51
|
Lepagnol-Bestel AM, Kvajo M, Karayiorgou M, Simonneau M, Gogos JA. A Disc1 mutation differentially affects neurites and spines in hippocampal and cortical neurons. Mol Cell Neurosci 2013; 54:84-92. [PMID: 23396153 DOI: 10.1016/j.mcn.2013.01.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Revised: 01/21/2013] [Accepted: 01/24/2013] [Indexed: 02/05/2023] Open
Abstract
A balanced chromosomal translocation segregating with schizophrenia and affective disorders in a large Scottish family disrupting DISC1 implicated this gene as a susceptibility gene for major mental illness. Here we study neurons derived from a genetically engineered mouse strain with a truncating lesion disrupting the endogenous Disc1 ortholog. We provide a detailed account of the consequences of this mutation on axonal and dendritic morphogenesis as well as dendritic spine development in cultured hippocampal and cortical neurons. We show that the mutation has distinct effects on these two types of neurons, supporting a cell-type specific role of Disc1 in establishing structural connections among neurons. Moreover, using a validated antibody we provide evidence indicating that Disc1 localizes primarily to Golgi apparatus-related vesicles. Our results support the notion that in vitro cultures derived from Disc1(Tm1Kara) mice provide a valuable model for future mechanistic analysis of the cellular and biochemical effects of this mutation, and can thus serve as a platform for drug discovery efforts.
Collapse
Affiliation(s)
- A M Lepagnol-Bestel
- Department of Physiology and Cellular Biophysics, Columbia University, 630 West, 168th Street, New York, NY 10032, USA
| | | | | | | | | |
Collapse
|
52
|
Thomson PA, Malavasi ELV, Grünewald E, Soares DC, Borkowska M, Millar JK. DISC1 genetics, biology and psychiatric illness. FRONTIERS IN BIOLOGY 2013; 8:1-31. [PMID: 23550053 PMCID: PMC3580875 DOI: 10.1007/s11515-012-1254-7] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Psychiatric disorders are highly heritable, and in many individuals likely arise from the combined effects of genes and the environment. A substantial body of evidence points towards DISC1 being one of the genes that influence risk of schizophrenia, bipolar disorder and depression, and functional studies of DISC1 consequently have the potential to reveal much about the pathways that lead to major mental illness. Here, we review the evidence that DISC1 influences disease risk through effects upon multiple critical pathways in the developing and adult brain.
Collapse
Affiliation(s)
- Pippa A Thomson
- The Centre for Molecular Medicine at the Medical Research Council Institute of Genetics and Molecular Medicine, The University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK
| | | | | | | | | | | |
Collapse
|
53
|
Narayan S, Nakajima K, Sawa A. DISC1: a key lead in studying cortical development and associated brain disorders. Neuroscientist 2013; 19:451-64. [PMID: 23300216 DOI: 10.1177/1073858412470168] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
For the past decade, DISC1 has been studied as a promising lead to understand the biology underlying major mental illnesses, such as schizophrenia. Consequently, many review articles on DISC1 have been published. In this article, rather than repeating comprehensive overviews of research articles, we will introduce the utility of DISC1 in the study of cortical development in association with a wide range of developmental brain disorders. Cortical development involves cell autonomous and cell nonautonomous mechanisms as well as host responses to environmental factors, all of which involve DISC1 function. Thus, we will discuss the significance of DISC1 in forming an overall understanding of multiple mechanisms that orchestrate corticogenesis and can serve as therapeutic targets in diseases caused by abnormal cortical development.
Collapse
Affiliation(s)
- Soumya Narayan
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | | | | |
Collapse
|
54
|
Staples CJ, Myers KN, Beveridge RDD, Patil AA, Lee AJX, Swanton C, Howell M, Boulton SJ, Collis SJ. The centriolar satellite protein Cep131 is important for genome stability. J Cell Sci 2012; 125:4770-9. [PMID: 22797915 DOI: 10.1242/jcs.104059] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The centrosome acts as a centre for microtubule organisation and plays crucial roles in cell polarity, migration, growth and division. Cep131 has recently been described as a basal body component essential for cilium formation, but its function in non-ciliogenic cells is unknown. We identified human Cep131 (also known as AZI1) in a screen for regulators of genome stability. We show that centrosomal localisation of Cep131 is cell-cycle-regulated and requires both an intact microtubule network and a functional dynein-dynactin transport system. Cep131 is recruited to centriolar satellites by PCM1, and localised to the centriolar core region by both pericentrin and Cep290. Depletion of Cep131 results in a reduction in proliferation rate, centriole amplification, an increased frequency of multipolar mitosis, chromosomal instability and an increase in post-mitotic DNA damage. These data therefore highlight the importance of human Cep131 for maintaining genomic integrity.
