatric disorders. Furthermore, increasing evidence--both genetic and functional--indicates that many of its protein interaction partners are also involved in the development of these diseases. In this study, we applied a pooled sample 454 sequencing strategy, to explore the contribution of genetic variation in DISC1 and 10 of its interaction partners (ATF5, Grb2, FEZ1, LIS-1, PDE4B, NDE1, NDEL1, TRAF3IP1, YWHAE, and ZNF365) to schizophrenia susceptibility in an isolated northern Swedish population. Mutation burden analysis of the identified variants in a population of 486 SZ patients and 514 control individuals, revealed that non-synonymous rare variants with a MAF<0.01 were significantly more present in patients compared to controls (8.64% versus 4.7%, P = 0.018), providing further evidence for the involvement of DISC1 and some of its interaction partners in psychiatric disorders. This increased burden of rare missense variants was even more striking in a subgroup of early onset patients (12.9% versus 4.7%, P = 0.0004), highlighting the importance of studying subgroups of patients and identifying endophenotypes. Upon investigation of the potential functional effects associated with the identified missense variants, we found that ∼90% of these variants reside in intrinsically disordered protein regions. The observed increase in mutation burden in patients provides further support for the role of the DISC1 pathway in schizophrenia. Furthermore, this study presents the first evidence supporting the involvement of mutations within intrinsically disordered protein regions in the pathogenesis of psychiatric disorders. As many important biological functions depend directly on the disordered state, alteration of this disorder in key pathways may represent an intriguing new disease mechanism for schizophrenia and related neuropsychiatric diseases. Further research into this unexplored domain will be required to elucidate the role of the identified variants in schizophrenia etiology.
Collapse
Affiliation(s)
- Lotte N. Moens
- Applied Molecular Genomics Group, Department of Molecular Genetics, Flanders Institute for Biotechnology (VIB), Flanders, Belgium
- University of Antwerp (UA), Antwerp, Belgium
| | - Peter De Rijk
- Applied Molecular Genomics Group, Department of Molecular Genetics, Flanders Institute for Biotechnology (VIB), Flanders, Belgium
- University of Antwerp (UA), Antwerp, Belgium
| | - Joke Reumers
- SWITCH Laboratory, Flanders Institute for Biotechnology (VIB), Flanders, Belgium
- Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Maarten J. A. Van Den Bossche
- Applied Molecular Genomics Group, Department of Molecular Genetics, Flanders Institute for Biotechnology (VIB), Flanders, Belgium
- University of Antwerp (UA), Antwerp, Belgium
| | - Wim Glassee
- Applied Molecular Genomics Group, Department of Molecular Genetics, Flanders Institute for Biotechnology (VIB), Flanders, Belgium
- University of Antwerp (UA), Antwerp, Belgium
| | - Sonia De Zutter
- Applied Molecular Genomics Group, Department of Molecular Genetics, Flanders Institute for Biotechnology (VIB), Flanders, Belgium
- University of Antwerp (UA), Antwerp, Belgium
| | - An-Sofie Lenaerts
- Applied Molecular Genomics Group, Department of Molecular Genetics, Flanders Institute for Biotechnology (VIB), Flanders, Belgium
- University of Antwerp (UA), Antwerp, Belgium
| | - Annelie Nordin
- Division of Psychiatry, Department of Clinical Sciences, Umeå University, Umeå, Sweden
| | | | - Ignacio Medina Castello
- Functional Genomics Unit, Bioinformatics and Genomics Department, Prince Felipe Research Centre (CIPF), Valencia, Spain
| | - Karl-Fredrik Norrback
- Division of Psychiatry, Department of Clinical Sciences, Umeå University, Umeå, Sweden
| | - Dirk Goossens
- Applied Molecular Genomics Group, Department of Molecular Genetics, Flanders Institute for Biotechnology (VIB), Flanders, Belgium
- University of Antwerp (UA), Antwerp, Belgium
| | - Kristel Van Steen
- Systems and Modeling Unit, Montefiore Institute/GIGA, University of Liège, Liège, Belgium
| | - Rolf Adolfsson
- Division of Psychiatry, Department of Clinical Sciences, Umeå University, Umeå, Sweden
| | - Jurgen Del-Favero
- Applied Molecular Genomics Group, Department of Molecular Genetics, Flanders Institute for Biotechnology (VIB), Flanders, Belgium
- University of Antwerp (UA), Antwerp, Belgium
| |
Collapse
|
268
|
Katsel P, Tan W, Abazyan B, Davis KL, Ross C, Pletnikov MV, Haroutunian V. Expression of mutant human DISC1 in mice supports abnormalities in differentiation of oligodendrocytes. Schizophr Res 2011; 130:238-49. [PMID: 21605958 PMCID: PMC3139741 DOI: 10.1016/j.schres.2011.04.021] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2011] [Revised: 04/18/2011] [Accepted: 04/20/2011] [Indexed: 01/15/2023]
Abstract
Abnormalities in oligodendrocyte (OLG) differentiation and OLG gene expression deficit have been described in schizophrenia (SZ). Recent studies revealed a critical requirement for Disrupted-in-Schizophrenia 1 (DISC1) in neural development. Transgenic mice with forebrain restricted expression of mutant human DISC1 (ΔhDISC1) are characterized by neuroanatomical and behavioral abnormalities reminiscent of some features of SZ. We sought to determine whether the expression of ΔhDISC1 may influence the development of OLGs in this mouse model. OLG- and cell cycle-associated gene and protein expression were characterized in the forebrain of ΔhDISC1 mice during different stages of neurodevelopment (E15 and P1 days) and in adulthood. The results suggest that the expression of ΔhDISC1 exerts a significant influence on oligodendrocyte differentiation and function, evidenced by premature OLG differentiation and increased proliferation of their progenitors. Additional findings showed that neuregulin 1 and its receptors may be contributing factors to the observed upregulation of OLG genes. Thus, OLG function may be perturbed by mutant hDISC1 in a model system that provides new avenues for studying aspects of the pathogenesis of SZ.
Collapse
Affiliation(s)
- Pavel Katsel
- Department of Psychiatry, The Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029-6575, USA.
| | | | | | | | | | | | | |
Collapse
|
269
|
Abstract
Schizophrenia is a common mental illness resulting from a complex interplay of genetic and environmental risk factors. Establishing its primary molecular and cellular aetiopathologies has proved difficult. However, this is a vital step towards the rational development of useful disease biomarkers and new therapeutic strategies. The advent and large-scale application of genomic, transcriptomic, proteomic and metabolomic technologies are generating data sets required to achieve this goal. This discovery phase, typified by its objective and hypothesis-free approach, is described in the first part of the review. The accumulating biological information, when viewed as a whole, reveals a number of biological process and subcellular locations that contribute to schizophrenia causation. The data also show that each technique targets different aspects of central nervous system function in the disease state. In the second part of the review, key schizophrenia candidate genes are discussed more fully. Two higher-order processes - adult neurogenesis and inflammation - that appear to have pathological relevance are also described in detail. Finally, three areas where progress would have a large impact on schizophrenia biology are discussed: deducing the causes of schizophrenia in the individual, explaining the phenomenon of cross-disorder risk factors, and distinguishing causative disease factors from those that are reactive or compensatory.
Collapse
|
270
|
Kvajo M, McKellar H, Gogos JA. Avoiding mouse traps in schizophrenia genetics: lessons and promises from current and emerging mouse models. Neuroscience 2011; 211:136-64. [PMID: 21821099 DOI: 10.1016/j.neuroscience.2011.07.051] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2011] [Revised: 07/15/2011] [Accepted: 07/19/2011] [Indexed: 01/31/2023]
Abstract
Schizophrenia is one of the most common psychiatric disorders, but despite progress in identifying the genetic factors implicated in its development, the mechanisms underlying its etiology and pathogenesis remain poorly understood. Development of mouse models is critical for expanding our understanding of the causes of schizophrenia. However, translation of disease pathology into mouse models has proven to be challenging, primarily due to the complex genetic architecture of schizophrenia and the difficulties in the re-creation of susceptibility alleles in the mouse genome. In this review we highlight current research on models of major susceptibility loci and the information accrued from their analysis. We describe and compare the different approaches that are necessitated by diverse susceptibility alleles, and discuss their advantages and drawbacks. Finally, we discuss emerging mouse models, such as second-generation pathophysiology models based on innovative approaches that are facilitated by the information gathered from the current genetic mouse models.
Collapse
Affiliation(s)
- M Kvajo
- Department of Physiology and Cellular Biophysics, College of Physicians & Surgeons, Columbia University Medical Center, 630 West 168th Street, New York, NY 10032, USA
| | | | | |
Collapse
|
271
|
Verhoeven WM, Tuinier S, van der Burgt I. Top-down or bottom-up: Contrasting perspectives on psychiatric diagnoses. Biologics 2011; 2:409-17. [PMID: 19707372 PMCID: PMC2721407 DOI: 10.2147/btt.s3053] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Clinical psychiatry is confronted with the expanding knowledge of medical genetics. Most of the research into the genetic underpinnings of major mental disorders as described in the categorical taxonomies, however, did reveal linkage with a variety of chromosomes. This heterogeneity of results is most probably due to the assumption that the nosological categories as used in these studies are disease entities with clear boundaries. If the reverse way of looking, the so-called bottom-up approach, is applied, it becomes clear that genetic abnormalities are in most cases not associated with a single psychiatric disorder but with a certain probability to develop a variety of aspecific psychiatric symptoms. The adequacy of the categorical taxonomy, the so-called top-down approach, seems to be inversely related to the amount of empirical etiological data. This is illustrated by four rather prevalent genetic syndromes, fragile X syndrome, Prader-Willi syndrome, 22q11 deletion syndrome, and Noonan syndrome, as well as by some cases with rare chromosomal abnormalities. From these examples, it becomes clear that psychotic symptoms as well as mood, anxiety, and autistic features can be found in a great variety of different genetic syndromes. A psychiatric phenotype exists, but comprises, apart from the chance to present several psychiatric symptoms, all elements from developmental, neurocognitive, and physical characteristics.
Collapse
|
272
|
Wang Q, Brandon NJ. Regulation of the cytoskeleton by Disrupted-in-schizophrenia 1 (DISC1). Mol Cell Neurosci 2011; 48:359-64. [PMID: 21757008 DOI: 10.1016/j.mcn.2011.06.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2011] [Revised: 05/27/2011] [Accepted: 06/02/2011] [Indexed: 12/21/2022] Open
Abstract
Disrupted in schizophrenia 1 (DISC1) is one of the strongest supported risk genes for psychiatric disorders, such as schizophrenia, major depression, bipolar disorder, and autism. Intensive study over the past 11 years, since the gene was cloned, has tried to understand at the molecular and cellular levels how mutations in DISC1 contribute to these diseases. The DISC1 protein has been reported to be localized to cytoskeleton-rich regions in cells, including the centrosome, base of primary cilia, axon and dendritic shafts and spines. Here we review the functions of DISC1 which are relevant for cytoskeletal regulation and its crucial roles during normal brain development and in adult brain function. This article is part of a Special Issue entitled Neuronal Function.
Collapse
Affiliation(s)
- Qi Wang
- Pfizer Neuroscience Research Unit, Eastern Point Road, Groton, CT 06340, USA.
| | | |
Collapse
|
273
|
Green EK, Grozeva D, Sims R, Raybould R, Forty L, Gordon-Smith K, Russell E, St Clair D, Young AH, Ferrier IN, Kirov G, Jones I, Jones L, Owen MJ, O'Donovan MC, Craddock N. DISC1 exon 11 rare variants found more commonly in schizoaffective spectrum cases than controls. Am J Med Genet B Neuropsychiatr Genet 2011; 156B:490-2. [PMID: 21445958 DOI: 10.1002/ajmg.b.31187] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Accepted: 03/03/2011] [Indexed: 12/12/2022]
Abstract
We previously performed a linkage study using families identified through probands meeting criteria for DSM-IV schizoaffective disorder, bipolar type (SABP) and observed a genome-wide significant signal (LOD = 3.54) at chromosome 1q42 close to DISC1. An initial sequencing study of DISC1 using 14 unrelated DSM-IV SABP samples from the linkage study identified 2 non-synonymous coding SNPs in exon 11 in 2 separate individuals. Here we provide evidence of additional rare coding SNPs within exon 11. In sequencing exon 11 in 506 cases and 1,211 controls for variants that occurred only once, 4 additional rare variants were found in cases (P-value = 0.008, Fisher's exact trend test).
Collapse
Affiliation(s)
- E K Green
- MRC Centre for Neuropsychiatric Genetics and Genomics, Department of Psychological Medicine and Neurology, School of Medicine, Cardiff University, Heath Park, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
274
|
Zheng F, Wang L, Jia M, Yue W, Ruan Y, Lu T, Liu J, Li J, Zhang D. Evidence for association between Disrupted-in-Schizophrenia 1 (DISC1) gene polymorphisms and autism in Chinese Han population: a family-based association study. BEHAVIORAL AND BRAIN FUNCTIONS : BBF 2011; 7:14. [PMID: 21569632 PMCID: PMC3113723 DOI: 10.1186/1744-9081-7-14] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2010] [Accepted: 05/15/2011] [Indexed: 12/27/2022]
Abstract
BACKGROUND Disrupted-in-Schizophrenia 1 (DISC1) gene is one of the most promising candidate genes for major mental disorders. In a previous study, a Finnish group demonstrated that DISC1 polymorphisms were associated with autism and Asperger syndrome. However, the results were not replicated in Korean population. To determine whether DISC1 is associated with autism in Chinese Han population, we performed a family-based association study between DISC1 polymorphisms and autism. METHODS We genotyped seven tag single nucleotide polymorphisms (SNPs) in DISC1, spanning 338 kb, in 367 autism trios (singleton and their biological parents) including 1,101 individuals. Single SNP association and haplotype association analysis were performed using the family-based association test (FBAT) and Haploview software. RESULTS We found three SNPs showed significant associations with autism (rs4366301: G>C, Z=2.872, p=0.004; rs11585959: T>C, Z=2.199, p=0.028; rs6668845: A>G, Z=2.326, p=0.02). After the Bonferroni correction, SNP rs4366301, which located in the first intron of DISC1, remained significant. When haplotype were constructed with two-markers, three haplotypes displayed significant association with autism. These results were still significant after using the permutation method to obtain empirical p values. CONCLUSIONS Our study provided evidence that the DISC1 may be the susceptibility gene of autism. It suggested DISC1 might play a role in the pathogenesis of autism.
Collapse
Affiliation(s)
- Fanfan Zheng
- Key Laboratory for Mental Health, Ministry of Health, Beijing, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
275
|
Nicodemus KK, Callicott JH, Higier RG, Luna A, Nixon DC, Lipska BK, Vakkalanka R, Giegling I, Rujescu D, St Clair D, Muglia P, Shugart YY, Weinberger DR. Evidence of statistical epistasis between DISC1, CIT and NDEL1 impacting risk for schizophrenia: biological validation with functional neuroimaging. Hum Genet 2011; 127:441-52. [PMID: 20084519 DOI: 10.1007/s00439-009-0782-y] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2009] [Accepted: 12/24/2009] [Indexed: 02/05/2023]
Abstract
The etiology of schizophrenia likely involves genetic interactions. DISC1, a promising candidate susceptibility gene, encodes a protein which interacts with many other proteins, including CIT, NDEL1, NDE1, FEZ1 and PAFAH1B1, some of which also have been associated with psychosis. We tested for epistasis between these genes in a schizophrenia case-control study using machine learning algorithms (MLAs: random forest, generalized boosted regression andMonteCarlo logic regression). Convergence of MLAs revealed a subset of seven SNPs that were subjected to 2-SNP interaction modeling using likelihood ratio tests for nested unconditional logistic regression models. Of the 7C2 = 21 interactions, four were significant at the α = 0.05 level: DISC1 rs1411771-CIT rs10744743 OR = 3.07 (1.37, 6.98) p = 0.007; CIT rs3847960-CIT rs203332 OR = 2.90 (1.45, 5.79) p = 0.003; CIT rs3847960-CIT rs440299 OR = 2.16 (1.04, 4.46) p = 0.038; one survived Bonferroni correction (NDEL1 rs4791707-CIT rs10744743 OR = 4.44 (2.22, 8.88) p = 0.00013). Three of four interactions were validated via functional magnetic resonance imaging (fMRI) in an independent sample of healthy controls; risk associated alleles at both SNPs predicted prefrontal cortical inefficiency during the N-back task, a schizophrenia-linked intermediate biological phenotype: rs3847960-rs440299; rs1411771-rs10744743, rs4791707-rs10744743 (SPM5 p < 0.05, corrected), although we were unable to statistically replicate the interactions in other clinical samples. Interestingly, the CIT SNPs are proximal to exons that encode theDISC1 interaction domain. In addition, the 3' UTR DISC1 rs1411771 is predicted to be an exonic splicing enhancer and the NDEL1 SNP is ~3,000 bp from the exon encoding the region of NDEL1 that interacts with the DISC1 protein, giving a plausible biological basis for epistasis signals validated by fMRI.