Collapse
Affiliation(s)
- Christopher J Staples
- Genome Stability Group, CR-UK/YCR Sheffield Cancer Research Centre, YCR Institute for Cancer Studies, Department of Oncology, University of Sheffield Medical School, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
55
|
Rao S, Ge S, Shelly M. Centrosome positioning and primary cilia assembly orchestrate neuronal development. ACTA ACUST UNITED AC 2012. [DOI: 10.1007/s11515-012-1231-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
56
|
Stowe TR, Wilkinson CJ, Iqbal A, Stearns T. The centriolar satellite proteins Cep72 and Cep290 interact and are required for recruitment of BBS proteins to the cilium. Mol Biol Cell 2012; 23:3322-35. [PMID: 22767577 PMCID: PMC3431927 DOI: 10.1091/mbc.e12-02-0134] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The ciliopathy-associated proteins Cep290 and BBS4 localize to cytoplasmic particles called centriolar satellites, yet the significance of this association is unknown. A new component of satellites, Cep72, is identified. Its role in the regulation of Cep290 and BBS4 is described, as are developmental defects resulting from loss of satellites in zebrafish. Defects in centrosome and cilium function are associated with phenotypically related syndromes called ciliopathies. Centriolar satellites are centrosome-associated structures, defined by the protein PCM1, that are implicated in centrosomal protein trafficking. We identify Cep72 as a PCM1-interacting protein required for recruitment of the ciliopathy-associated protein Cep290 to centriolar satellites. Loss of centriolar satellites by depletion of PCM1 causes relocalization of Cep72 and Cep290 from satellites to the centrosome, suggesting that their association with centriolar satellites normally restricts their centrosomal localization. We identify interactions between PCM1, Cep72, and Cep290 and find that disruption of centriolar satellites by overexpression of Cep72 results in specific aggregation of these proteins and the BBSome component BBS4. During ciliogenesis, BBS4 relocalizes from centriolar satellites to the primary cilium. This relocalization occurs normally in the absence of centriolar satellites (PCM1 depletion) but is impaired by depletion of Cep290 or Cep72, resulting in defective ciliary recruitment of the BBSome subunit BBS8. We propose that Cep290 and Cep72 in centriolar satellites regulate the ciliary localization of BBS4, which in turn affects assembly and recruitment of the BBSome. Finally, we show that loss of centriolar satellites in zebrafish leads to phenotypes consistent with cilium dysfunction and analogous to those observed in human ciliopathies.
Collapse
Affiliation(s)
- Timothy R Stowe
- Department of Biology, Stanford School of Medicine, Stanford, CA 94305-5020, USA
| | | | | | | |
Collapse
|
57
|
Kwan KY, Sestan N, Anton ES. Transcriptional co-regulation of neuronal migration and laminar identity in the neocortex. Development 2012; 139:1535-46. [PMID: 22492350 DOI: 10.1242/dev.069963] [Citation(s) in RCA: 258] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The cerebral neocortex is segregated into six horizontal layers, each containing unique populations of molecularly and functionally distinct excitatory projection (pyramidal) neurons and inhibitory interneurons. Development of the neocortex requires the orchestrated execution of a series of crucial processes, including the migration of young neurons into appropriate positions within the nascent neocortex, and the acquisition of layer-specific neuronal identities and axonal projections. Here, we discuss emerging evidence supporting the notion that the migration and final laminar positioning of cortical neurons are also co-regulated by cell type- and layer-specific transcription factors that play concomitant roles in determining the molecular identity and axonal connectivity of these neurons. These transcriptional programs thus provide direct links between the mechanisms controlling the laminar position and identity of cortical neurons.