Collapse
Affiliation(s)
- Kristin K Nicodemus
- Genes, Cognition and Psychosis Program, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
276
|
Tomita K, Kubo KI, Ishii K, Nakajima K. Disrupted-in-Schizophrenia-1 (Disc1) is necessary for migration of the pyramidal neurons during mouse hippocampal development. Hum Mol Genet 2011; 20:2834-45. [PMID: 21540240 DOI: 10.1093/hmg/ddr194] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
The hippocampus has a highly ordered structure and is composed of distinct layers. Neuronal migration is an essential part of the process of the layer formation because neurons are primarily generated near the ventricle and must migrate to arrive at their final locations during brain development. Impairment of brain development is thought to underlie the etiology of psychiatric disorders. Consistent with this idea, many genetic risk factors for psychiatric disorders play critical roles during brain development. As one example, Disrupted-in-Schizophrenia-1 (DISC1) is a genetic risk factor for major psychiatric disorders and plays various roles during neurodevelopment. To examine the role of Disc1 in the hippocampal development, we suppressed expression of Disc1 in the CA1 region of the developing mouse hippocampus by using the RNA interference (RNAi) technology and an in utero electroporation system. Disc1 suppression was found to impair migration of the CA1 pyramidal neurons. This effect was especially apparent while the majority of the transfected neurons were passing through the stratum pyramidale of the developing hippocampus. The migration of neurons was restored by expression of an RNAi-resistant wild-type mouse Disc1, indicating that the migration defect was caused by specific suppression of Disc1. In the mature hippocampus, the migration defect resulted in malposition and disarray of the pyramidal neurons. These findings indicate that Disc1 is required for migration and layer formation by the CA1 pyramidal neurons during hippocampal development.
Collapse
Affiliation(s)
- Kenji Tomita
- Department of Anatomy, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan
| | | | | | | |
Collapse
|
277
|
Chakirova G, Whalley HC, Thomson PA, Hennah W, Moorhead TWJ, Welch KA, Giles S, Hall J, Johnstone EC, Lawrie SM, Porteous DJ, Brown VJ, McIntosh AM. The effects of DISC1 risk variants on brain activation in controls, patients with bipolar disorder and patients with schizophrenia. Psychiatry Res 2011; 192:20-8. [PMID: 21376542 DOI: 10.1016/j.pscychresns.2011.01.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2010] [Revised: 11/12/2010] [Accepted: 01/13/2011] [Indexed: 01/15/2023]
Abstract
Three risk variants (rs1538979, rs821577, and rs821633) in the Disrupted-in-Schizophrenia-1 (DISC1) gene have previously been associated with both schizophrenia and bipolar disorder in a recent collaborative analysis of European cohorts. In this study we examined the effects of these risk variants on brain activation during functional magnetic resonance imaging (fMRI) of the Hayling Sentence Completion Task (HSCT) in healthy volunteers (n=33), patients with schizophrenia (n=20) and patients with bipolar disorder (n=36). In the healthy controls the risk associated allele carriers of SNPs rs1538979 and rs821633 demonstrated decreased activation of the cuneus. Moreover, there was an effect of SNP rs1538979 in the pre/postcentral gyrus with decreased activation in healthy controls and increased activation in patients with schizophrenia. In the bipolar group there was decreased activation in the risk carriers of SNP rs821633 in the inferior parietal lobule and left cingulate cortex. Clusters in the precentral gyrus, left middle temporal gyrus and left cerebellum were found to be significant on examining the group × genotype interactions. These findings may provide a better understanding of the neural effects of DISC1 variants and on the pathophysiology of schizophrenia and bipolar disorder.
Collapse
|
278
|
Integration-free induced pluripotent stem cells derived from schizophrenia patients with a DISC1 mutation. Mol Psychiatry 2011; 16:358-60. [PMID: 21339753 PMCID: PMC4005725 DOI: 10.1038/mp.2011.13] [Citation(s) in RCA: 147] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
279
|
Association study between Disrupted-in-Schizophrenia-1 (DISC1) and Japanese patients with treatment-resistant schizophrenia (TRS). Prog Neuropsychopharmacol Biol Psychiatry 2011; 35:636-9. [PMID: 21256178 DOI: 10.1016/j.pnpbp.2011.01.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2010] [Revised: 01/10/2011] [Accepted: 01/11/2011] [Indexed: 10/18/2022]
Abstract
Treating the 20-30% of patients with schizophrenia whose symptoms are resistant to antipsychotic treatment, a condition known as treatment-resistant schizophrenia (TRS), can be problematic. Recently, an association between Disrupted-in-Schizophrenia-1 (DISC1), a candidate susceptibility gene for schizophrenia, and TRS was reported. Associations between three missense SNPs, rs3738401 (Q264R), rs6675281 (L607F), and rs821616 (S704C) in DISC1, especially rs3738401, showed strong significance. Thus, the main aim of our current study was to examine if the reported possible functional polymorphisms in DISC1 were related to Japanese TRS. First, DISC1 was re-investigated in 485 Japanese patients with schizophrenia and 660 healthy controls with a case-control study using four candidate SNPs, rs751229, rs3738401, rs821597, and rs821616. DISC1 was not associated with schizophrenia in the Japanese population. Second, we investigated whether these SNPs contributed to TRS in 127 inpatients with schizophrenia (35 patients; TRS and 92 patients; non-TRS). The genotypic distributions of these four SNPs were not significantly different between TRS and non-TRS in either genotypic or recessive models of minor alleles. In addition, clinical variables, such as improvement in clinical symptoms, duration of hospitalization, and total antipsychotics dose amounts, were not different among the genotypes of these SNPs. Taken together, results showed that DISC1 had no apparent degree of association with Japanese patients with schizophrenia as a candidate susceptibility gene for disease per se or TRS.
Collapse
|
280
|
Penzes P, Cahill ME, Jones KA, VanLeeuwen JE, Woolfrey KM. Dendritic spine pathology in neuropsychiatric disorders. Nat Neurosci 2011; 14:285-93. [PMID: 21346746 PMCID: PMC3530413 DOI: 10.1038/nn.2741] [Citation(s) in RCA: 1168] [Impact Index Per Article: 83.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Substantial progress has been made toward understanding the genetic architecture, cellular substrates, brain circuits and endophenotypic profiles of neuropsychiatric disorders, including autism spectrum disorders (ASD), schizophrenia and Alzheimer's disease. Recent evidence implicates spiny synapses as important substrates of pathogenesis in these disorders. Although synaptic perturbations are not the only alterations relevant for these diseases, understanding the molecular underpinnings of spine pathology may provide insight into their etiologies and may reveal new drug targets. Here we discuss recent neuropathological, genetic, molecular and animal model studies that implicate structural alterations at spiny synapses in the pathogenesis of major neurological disorders, focusing on ASD, schizophrenia and Alzheimer's disease as representatives of these categories across different ages of onset. We stress the importance of reverse translation, collaborative and multidisciplinary approaches, and the study of the spatio-temporal roles of disease molecules in the context of synaptic regulatory pathways and neuronal circuits that underlie disease endophenotypes.
Collapse
Affiliation(s)
- Peter Penzes
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
| | | | | | | | | |
Collapse
|
281
|
Liu J, Li J, Li T, Wang T, Li Y, Zeng Z, Li Z, Chen P, Hu Z, Zheng L, Ji J, Lin H, Feng G, Shi Y. CTLA-4 confers a risk of recurrent schizophrenia, major depressive disorder and bipolar disorder in the Chinese Han population. Brain Behav Immun 2011; 25:429-33. [PMID: 21040781 DOI: 10.1016/j.bbi.2010.10.024] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2010] [Revised: 10/21/2010] [Accepted: 10/23/2010] [Indexed: 10/18/2022] Open
Abstract
Previous studies have reported that the cytotoxic T lymphocyte antigen-4 (CTLA-4) gene, which is related to immunological function such as T-cell regulation, is associated with psychiatric disorders. In this study, we studied the relationship between CTLA-4 and three major psychiatric disorders, schizophrenia, major depressive disorder and bipolar disorder in the Chinese Han population. We recruited 1140 schizophrenia patients, 1140 major depressive disorder patients, 1140 bipolar disorder patients, and 1140 normal controls to examine the risk conferred by 6 tag SNPs (rs231777, rs231775, rs231779, rs3087243, rs5742909, rs16840252) in the CTLA-4 gene. We found that rs231779 conferred a risk for schizophrenia (P(allele)=0.0003, P(genotype)=0.0016), major depressive disorder (P(allele)=0.0006, P(genotype)=0.0026) and bipolar disorder (P(allele)=0.0004, P(genotype)=0.0018). In addition, rs231777 and rs16840252 had a significant association with schizophrenia (rs231777: P(allele)=0.0201, rs16840252: P(allele)=0.0081, P(genotype)=0.0117), and rs231777 had significant association with bipolar disorder (rs231777: P(allele)=0.0199). However, after 10,000 permutations, only rs231779 remained significant (schizophrenia: P(allele)=0.0010, P(genotype)=0.0145, major depressive disorder: P(allele)=0.0010, P(genotype)=0.0201, bipolar disorder: P(allele)=0.0008, P(genotype)=0.0125). Our results suggest that shared common risk factors for schizophrenia, major depressive disorder and bipolar disorder exist in the CTLA-4 gene in the Chinese Han population.