Collapse
Affiliation(s)
- Kenneth Y Kwan
- Department of Neurobiology and Kavli Institute for Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
| | | | | |
Collapse
|
58
|
Whalley HC, Sussmann JE, Johnstone M, Romaniuk L, Redpath H, Chakirova G, Mukherjee P, Hall J, Johnstone EC, Lawrie SM, McIntosh AM. Effects of a mis-sense DISC1 variant on brain activation in two cohorts at high risk of bipolar disorder or schizophrenia. Am J Med Genet B Neuropsychiatr Genet 2012; 159B:343-53. [PMID: 22337479 DOI: 10.1002/ajmg.b.32035] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Accepted: 01/24/2012] [Indexed: 12/11/2022]
Abstract
Bipolar disorder and schizophrenia share a number of clinical features and genetic risk variants of small effect, suggesting overlapping pathogenic mechanisms. The effect of single genetic risk variants on brain function is likely to differ in people at high familial risk versus controls as these individuals have a higher overall genetic loading and are therefore closer to crossing a threshold of disease liability. Therefore, whilst the effects of genetic risk variants on brain function may be similar across individuals at risk of both disorders, they are hypothesized to differ compared to that seen in control subjects. We sought to examine the effects of the DISC1 Leu(607) Phe polymorphism on brain activation in young healthy individuals at familial risk of bipolar disorder (n = 84), in a group of controls (n = 78), and in a group at familial risk of schizophrenia (n = 47), performing a language task. We assessed whether genotype effects on brain activation differed according to risk status. There was a significant genotype × group interaction in a cluster centered on the left pre/postcentral gyrus, extending to the inferior frontal gyrus. The origin of this genotype × group effect originated from a significant effect of the presumed risk variant (Phe) on brain activation in the control group, which was absent in both high-risk groups. Differential effects of this polymorphism in controls compared to the two familial groups suggests a commonality of effect across individuals at high-risk of the disorders, which is likely to be dependant upon existing genetic background.
Collapse
Affiliation(s)
- Heather C Whalley
- Division of Psychiatry, University of Edinburgh, Royal Edinburgh Hospital, Edinburgh, UK.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
59
|
Abstract
Chronic mental diseases (CMD) like the schizophrenias are progressive diseases of heterogenous but poorly understood biological origin. An imbalance in proteostasis is a hallmark of dysfunctional neurons, leading to impaired clearance and abnormal deposition of protein aggregates. Thus, it can be hypothesized that unbalanced proteostasis in such neurons may also lead to protein aggregates in schizophrenia. These protein aggregates, however, would be more subtle then in the classical neurodegenerative diseases and as such have not yet been detected. The DISC1 (Disrupted-in-schizophrenia 1) gene is considered among the most promising candidate genes for CMD having been identified as linked to CMD in a Scottish pedigree and having since been found to associate to various phenotypes of CMD. We have recently demonstrated increased insoluble DISC1 protein in the cingular cortex in approximately 20% of cases of CMD within the widely used Stanley Medical Research Institute Consortium Collection. Surprisingly, in vitro, DISC1 aggregates were cell-invasive, i.e., purified aggresomes or recombinant DISC1 fragments where internalized at an efficiency comparable to that of α-synuclein. Intracellular DISC1 aggresomes acquired gain-of-function properties in recruiting otherwise soluble proteins such as the candidate schizophrenia protein dysbindin. Disease-associated DISC1 polymorphism S704C led to a higher oligomerization tendency of DISC1. These findings justify classification of DISC1-dependent brain disorders as protein conformational disorders which we have tentatively termed DISC1opathies. The notion of disturbed proteostasis and protein aggregation as a mechanism of mental diseases is thus emerging. The yet unidentified form of neuronal impairment in CMD is more subtle than in the classical neurodegenerative diseases without leading to massive cell death and as such present a different kind of neuronal dysfunctionality, eventually confined to highly selective CNS subpopulations.
Collapse
Affiliation(s)
- Carsten Korth
- Department of Neuropathology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
60
|
Bradshaw NJ, Porteous DJ. DISC1-binding proteins in neural development, signalling and schizophrenia. Neuropharmacology 2012; 62:1230-41. [PMID: 21195721 PMCID: PMC3275753 DOI: 10.1016/j.neuropharm.2010.12.027] [Citation(s) in RCA: 150] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Revised: 12/17/2010] [Accepted: 12/22/2010] [Indexed: 12/18/2022]
Abstract
In the decade since Disrupted in Schizophrenia 1 (DISC1) was first identified it has become one of the most convincing risk genes for major mental illness. As a multi-functional scaffold protein, DISC1 has multiple identified protein interaction partners that highlight pathologically relevant molecular pathways with potential for pharmaceutical intervention. Amongst these are proteins involved in neuronal migration (e.g. APP, Dixdc1, LIS1, NDE1, NDEL1), neural progenitor proliferation (GSK3β), neurosignalling (Girdin, GSK3β, PDE4) and synaptic function (Kal7, TNIK). Furthermore, emerging evidence of genetic association (NDEL1, PCM1, PDE4B) and copy number variation (NDE1) implicate several DISC1-binding partners as risk factors for schizophrenia in their own right. Thus, a picture begins to emerge of DISC1 as a key hub for multiple critical developmental pathways within the brain, disruption of which can lead to a variety of psychiatric illness phenotypes.
Collapse
Key Words
- disc1
- schizophrenia
- neurodevelopment
- signalling
- synapse
- association studies
- app, amyloid precursor protein
- atf4, activating transcription factor 4
- bace1, β-site app-cleaving enzyme-1
- bbs4, bardet–biedl syndrome 4
- cep290, centrosomal protein 290 kda
- cnv, copy number variation
- cre, camp response element
- dbz, disc1-binding zinc finger
- disc1, disrupted in schizophrenia 1
- dixdc1, dishevelled-axin domain containing-1
- fez1, fasciculation and elongation protein zeta 1
- glur, glutamate receptor
- gsk3β, glycogen synthase kinase 3β
- kal7, kalirin-7
- lef/tcf, lymphoid enhancer factor/t cell factor
- lis1, lissencephaly 1
- mtor, mammalian target of rapamycin
- nde1, nuclear distribution factor e homologue 1 or nuclear distribution element 1
- ndel1, nde-like 1
- nrg, neuregulin
- pacap, pituitary adenylate cyclase-activating polypeptide
- pcm1, pericentriolar material 1
- pcnt, pericentrin
- pde4, phosphodiesterase 4
- pi3 k, phosphatidylinositiol 3-kinase
- psd, post-synaptic density
- rac1, ras-related c3 botulinum toxin substrate 1
- tnik, traf2 and nck interacting kinase
Collapse
Affiliation(s)
- Nicholas J. Bradshaw
- Medical Genetics Section, Molecular Medicine Centre, Institute of Genetics & Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road South, Edinburgh, Midlothian EH4 2XU, UK
| | | |
Collapse
|
61
|
Atkin TA, Brandon NJ, Kittler JT. Disrupted in Schizophrenia 1 forms pathological aggresomes that disrupt its function in intracellular transport. Hum Mol Genet 2012; 21:2017-28. [DOI: 10.1093/hmg/dds018] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
62
|
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: 4.7] [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
|
63
|
Kamiya A, Sedlak TW, Pletnikov MV. DISC1 Pathway in Brain Development: Exploring Therapeutic Targets for Major Psychiatric Disorders. Front Psychiatry 2012; 3:25. [PMID: 22461775 PMCID: PMC3310233 DOI: 10.3389/fpsyt.2012.00025] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2011] [Accepted: 03/06/2012] [Indexed: 01/30/2023] Open
Abstract
Genetic risk factors for major psychiatric disorders play key roles in neurodevelopment. Thus, exploring the molecular pathways of risk genes is important not only for understanding the molecular mechanisms underlying brain development, but also to decipher how genetic disturbances affect brain maturation and functioning relevant to major mental illnesses. During the last decade, there has been significant progress in determining the mechanisms whereby risk genes impact brain development. Nonetheless, given that the majority of psychiatric disorders have etiological complexities encompassing multiple risk genes and environmental factors, the biological mechanisms of these diseases remain poorly understood. How can we move forward to our research for discovery of the biological markers and novel therapeutic targets for major mental disorders? Here we review recent progress in the neurobiology of disrupted in schizophrenia 1 (DISC1), a major risk gene for major mental disorders, with a particular focus on its roles in cerebral cortex development. Convergent findings implicate DISC1 as part of a large, multi-step pathway implicated in various cellular processes and signal transduction. We discuss links between the DISC1 pathway and environmental factors, such as immune/inflammatory responses, which may suggest novel therapeutic targets. Existing treatments for major mental disorders are hampered by a limited number of pharmacological targets. Consequently, elucidation of the DISC1 pathway, and its association with neuropsychiatric disorders, may offer hope for novel treatment interventions.