Collapse
Affiliation(s)
- Jie Liu
- Bio-X Center, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, PR China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
282
|
Myers RA, Casals F, Gauthier J, Hamdan FF, Keebler J, Boyko AR, Bustamante CD, Piton AM, Spiegelman D, Henrion E, Zilversmit M, Hussin J, Quinlan J, Yang Y, Lafrenière RG, Griffing AR, Stone EA, Rouleau GA, Awadalla P. A population genetic approach to mapping neurological disorder genes using deep resequencing. PLoS Genet 2011; 7:e1001318. [PMID: 21383861 PMCID: PMC3044677 DOI: 10.1371/journal.pgen.1001318] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2010] [Accepted: 01/24/2011] [Indexed: 01/15/2023] Open
Abstract
Deep resequencing of functional regions in human genomes is key to identifying potentially causal rare variants for complex disorders. Here, we present the results from a large-sample resequencing (n = 285 patients) study of candidate genes coupled with population genetics and statistical methods to identify rare variants associated with Autism Spectrum Disorder and Schizophrenia. Three genes, MAP1A, GRIN2B, and CACNA1F, were consistently identified by different methods as having significant excess of rare missense mutations in either one or both disease cohorts. In a broader context, we also found that the overall site frequency spectrum of variation in these cases is best explained by population models of both selection and complex demography rather than neutral models or models accounting for complex demography alone. Mutations in the three disease-associated genes explained much of the difference in the overall site frequency spectrum among the cases versus controls. This study demonstrates that genes associated with complex disorders can be mapped using resequencing and analytical methods with sample sizes far smaller than those required by genome-wide association studies. Additionally, our findings support the hypothesis that rare mutations account for a proportion of the phenotypic variance of these complex disorders. It is widely accepted that genetic factors play important roles in the etiology of neurological diseases. However, the nature of the underlying genetic variation remains unclear. Critical questions in the field of human genetics relate to the frequency and size effects of genetic variants associated with disease. For instance, the common disease–common variant model is based on the idea that sets of common variants explain a significant fraction of the variance found in common disease phenotypes. On the other hand, rare variants may have strong effects and therefore largely contribute to disease phenotypes. Due to their high penetrance and reduced fitness, such variants are maintained in the population at low frequencies, thus limiting their detection in genome-wide association studies. Here, we use a resequencing approach on a cohort of 285 Autism Spectrum Disorder and Schizophrenia patients and preformed several analyses, enhanced with population genetic approaches, to identify variants associated with both diseases. Our results demonstrate an excess of rare variants in these disease cohorts and identify genes with negative (deleterious) selection coefficients, suggesting an accumulation of variants of detrimental effects. Our results present further evidence for rare variants explaining a component of the genetic etiology of autism and schizophrenia.
Collapse
Affiliation(s)
- Rachel A. Myers
- Department of Pediatrics, University of Montreal, Montreal, Canada
- CHU Sainte-Justine Research Centre, University of Montreal, Montreal, Canada
- Bioinformatics Research Centre, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Ferran Casals
- Department of Pediatrics, University of Montreal, Montreal, Canada
| | - Julie Gauthier
- Centre of Excellence in Neuromics of Université de Montréal, Centre Hospitalier de l'Université de Montréal, Montreal, Canada
- Department of Medicine, Université of Montréal, Montreal, Canada
| | - Fadi F. Hamdan
- Centre of Excellence in Neuromics of Université de Montréal, Centre Hospitalier de l'Université de Montréal, Montreal, Canada
- Department of Medicine, Université of Montréal, Montreal, Canada
| | - Jon Keebler
- Department of Pediatrics, University of Montreal, Montreal, Canada
- CHU Sainte-Justine Research Centre, University of Montreal, Montreal, Canada
- Bioinformatics Research Centre, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Adam R. Boyko
- Department of Genetics, Stanford University School of Medicine, Stanford, California, United States of America
| | - Carlos D. Bustamante
- Department of Genetics, Stanford University School of Medicine, Stanford, California, United States of America
| | - Amelie M. Piton
- Centre of Excellence in Neuromics of Université de Montréal, Centre Hospitalier de l'Université de Montréal, Montreal, Canada
- Department of Medicine, Université of Montréal, Montreal, Canada
| | - Dan Spiegelman
- Centre of Excellence in Neuromics of Université de Montréal, Centre Hospitalier de l'Université de Montréal, Montreal, Canada
- Department of Medicine, Université of Montréal, Montreal, Canada
| | - Edouard Henrion
- Centre of Excellence in Neuromics of Université de Montréal, Centre Hospitalier de l'Université de Montréal, Montreal, Canada
- Department of Medicine, Université of Montréal, Montreal, Canada
| | | | - Julie Hussin
- Department of Pediatrics, University of Montreal, Montreal, Canada
| | - Jacklyn Quinlan
- Department of Pediatrics, University of Montreal, Montreal, Canada
| | - Yan Yang
- Centre of Excellence in Neuromics of Université de Montréal, Centre Hospitalier de l'Université de Montréal, Montreal, Canada
- Department of Medicine, Université of Montréal, Montreal, Canada
| | - Ronald G. Lafrenière
- Centre of Excellence in Neuromics of Université de Montréal, Centre Hospitalier de l'Université de Montréal, Montreal, Canada
- Department of Medicine, Université of Montréal, Montreal, Canada
| | - Alexander R. Griffing
- Bioinformatics Research Centre, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Eric A. Stone
- Bioinformatics Research Centre, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Guy A. Rouleau
- CHU Sainte-Justine Research Centre, University of Montreal, Montreal, Canada
- Centre of Excellence in Neuromics of Université de Montréal, Centre Hospitalier de l'Université de Montréal, Montreal, Canada
- Department of Medicine, Université of Montréal, Montreal, Canada
- * E-mail: (PA); (GAR)
| | - Philip Awadalla
- Department of Pediatrics, University of Montreal, Montreal, Canada
- CHU Sainte-Justine Research Centre, University of Montreal, Montreal, Canada
- Bioinformatics Research Centre, North Carolina State University, Raleigh, North Carolina, United States of America
- Centre of Excellence in Neuromics of Université de Montréal, Centre Hospitalier de l'Université de Montréal, Montreal, Canada
- Department of Medicine, Université of Montréal, Montreal, Canada
- * E-mail: (PA); (GAR)
| |
Collapse
|
283
|
Lawrie SM, Olabi B, Hall J, McIntosh AM. Do we have any solid evidence of clinical utility about the pathophysiology of schizophrenia? World Psychiatry 2011; 10:19-31. [PMID: 21379347 PMCID: PMC3048512 DOI: 10.1002/j.2051-5545.2011.tb00004.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
A diagnosis of schizophrenia, as in most of psychiatric practice, is made largely by eliciting symptoms with reference to subjective, albeit operationalized, criteria. This diagnosis then provides some rationale for management. Objective diagnostic and therapeutic tests are much more desirable, provided they are reliably measured and interpreted. Definite advances have been made in our understanding of schizophrenia in recent decades, but there has been little consideration of how this information could be used in clinical practice. We review here the potential utility of the strongest and best replicated risk factors for and manifestations of schizophrenia within clinical, epidemiological, cognitive, blood biomarker and neuroimaging domains. We place particular emphasis on the sensitivity, specificity and predictive power of pathophysiological indices for making a diagnosis, establishing an early diagnosis or predicting treatment response in schizophrenia. We conclude that a number of measures currently available have the potential to increase the rigour of clinical assessments in schizophrenia. We propose that the time has come to more fully evaluate these and other well replicated abnormalities as objective potential diagnostic and prognostic guides, and to steer future clinical, therapeutic and nosological research in this direction.
Collapse
Affiliation(s)
- Stephen M Lawrie
- Division of Psychiatry, School of Molecular and Clinical Medicine, Royal Edinburgh Hospital, Morningside, Edinburgh EH10 5HF, UK
| | | | | | | |
Collapse
|
284
|
Gondo Y, Murata T, Makino S, Fukumura R, Ishitsuka Y. Mouse mutagenesis and disease models for neuropsychiatric disorders. Curr Top Behav Neurosci 2011; 7:1-35. [PMID: 21298381 DOI: 10.1007/7854_2010_106] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
In this chapter, mutant mouse resources which have been developed by classical genetics as well as by modern large-scale mutagenesis projects are summarized. Various spontaneous and induced mouse mutations have been archived since the rediscovery of Mendel's genetics in 1900. Moreover, genome-wide, large-scale mutagenesis efforts have recently been expanding the available mutant mouse resources. Forward genetics projects using ENU mutagenesis in the mouse were started in the mid-1990s. The widespread adoption of reverse genetics, using knockouts and conditional mutagenesis based on gene-targeting technology, followed. ENU mutagenesis has now evolved to provide a further resource for reverse genetics, with multiple point mutations in a single gene and this new approach is described. Researchers now have various options to obtain mutant mice: point mutations, transgenic mouse strains, and constitutional or conditional knockout mice. The established mutant strains have already contributed to modeling human diseases by elucidating the underlying molecular mechanisms as well as by providing preclinical applications. Examples of mutant mice, focusing on neurological and behavioral models for human diseases, are reviewed. Human diseases caused by a single gene or a small number of major genes have been well modeled by corresponding mutant mice. Current evidence suggests that quantitative traits based on polygenes are likely to be associated with a range of psychiatric diseases, and these are now coming within the range of modeling by mouse mutagenesis.
Collapse
Affiliation(s)
- Yoichi Gondo
- Mutagenesis and Genomics Team, RIKEN BioResource Center, 3-1-1 Koyadai, Tsukuba, Ibaraki, 305-0074, Japan,
| | | | | | | | | |
Collapse
|
285
|
Sarnyai Z, Alsaif M, Bahn S, Ernst A, Guest PC, Hradetzky E, Kluge W, Stelzhammer V, Wesseling H. Behavioral and molecular biomarkers in translational animal models for neuropsychiatric disorders. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2011; 101:203-38. [PMID: 22050853 DOI: 10.1016/b978-0-12-387718-5.00008-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Modeling neuropsychiatric disorders in animals poses a significant challenge due to the subjective nature of diverse often overlapping symptoms, lack of objective biomarkers and diagnostics, and the rudimentary understanding of the pathophysiology. Successful translational research requires animal models that can inform about disease mechanisms and therapeutic targets. Here, we review behavioral and neurobiological findings from selected animal models, based on presumed etiology and risk factors, for schizophrenia, bipolar disorder, and major depressive disorder. We focus on the use of appropriate statistical tools and newly developed Research Domain Criteria (RDoC) to link biomarkers from animal models with the human disease. We argue that this approach will lead to development of only the most robust animal models for specific psychiatric disorders and may ultimately lead to better understanding of the pathophysiology and identification of novel biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Zoltán Sarnyai
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
286
|
Balu DT, Coyle JT. Neuroplasticity signaling pathways linked to the pathophysiology of schizophrenia. Neurosci Biobehav Rev 2011; 35:848-70. [PMID: 20951727 PMCID: PMC3005823 DOI: 10.1016/j.neubiorev.2010.10.005] [Citation(s) in RCA: 130] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2010] [Revised: 10/06/2010] [Accepted: 10/10/2010] [Indexed: 12/15/2022]
Abstract
Schizophrenia is a severe mental illness that afflicts nearly 1% of the world's population. One of the cardinal pathological features of schizophrenia is perturbation in synaptic connectivity. Although the etiology of schizophrenia is unknown, it appears to be a developmental disorder involving the interaction of a potentially large number of risk genes, with no one gene producing a strong effect except rare, highly penetrant copy number variants. The purpose of this review is to detail how putative schizophrenia risk genes (DISC-1, neuregulin/ErbB4, dysbindin, Akt1, BDNF, and the NMDA receptor) are involved in regulating neuroplasticity and how alterations in their expression may contribute to the disconnectivity observed in schizophrenia. Moreover, this review highlights how many of these risk genes converge to regulate common neurotransmitter systems and signaling pathways. Future studies aimed at elucidating the functions of these risk genes will provide new insights into the pathophysiology of schizophrenia and will likely lead to the nomination of novel therapeutic targets for restoring proper synaptic connectivity in the brain in schizophrenia and related disorders.
Collapse
Affiliation(s)
- Darrick T Balu
- Department of Psychiatry, Harvard Medical School, Belmont, MA, USA.
| | | |
Collapse
|
287
|
Ingason A, Rujescu D, Cichon S, Sigurdsson E, Sigmundsson T, Pietiläinen OPH, Buizer-Voskamp JE, Strengman E, Francks C, Muglia P, Gylfason A, Gustafsson O, Olason PI, Steinberg S, Hansen T, Jakobsen KD, Rasmussen HB, Giegling I, Möller HJ, Hartmann A, Crombie C, Fraser G, Walker N, Lonnqvist J, Suvisaari J, Tuulio-Henriksson A, Bramon E, Kiemeney LA, Franke B, Murray R, Vassos E, Toulopoulou T, Mühleisen TW, Tosato S, Ruggeri M, Djurovic S, Andreassen OA, Zhang Z, Werge T, Ophoff RA, GROUP Investigators, Rietschel M, Nöthen MM, Petursson H, Stefansson H, Peltonen L, Collier D, Stefansson K, St Clair DM. Copy number variations of chromosome 16p13.1 region associated with schizophrenia. Mol Psychiatry 2011; 16:17-25. [PMID: 19786961 PMCID: PMC3330746 DOI: 10.1038/mp.2009.101] [Citation(s) in RCA: 192] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2009] [Revised: 08/18/2009] [Accepted: 08/21/2009] [Indexed: 01/22/2023]
Abstract
Deletions and reciprocal duplications of the chromosome 16p13.1 region have recently been reported in several cases of autism and mental retardation (MR). As genomic copy number variants found in these two disorders may also associate with schizophrenia, we examined 4345 schizophrenia patients and 35,079 controls from 8 European populations for duplications and deletions at the 16p13.1 locus, using microarray data. We found a threefold excess of duplications and deletions in schizophrenia cases compared with controls, with duplications present in 0.30% of cases versus 0.09% of controls (P=0.007) and deletions in 0.12 % of cases and 0.04% of controls (P>0.05). The region can be divided into three intervals defined by flanking low copy repeats. Duplications spanning intervals I and II showed the most significant (P = 0.00010) association with schizophrenia. The age of onset in duplication and deletion carriers among cases ranged from 12 to 35 years, and the majority were males with a family history of psychiatric disorders. In a single Icelandic family, a duplication spanning intervals I and II was present in two cases of schizophrenia, and individual cases of alcoholism, attention deficit hyperactivity disorder and dyslexia. Candidate genes in the region include NTAN1 and NDE1. We conclude that duplications and perhaps also deletions of chromosome 16p13.1, previously reported to be associated with autism and MR, also confer risk of schizophrenia.