Collapse
Affiliation(s)
- Atsushi Kamiya
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine Baltimore, MD, USA
| | | | | |
Collapse
|
64
|
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: 331] [Impact Index Per Article: 25.5] [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
|
65
|
Soares DC, Carlyle BC, Bradshaw NJ, Porteous DJ. DISC1: Structure, Function, and Therapeutic Potential for Major Mental Illness. ACS Chem Neurosci 2011; 2:609-632. [PMID: 22116789 PMCID: PMC3222219 DOI: 10.1021/cn200062k] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Accepted: 08/05/2011] [Indexed: 01/09/2023] Open
Abstract
![]()
Disrupted in schizophrenia 1 (DISC1) is well established
as a genetic risk factor across a spectrum of psychiatric disorders,
a role supported by a growing body of biological studies, making the
DISC1 protein interaction network an attractive therapeutic target.
By contrast, there is a relative deficit of structural information
to relate to the myriad biological functions of DISC1. Here, we critically
appraise the available bioinformatics and biochemical analyses on
DISC1 and key interacting proteins, and integrate this with the genetic
and biological data. We review, analyze, and make predictions regarding
the secondary structure and propensity for disordered regions within
DISC1, its protein-interaction domains, subcellular localization motifs,
and the structural and functional implications of common and ultrarare DISC1 variants associated with major mental illness. We
discuss signaling pathways of high pharmacological potential wherein
DISC1 participates, including those involving phosphodiesterase 4
(PDE4) and glycogen synthase kinase 3 (GSK3). These predictions and
priority areas can inform future research in the translational and
potentially guide the therapeutic processes.
Collapse
Affiliation(s)
- Dinesh C. Soares
- Medical Genetics Section, Molecular
Medicine Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital,
Crewe Road South, Edinburgh EH4 2XU, United Kingdom
| | - Becky C. Carlyle
- Department of Psychiatry, Yale University School of Medicine, 300 George Street,
Suite 901, New Haven, Connecticut 06511, United States
| | - Nicholas J. Bradshaw
- Medical Genetics Section, Molecular
Medicine Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital,
Crewe Road South, Edinburgh EH4 2XU, United Kingdom
| | - David J. Porteous
- Medical Genetics Section, Molecular
Medicine Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital,
Crewe Road South, Edinburgh EH4 2XU, United Kingdom
| |
Collapse
|
66
|
Porteous DJ, Millar JK, Brandon NJ, Sawa A. DISC1 at 10: connecting psychiatric genetics and neuroscience. Trends Mol Med 2011; 17:699-706. [PMID: 22015021 DOI: 10.1016/j.molmed.2011.09.002] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Revised: 09/12/2011] [Accepted: 09/20/2011] [Indexed: 11/17/2022]
Abstract
Psychiatric genetics research, as exemplified by the DISC1 gene, aspires to inform on mental health etiology and to suggest improved strategies for intervention. DISC1 was discovered in 2000 through the molecular cloning of a chromosomal translocation that segregated with a spectrum of major mental illnesses in a single large Scottish family. Through in vitro experiments and mouse models, DISC1 has been firmly established as a genetic risk factor for a spectrum of psychiatric illness. As a consequence of its protein scaffold function, the DISC1 protein impacts on many aspects of brain function, including neurosignaling and neurodevelopment. DISC1 is a pathfinder for understanding psychopathology, brain development, signaling and circuitry. Although much remains to be learnt and understood, potential targets for drug development are starting to emerge, and in this review, we will discuss the 10 years of research that has helped us understand key roles of DISC1 in psychiatric disease.