Collapse
Affiliation(s)
- A Ingason
- deCODE genetics, Reykjavík, Iceland
- Research Institute of Biological Psychiatry, Mental Health Centre Sct. Hans, Copenhagen University Hospital, Roskilde, Denmark
| | - D Rujescu
- Division of Molecular and Clinical Neurobiology, Department of Psychiatry, Ludwig-Maximilians-University and Genetics Research Centre GmbH, Munich, Germany
| | - S Cichon
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
- Institute of Human Genetics, University of Bonn, Bonn, Germany
| | - E Sigurdsson
- Department of Psychiatry, National University Hospital, Reykjavík, Iceland
| | - T Sigmundsson
- Department of Psychiatry, National University Hospital, Reykjavík, Iceland
| | - OPH Pietiläinen
- Department for Molecular Medicine, National Public Health Institute, Helsinki, Finland
| | - JE Buizer-Voskamp
- Department of Psychiatry, Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Medical Genetics and Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands
| | - E Strengman
- Department of Medical Genetics and Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands
| | - C Francks
- Medical Genetics, GlaxoSmithKline R&D, Verona, Italy
| | - P Muglia
- Medical Genetics, GlaxoSmithKline R&D, Verona, Italy
| | | | | | | | | | - T Hansen
- Research Institute of Biological Psychiatry, Mental Health Centre Sct. Hans, Copenhagen University Hospital, Roskilde, Denmark
| | - KD Jakobsen
- Research Institute of Biological Psychiatry, Mental Health Centre Sct. Hans, Copenhagen University Hospital, Roskilde, Denmark
| | - HB Rasmussen
- Research Institute of Biological Psychiatry, Mental Health Centre Sct. Hans, Copenhagen University Hospital, Roskilde, Denmark
| | - I Giegling
- Division of Molecular and Clinical Neurobiology, Department of Psychiatry, Ludwig-Maximilians-University and Genetics Research Centre GmbH, Munich, Germany
| | - H-J Möller
- Division of Molecular and Clinical Neurobiology, Department of Psychiatry, Ludwig-Maximilians-University and Genetics Research Centre GmbH, Munich, Germany
| | - A Hartmann
- Division of Molecular and Clinical Neurobiology, Department of Psychiatry, Ludwig-Maximilians-University and Genetics Research Centre GmbH, Munich, Germany
| | - C Crombie
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, Scotland
| | - G Fraser
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, Scotland
| | - N Walker
- Ravenscraig Hospital, Greenock, Scotland
| | - J Lonnqvist
- Department of Mental Health and Addiction, National Public Health Institute, Helsinki, Finland
| | - J Suvisaari
- Department of Mental Health and Addiction, National Public Health Institute, Helsinki, Finland
| | - A Tuulio-Henriksson
- Department of Mental Health and Addiction, National Public Health Institute, Helsinki, Finland
| | - E Bramon
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, King’s College, London, UK
| | - LA Kiemeney
- Department of Epidemiology & Biostatistics (133 EPIB)/Department of Urology (659 URO), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - B Franke
- Department of Human Genetics, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - R Murray
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, King’s College, London, UK
| | - E Vassos
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, King’s College, London, UK
| | - T Toulopoulou
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, King’s College, London, UK
| | - TW Mühleisen
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
| | - S Tosato
- Section of Psychiatry and Clinical Psychology, University of Verona, Verona, Italy
| | - M Ruggeri
- Section of Psychiatry and Clinical Psychology, University of Verona, Verona, Italy
| | - S Djurovic
- Institute of Psychiatry, University of Oslo, Oslo, Norway
- Departments of Medical Genetics and Psychiatry, Ulleval University Hospital, Oslo, Norway
| | - OA Andreassen
- Institute of Psychiatry, University of Oslo, Oslo, Norway
- Departments of Medical Genetics and Psychiatry, Ulleval University Hospital, Oslo, Norway
| | - Z Zhang
- Department of Statistics, UCLA, Los Angeles, CA, USA
| | - T Werge
- Research Institute of Biological Psychiatry, Mental Health Centre Sct. Hans, Copenhagen University Hospital, Roskilde, Denmark
| | - RA Ophoff
- Department of Medical Genetics and Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands
- UCLA Center for Neurobehavioral Genetics and Department of Human Genetics, Los Angeles, CA, USA
| | | | - M Rietschel
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health Mannheim, University of Heidelberg, Mannheim, Germany
| | - MM Nöthen
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
- Institute of Human Genetics, University of Bonn, Bonn, Germany
| | - H Petursson
- Department of Psychiatry, National University Hospital, Reykjavík, Iceland
| | | | - L Peltonen
- Department for Molecular Medicine, National Public Health Institute, Helsinki, Finland
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK
- The Broad Institute, Cambridge, MA, USA
| | - D Collier
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, King’s College, London, UK
| | | | - DM St Clair
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, Scotland
| |
Collapse
|
288
|
Johnstone M, Thomson PA, Hall J, McIntosh AM, Lawrie SM, Porteous DJ. DISC1 in schizophrenia: genetic mouse models and human genomic imaging. Schizophr Bull 2011; 37:14-20. [PMID: 21149852 PMCID: PMC3004186 DOI: 10.1093/schbul/sbq135] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Schizophrenia and related disorders have a major genetic component. Several large-scale studies have uncovered a number of possible candidate genes, but these have yet to be consistently replicated and their underlying biological function remains elusive. One exception is 'Disrupted in schizophrenia 1' (DISC1), a gene locus originally identified in a large Scottish family, showing a heavy burden of major mental illnesses associated with a balanced t(1;11)(q42.1;q14.3) chromosome translocation. Substantial genetic and biological research on DISC1 has been reported in the intervening 10 years: DISC1 is now recognized as a genetic risk factor for a spectrum of psychiatric disorders and DISC1 impacts on many aspects of central nervous system (CNS) function, including neurodevelopment, neurosignaling, and synaptic functioning. Evidence has emerged from genetic studies showing a relationship between DISC1 and quantitative traits, including working memory, cognitive aging, gray matter volume in the prefrontal cortex, and abnormalities in hippocampal structures and function. DISC1 interacts with numerous proteins also involved in neuronal migration, neurite outgrowth, cytoskeletal modulation, and signal transduction, some of which have been reported as independent genetic susceptibility factors for psychiatric morbidity. Here, we focus on the growing literature relating genetic variation in the DISC1 pathway to functional and structural studies of the brain in humans and in the mouse.
Collapse
Affiliation(s)
- Mandy Johnstone
- Department of Psychiatry, The Royal Edinburgh Hospital, Morningside Terrace, Edinburgh EH10 5HF, UK
- Medical Genetics Section, Institute of Genetics and Molecular Medicine, University of Edinburgh Molecular Medicine Centre, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK
| | - Pippa A. Thomson
- Medical Genetics Section, Institute of Genetics and Molecular Medicine, University of Edinburgh Molecular Medicine Centre, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK
| | - Jeremy Hall
- Department of Psychiatry, The Royal Edinburgh Hospital, Morningside Terrace, Edinburgh EH10 5HF, UK
- Medical Genetics Section, Institute of Genetics and Molecular Medicine, University of Edinburgh Molecular Medicine Centre, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK
| | - Andrew M. McIntosh
- Department of Psychiatry, The Royal Edinburgh Hospital, Morningside Terrace, Edinburgh EH10 5HF, UK
- Medical Genetics Section, Institute of Genetics and Molecular Medicine, University of Edinburgh Molecular Medicine Centre, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK
| | - Stephen M. Lawrie
- Department of Psychiatry, The Royal Edinburgh Hospital, Morningside Terrace, Edinburgh EH10 5HF, UK
| | - David J. Porteous
- Medical Genetics Section, Institute of Genetics and Molecular Medicine, University of Edinburgh Molecular Medicine Centre, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK
| |
Collapse
|
289
|
No Association of the YWHAE Gene with Schizophrenia, Major Depressive Disorder or Bipolar Disorder in the Han Chinese Population. Behav Genet 2010; 41:557-64. [DOI: 10.1007/s10519-010-9426-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2010] [Accepted: 11/30/2010] [Indexed: 11/27/2022]
|
290
|
Dwyer S, Williams H, Holmans P, Moskvina V, Craddock N, Owen MJ, O'Donovan MC. No evidence that rare coding variants in ZNF804A confer risk of schizophrenia. Am J Med Genet B Neuropsychiatr Genet 2010; 153B:1411-6. [PMID: 20862696 DOI: 10.1002/ajmg.b.31117] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2010] [Accepted: 07/27/2010] [Indexed: 01/27/2023]
Abstract
Strong evidence that rare variants of relatively high penetrance are involved in the etiology of schizophrenia is currently restricted to the data from studies investigating copy number variants and major structural re-arrangements in that disorder. Global tests of the hypothesis of the involvement of fairly high penetrance rare single nucleotide changes or small insertion deletion events await the genesis of data from large-scale sequencing studies, meanwhile, a pragmatic approach to trying to detect such alleles is to target sequencing efforts on genes for which there is compelling evidence from other sources for their involvement in this disorder. We have undertaken a study, which aimed to identify whether rare (frequency ∼0.001%) coding variants in the schizophrenia susceptibility gene ZNF804A are involved in this disorder. We screened the coding regions of the gene in 517 schizophrenic cases and 501 controls, and genotyped rare non-synonymous variants in a case-control sample powered to detect association to rare alleles with an effect size (odds ratio) of 5. No single rare variant was associated with schizophrenia, nor was the burden of rare, or even fairly common, non-synonymous variants. Our results do not support the hypothesis that moderately rare non-synonymous variants at the ZNF804A locus are involved in schizophrenia susceptibility. © 2010 Wiley-Liss, Inc.
Collapse
Affiliation(s)
- S Dwyer
- MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, UK
| | | | | | | | | | | | | |
Collapse
|
291
|
Tiwari AK, Zai CC, Müller DJ, Kennedy JL. Genetics in schizophrenia: where are we and what next? DIALOGUES IN CLINICAL NEUROSCIENCE 2010. [PMID: 20954426 PMCID: PMC3181975 DOI: 10.31887/dcns.2010.12.3/atiwari] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Understanding the genetic basis of schizophrenia continues to be major challenge. The research done during the last two decades has provided several candidate genes which unfortunately have not been consistently replicated across or within a population. The recent genome-wide association studies (GWAS) and copy number variation (CNV) studies have provided important evidence suggesting a role of both common and rare large CNVs in schizophrenia genesis. The burden of rare copy number variations appears to be increased in schizophrenia patients. A consistent observation among the GWAS studies is the association with schizophrenia of genetic markers in the major histocompatibility complex (6p22.1)-containing genes including NOTCH4 and histone protein loci. Molecular genetic studies are also demonstrating that there is more overlap between the susceptibility genes for schizophrenia and bipolar disorder than previously suspected. In this review we summarize the major findings of the past decade and suggest areas of future research.
Collapse
Affiliation(s)
- Arun K Tiwari
- Neurogenetics section, Neuroscience department, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | | | | | | |
Collapse
|
292
|
Evidence for rare and common genetic risk variants for schizophrenia at protein kinase C, alpha. Mol Psychiatry 2010; 15:1101-11. [PMID: 19786960 DOI: 10.1038/mp.2009.96] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
We earlier reported a genome-wide significant linkage to schizophrenia at chromosome 17 that was identified in a single pedigree (C702) consisting of six affected, male siblings with DSM-IV schizophrenia and prominent mood symptoms. In this study, we adopted several approaches in an attempt to map the putative disease locus. First, mapping the source of linkage to chromosome 17 in pedigree C702. We refined the linkage region in family C702 to a 21-marker segment spanning 11.7 Mb at 17q23-q24 by genotyping a total of 50 microsatellites across chromosome 17 in the pedigree. Analysis of data from 1028 single nucleotide polymorphisms (SNPs) across the refined linkage region identified a single region of homozygosity present in pedigree C702 but not in 2938 UK controls. This spanned ~432 kb of the gene encoding protein kinase C, alpha (PRKCA), the encoded protein of which has been implicated in the pathogenesis of psychiatric disorders. Analysis of pedigree C702 by oligonucleotide-array comparative genome hybridization excluded the possibility that this region of homozygosity was because of a deletion. Mutation screening of PRKCA identified a rare, four-marker haplotype (C-HAP) in the 3' untranslated region of the gene, which was present in the homozygous state in all six affected members of pedigree C702. No other homozygotes were observed in genotype data for a total of 6597 unrelated Europeans (case N=1755, control N=3580 and parents of probands N=1262). Second, association analysis of C702 alleles at PRKCA. The low-frequency haplotype (C-HAP) showed a trend for association in a study of unrelated schizophrenia cases and controls from the UK (661 cases, 2824 controls, P=0.078 and odd ratio (OR)=1.9) and significant evidence for association when the sample was expanded to include cases with bipolar (N=710) and schizoaffective disorder (N=50) (psychosis sample: 1421 cases, 2824 controls, P=0.037 and OR=1.9). Given that all the affected members of C702 are male, we also undertook sex-specific analyses. This revealed that the association was strongest in males for both schizophrenia (446 male cases, 1421 male controls, P=0.008 and OR=3.9) and in the broader psychosis group (730 male cases, 1421 male controls, P=0.008 and OR=3.6). Analysis of C-HAP in follow-up samples from Ireland and Bulgaria revealed no evidence for association in either the whole sample or in males alone, and meta-analysis of all male psychosis samples yielded no significant evidence of association (969 male cases, 1939 male controls, 311 male probands P=0.304 and OR=1.4). Third, association mapping of the pedigree C702 linkage region. Independent of pedigree C702, genotype data from the Affymetrix 500k GeneChip set were available for 476 patients with schizophrenia and 2938 controls from the United Kingdom. SNPs in PRKCA showed evidence for association with schizophrenia that achieved gene-wide significance (P=0.027). Moreover, the same SNP was the most significantly associated marker out of the 1028 SNPs genotyped across the linkage region (rs873417, allelic P=0.0004). Follow-up genotyping in samples from Ireland, Bulgaria and Germany did not show consistent replication, but meta-analysis of all samples (4116 cases and 6491 controls) remained nominally significant (meta-analysis P=0.026, OR=1.1). We conclude that, although we have obtained convergent lines of evidence implicating both rare and common schizophrenia risk variants at PRKCA, none of these is individually compelling. However, the evidence across all approaches suggests that further study of this locus is warranted.
Collapse
|
293
|
Yang H, Liu J, Sui J, Pearlson G, Calhoun VD. A Hybrid Machine Learning Method for Fusing fMRI and Genetic Data: Combining both Improves Classification of Schizophrenia. Front Hum Neurosci 2010; 4:192. [PMID: 21119772 PMCID: PMC2990459 DOI: 10.3389/fnhum.2010.00192] [Citation(s) in RCA: 129] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2010] [Accepted: 09/24/2010] [Indexed: 11/25/2022] Open
Abstract
We demonstrate a hybrid machine learning method to classify schizophrenia patients and healthy controls, using functional magnetic resonance imaging (fMRI) and single nucleotide polymorphism (SNP) data. The method consists of four stages: (1) SNPs with the most discriminating information between the healthy controls and schizophrenia patients are selected to construct a support vector machine ensemble (SNP-SVME). (2) Voxels in the fMRI map contributing to classification are selected to build another SVME (Voxel-SVME). (3) Components of fMRI activation obtained with independent component analysis (ICA) are used to construct a single SVM classifier (ICA-SVMC). (4) The above three models are combined into a single module using a majority voting approach to make a final decision (Combined SNP-fMRI). The method was evaluated by a fully validated leave-one-out method using 40 subjects (20 patients and 20 controls). The classification accuracy was: 0.74 for SNP-SVME, 0.82 for Voxel-SVME, 0.83 for ICA-SVMC, and 0.87 for Combined SNP-fMRI. Experimental results show that better classification accuracy was achieved by combining genetic and fMRI data than using either alone, indicating that genetic and brain function representing different, but partially complementary aspects, of schizophrenia etiopathology. This study suggests an effective way to reassess biological classification of individuals with schizophrenia, which is also potentially useful for identifying diagnostically important markers for the disorder.
Collapse
Affiliation(s)
- Honghui Yang
- Department of Environment Engineering, Northwestern Polytechnical University Xi'an, China
| | | | | | | | | |
Collapse
|
294
|
Li X, Jope RS. Is glycogen synthase kinase-3 a central modulator in mood regulation? Neuropsychopharmacology 2010; 35:2143-54. [PMID: 20668436 PMCID: PMC3055312 DOI: 10.1038/npp.2010.105] [Citation(s) in RCA: 233] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2010] [Revised: 06/04/2010] [Accepted: 06/21/2010] [Indexed: 02/05/2023]
Abstract
Little is known regarding the mechanisms underlying the complex etiology of mood disorders, represented mainly by major depressive disorder and bipolar disorder. The 1996 discovery that lithium inhibits glycogen synthase kinase-3 (GSK3) raised the possibility that impaired inhibition of GSK3 is associated with mood disorders. This is now supported by evidence from animal biochemical, pharmacological, molecular, and behavioral studies and from human post-mortem brain, peripheral tissue, and genetic studies that are reviewed here. Mood disorders may result in part from impairments in mechanisms controlling the activity of GSK3 or GSK3-regulated functions, and disruptions of these regulating systems at different signaling sites may contribute to the heterogeneity of mood disorders. This substantial evidence supports the conclusion that bolstering the inhibitory control of GSK3 is an important component of the therapeutic actions of drugs used to treat mood disorders and that GSK3 is a valid target for developing new therapeutic interventions.
Collapse
Affiliation(s)
- Xiaohua Li
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | | |
Collapse
|
295
|
Buonanno A. The neuregulin signaling pathway and schizophrenia: from genes to synapses and neural circuits. Brain Res Bull 2010; 83:122-31. [PMID: 20688137 PMCID: PMC2958213 DOI: 10.1016/j.brainresbull.2010.07.012] [Citation(s) in RCA: 129] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2010] [Revised: 07/14/2010] [Accepted: 07/20/2010] [Indexed: 02/07/2023]
Abstract
Numerous genetic linkage and association studies implicate members of the Neuregulin-ErbB receptor (NRG-ErbB) signaling pathway as schizophrenia "at risk" genes. An emphasis of this review is to propose plausible neurobiological mechanisms, regulated by the Neuregulin-ErbB signaling network, that may be altered in schizophrenia and contribute to its etiology. To this end, the distinct neurotransmitter pathways, neuronal subtypes and neural network systems altered in schizophrenia are initially discussed. Next, the review focuses on the possible significance of genetic studies associating NRG1 and ErbB4 with schizophrenia, in light of the functional role of this signaling pathway in regulating glutamatergic, GABAergic and dopaminergic neurotransmission, as well as modulating synaptic plasticity and gamma oscillations. The importance of restricted ErbB4 receptor expression in GABAergic interneurons is emphasized, particularly their expression at glutamatergic synapses of parvalbumin-positive fast-spiking interneurons where modulation of inhibitory drive could account for the dramatic effects of NRG-ErbB signaling on gamma oscillations and pyramidal neuron output. A case is made for reasons that the NRG-ErbB signaling pathway constitutes a "biologically plausible" system for understanding the pathogenic mechanisms that may underlie the complex array of positive, negative and cognitive deficits associated with schizophrenia during development.