Collapse
Affiliation(s)
- David J Porteous
- Molecular Medicine Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK.
| | | | | | | |
Collapse
|
67
|
Centriolar satellites: busy orbits around the centrosome. Eur J Cell Biol 2011; 90:983-9. [PMID: 21945726 DOI: 10.1016/j.ejcb.2011.07.007] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2011] [Revised: 07/20/2011] [Accepted: 07/20/2011] [Indexed: 01/26/2023] Open
Abstract
Since its first description by Theodor Boveri in 1888, the centrosome has been studied intensely, and it revealed detailed information about its structure, molecular composition and its various functions. The centrosome consists of two centrioles, which generally appear in electron microscopy as barrel-shaped structures usually composed of nine microtubule triplets. An amorphous mass of pericentriolar material surrounds the centrioles and accumulates many proteins important for the integrity and function of centrosomes, such as the γ-tubulin ring complex (γ-TuRC) that mediates microtubule nucleation and capping. In animal somatic cells, the centrosome generally accounts for the major microtubule organizing center, and the duplicated pair of centrosomes determines the poles of the microtubule-based mitotic spindle. Despite detailed insights into the centrosome's structure and function, it has been a complete mystery until a few years ago how centrosomes duplicate and assemble. Moreover, it is still largely unclear if and how centrosomal proteins or protein complexes are exchanged, replaced or qualitatively altered. Previously identified cytoplasmic granules, named "pericentriolar" or "centriolar satellites", might fulfil such functions in protein targeting and exchange, and communication between the centrosomes and the cytoplasm. In this review, we summarize current knowledge about the structure, molecular composition and possible roles of the satellites that seem to surround the core of the centrosome in most animal cells.
Collapse
|
68
|
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: 9.1] [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
|
69
|
Lin CY, Sawa A, Jaaro-Peled H. Better understanding of mechanisms of schizophrenia and bipolar disorder: from human gene expression profiles to mouse models. Neurobiol Dis 2011; 45:48-56. [PMID: 21914480 DOI: 10.1016/j.nbd.2011.08.025] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Revised: 08/22/2011] [Accepted: 08/26/2011] [Indexed: 01/01/2023] Open
Abstract
The molecular mechanisms of major mental illnesses, such as schizophrenia and bipolar disorder, are unclear. To address this fundamental question, many groups have studied molecular expression profiles in postmortem brains and other tissues from patients compared with those from normal controls. Development of unbiased high-throughput approaches, such as microarray, RNA-seq, and proteomics, have supported and facilitated this endeavor. In addition to genes directly involved in neuron/glia signaling, especially those encoding for synaptic proteins, genes for metabolic cascades are differentially expressed in the brains of patients with schizophrenia and bipolar disorder, compared with those from normal controls in DNA microarray studies. Here we propose the importance and usefulness of genetic mouse models in which such differentially expressed molecules are modulated. These animal models allow us to dissect the mechanisms of how such molecular changes in patient brains may play a role in neuronal circuitries and overall behavioral phenotypes. We also point out that models in which the metabolic genes are modified are obviously untested from mental illness viewpoints, suggesting the potential to re-address these models with behavioral assays and neurochemical assessments.
Collapse
Affiliation(s)
- Chi-Ying Lin
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | |
Collapse
|
70
|
Kelly MP, Brandon NJ. Taking a bird’s eye view on a mouse model review: a comparison of findings from mouse models targeting DISC1 or DISC1-interacting proteins. FUTURE NEUROLOGY 2011. [DOI: 10.2217/fnl.11.39] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
DISC1 has garnered much interest from researchers trying to understand the neurobiology of psychiatric disease. DISC1 appears to function as a structural protein hub for a number of molecules, many of which are considered disease-relevant targets in their own right. Thus, in this article, we compare behavioral, anatomical and biochemical findings in genetic mouse models of DISC1 and DISC1-interacting proteins to better understand how dysfunction of DISC1 and/or its interactors could contribute to psychiatric pathophysiology through convergent effects on distinct cells, circuits and behaviors. Consistencies in phenotypes across mouse models suggest that DISC1 and its binding partners are particularly critical for working memory performance, proper neuronal migration and cortical volume, normal spine density, an intact monoaminergic system, proper levels of parvalbumin and normal cytokine/stress signaling in the rodent. If these DISC1 functions translate to humans, it would explain how alterations in DISC1 or DISC1 interactors could contribute to psychiatric pathophysiology. Identification of such a biological convergence will hopefully improve the development of novel therapeutics for patients by focusing efforts on specific domains that are affected by DISC1-related genetic risk architecture.