Collapse
Affiliation(s)
- Andrés Buonanno
- National Institutes of Health, Eunice Shriver Kennedy NICHD, Section on Molecular Neurobiology, Program of Developmental Neurobiology, 35 Lincoln Drive, Bethesda, MD 20892-3714, USA.
| |
Collapse
|
296
|
Song W, Li W, Noltner K, Yan J, Green E, Grozeva D, Jones IR, Craddock N, Longmate J, Feng J, Sommer SS. Identification of high risk DISC1 protein structural variants in patients with bipolar spectrum disorder. Neurosci Lett 2010; 486:136-40. [PMID: 20850505 DOI: 10.1016/j.neulet.2010.09.027] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2009] [Revised: 08/18/2010] [Accepted: 09/10/2010] [Indexed: 01/15/2023]
Abstract
In a large Scottish pedigree, a balanced translocation t (1;11)(q42.1;q14.3) disrupting the DISC1 and DISC2 genes segregates with major mental illness, including schizophrenia and depression. A frame-shift carboxyl-terminal deletion was reported in DISC1 in an American family with schizophrenia, but subsequently found in two controls. Herein, we test one hypothesis utilizing a large scale case-control mutation analysis: uncommon DISC1 variants are associated with high risk for bipolar spectrum disorder. We have analyzed the regions of likely functional significance in the DISC1 gene in 504 patients with bipolar spectrum disorder and 576 ethnically similar controls. Five patients were heterozygous for ultra-rare protein structural variants not found in the 576 controls (p=0.02, one-sided Fisher's exact test) and shown to be ultra-rare by their absence in a pool of 10,000 control alleles. In our sample, ultra-rare (private) protein structural variants in DISC1 are associated with an estimated attributable risk of about 0.5% in bipolar spectrum disorder. These data are consistent with: (i) the high frequency of depression in the large Scottish family with a translocation disrupting DISC1; (ii) linkage disequilibrium analysis demonstrating haplotypes associated with relatively small increases in risk for bipolar disorder (<3-fold odds ratio). The data illustrate how low/moderate risk haplotypes that might be found by the HapMap project can be followed up by resequencing to identify protein structural variants with high risk, low frequency and of potential clinical utility.
Collapse
Affiliation(s)
- Wenjia Song
- Department of Molecular Genetics, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, United States
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
297
|
Abstract
A pressing need for interrater reliability in the diagnosis of mental disorders emerged during the mid-twentieth century, prompted in part by the development of diverse new treatments. The Diagnostic and Statistical Manual of Mental Disorders (DSM), third edition answered this need by introducing operationalized diagnostic criteria that were field-tested for interrater reliability. Unfortunately, the focus on reliability came at a time when the scientific understanding of mental disorders was embryonic and could not yield valid disease definitions. Based on accreting problems with the current DSM-fourth edition (DSM-IV) classification, it is apparent that validity will not be achieved simply by refining criteria for existing disorders or by the addition of new disorders. Yet DSM-IV diagnostic criteria dominate thinking about mental disorders in clinical practice, research, treatment development, and law. As a result, the modern DSM system, intended to create a shared language, also creates epistemic blinders that impede progress toward valid diagnoses. Insights that are beginning to emerge from psychology, neuroscience, and genetics suggest possible strategies for moving forward.
Collapse
Affiliation(s)
- Steven E Hyman
- Department of Neurobiology, Harvard Medical School, Harvard University, Cambridge, Massachusetts 02138, USA.
| |
Collapse
|
298
|
Pedrosa E, Shah A, Tenore C, Capogna M, Villa C, Guo X, Zheng D, Lachman HM. β-catenin promoter ChIP-chip reveals potential schizophrenia and bipolar disorder gene network. J Neurogenet 2010; 24:182-93. [PMID: 20615089 DOI: 10.3109/01677063.2010.495182] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Therapeutic concentrations of lithium salts inhibit glycogen synthase kinase 3 beta (GSK3β) and phosphoinositide (PI) signaling suggesting that abnormal activation of these pathways could be a factor in the pathophysiology of bipolar disorder (BD). Involvement of these pathways is also supported by recent genome-wide association studies (GWASs). One way investigators have investigated the molecular basis of BD and the therapeutic action of lithium is by microarray expression studies, since both GSK3β- and PI-mediated signal transduction pathways are coupled to transcriptional activation and inhibition. However, expression profiling has some limitations and investigators cannot use the approach to analyze fetal brain tissue, arguably the most relevant biological structure related to the development of genetically based psychiatric disorders. To address these shortcomings, the authors have taken a novel approach using chromatin immunoprecipitation-enriched material annealed to microarrays (ChIP-chip) targeting genes in fetal brain tissue bound by β-catenin, a transcription factor that is directly regulated by GSK3β. The promoters for 640 genes were found to be bound by β-catenin, many of which are known schizophrenia (SZ), autism spectrum disorder (ASD), and BD candidates, including CACNA1B, NRNG, SNAP29, FGFR1, PCDH9, and nine others identified in recently published GWASs and genome-wide searches for copy number variants (CNVs). The findings suggest that seemingly disparate candidate genes for SZ and BD can be incorporated into a common molecular network revolving around GSK3β/β-catenin signaling. In addition, the finding that a putative lithium-responsive pathway may influence a subgroup of SZ and ASD candidate genes could have therapeutic implications.
Collapse
Affiliation(s)
- Erika Pedrosa
- Department of Psychiatry and Behavioral Sciences, Division of Basic Research, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | | | | | | | | | | | | | | |
Collapse
|
299
|
Karam CS, Ballon JS, Bivens NM, Freyberg Z, Girgis RR, Lizardi-Ortiz JE, Markx S, Lieberman JA, Javitch JA. Signaling pathways in schizophrenia: emerging targets and therapeutic strategies. Trends Pharmacol Sci 2010; 31:381-90. [PMID: 20579747 DOI: 10.1016/j.tips.2010.05.004] [Citation(s) in RCA: 127] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2010] [Revised: 05/17/2010] [Accepted: 05/20/2010] [Indexed: 02/07/2023]
Abstract
Dopamine D(2) receptor antagonism is a unifying property of all antipsychotic drugs in use for schizophrenia. While often effective at ameliorating psychosis, these drugs are largely ineffective at treating negative and cognitive symptoms. Increasing attention is being focused on the complex genetics of the illness and the signaling pathways implicated in its pathophysiology. We review targeted approaches for pharmacotherapy involving the glutamatergic, GABAergic and cholinergic pathways. We also describe several of the major genetic findings that identify signaling pathways representing potential targets for novel pharmacological intervention. These include genes in the 22q11 locus, DISC1, Neuregulin 1/ErbB4, and components of the Akt/GSK-3 pathway.
Collapse
Affiliation(s)
- Caline S Karam
- Department of Psychiatry, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
300
|
Miller BH, Wahlestedt C. MicroRNA dysregulation in psychiatric disease. Brain Res 2010; 1338:89-99. [PMID: 20303342 PMCID: PMC2891055 DOI: 10.1016/j.brainres.2010.03.035] [Citation(s) in RCA: 149] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2010] [Revised: 02/04/2010] [Accepted: 03/11/2010] [Indexed: 12/24/2022]
Abstract
MicroRNAs (miRNAs) are small regulatory RNAs that individually regulate up to several hundred genes, and collectively may regulate as much as two-thirds of the transcriptome. Recent evidence supports a role for miRNA dysregulation in psychiatric and neurological disorders, including schizophrenia, bipolar disorder, and autism. Small changes in miRNA expression can fine-tune the expression of multiple genes within a biological network, suggesting that miRNA dysregulation may underlie many of the molecular changes observed in psychiatric disease, and that therapeutic regulation of miRNA levels may represent a novel treatment option.
Collapse
Affiliation(s)
- Brooke H. Miller
- Department of Neuroscience, Scripps Florida, Jupiter, FL 33458 USA
| | - Claes Wahlestedt
- Department of Neuroscience, Scripps Florida, Jupiter, FL 33458 USA
| |
Collapse
|