Collapse
Affiliation(s)
- Michy P Kelly
- Pfizer Neuroscience Research Unit, Eastern Point Road, Groton, CT 06340, USA
| | - Nicholas J Brandon
- Pfizer Neuroscience Research Unit, Eastern Point Road, Groton, CT 06340, USA
| |
Collapse
|
71
|
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: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [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
|
72
|
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
|
73
|
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: 3.2] [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
|
74
|
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
|
75
|
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: 1.8] [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
|
76
|
No association between the PCM1 gene and schizophrenia: a multi-center case-control study and a meta-analysis. Schizophr Res 2011; 129:80-4. [PMID: 21481569 DOI: 10.1016/j.schres.2011.03.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2011] [Revised: 03/15/2011] [Accepted: 03/24/2011] [Indexed: 12/20/2022]
Abstract
Alterations in centrosomal function have been suggested in the pathology of schizophrenia. The molecule pericentriolar material 1 (PCM1) is involved in maintaining centrosome integrity and in the regulation of the microtubule cytoskeleton. PCM1 forms a complex at the centrosome with the disrupted-in-schizophrenia 1 (DISC1) protein, which is a major susceptibility factor for schizophrenia. The association between genetic variants in the PCM1 gene and schizophrenia has been reported by several case-control studies, linkage studies and a meta-analysis. The aims of this study are to replicate the association between four single-nucleotide polymorphisms (SNPs) in the PCM1 gene and schizophrenia in a Japanese population (1496 cases and 1845 controls) and to perform a meta-analysis of the combined sample groups (3289 cases and 3567 controls). We failed to find a significant association between SNPs or haplotypes of the PCM1 gene and schizophrenia in the Japanese population (P>0.28). The meta-analysis did not reveal an association between the four examined SNPs and schizophrenia. Our data did not support genetic variants in the PCM1 gene as a susceptibility locus for schizophrenia.
Collapse
|
77
|
Brauns S, Gollub RL, Roffman JL, Yendiki A, Ho BC, Wassink TH, Heinz A, Ehrlich S. DISC1 is associated with cortical thickness and neural efficiency. Neuroimage 2011; 57:1591-600. [PMID: 21642004 DOI: 10.1016/j.neuroimage.2011.05.058] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Revised: 04/19/2011] [Accepted: 05/19/2011] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Disrupted in schizophrenia 1 (DISC1) is known to play a major role during brain development and is a candidate gene for schizophrenia. Cortical thickness is highly heritable and several MRI studies have shown widespread reductions of cortical thickness in patients with schizophrenia. Here, we investigated the effects of variation in DISC1 on cortical thickness. In a subsequent analysis we tested whether the identified DISC1 risk variant is also associated with neural activity during working memory functioning. METHODS We acquired structural MRI (sMRI), functional MRI (fMRI) and genotype data from 96 healthy volunteers. Separate cortical statistical maps for five single nucleotide polymorphisms (SNP) of DISC1 were generated to detect differences of cortical thickness in genotype groups across the entire cortical surface. Working-memory related load-dependent activation was measured during the Sternberg Item Recognition Paradigm and analyzed using a region-of-interest approach. RESULTS Phe allele carriers of the DISC1 SNP Leu607Phe had significantly reduced cortical thickness in the left supramarginal gyrus compared to Leu/Leu homozygotes. Neural activity in the left dorsolateral prefrontal cortex (DLPFC) during working memory task was increased in Phe allele carriers, whereas working memory performance did not differ between genotype groups. CONCLUSIONS This study provides convergent evidence for the effect of DISC1 risk variants on two independent brain-based intermediate phenotypes of schizophrenia. The same risk variant was associated with cortical thickness reductions and signs of neural inefficiency during a working memory task. Our findings provide further evidence for a neurodevelopmental model of schizophrenia.
Collapse
Affiliation(s)
- Stefan Brauns
- MGH/MIT/HMS Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA 02129-2000, USA
| | | | | | | | | | | | | | | |
Collapse
|
78
|
Bennouna-Greene V, Kremer S, Stoetzel C, Christmann D, Schuster C, Durand M, Verloes A, Sigaudy S, Holder-Espinasse M, Godet J, Brandt C, Marion V, Danion A, Dietemann JL, Dollfus H. Hippocampal dysgenesis and variable neuropsychiatric phenotypes in patients with Bardet-Biedl syndrome underline complex CNS impact of primary cilia. Clin Genet 2011; 80:523-31. [PMID: 21517826 DOI: 10.1111/j.1399-0004.2011.01688.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The Bardet-Biedl syndrome (BBS) is a rare ciliopathy clinically defined by the association of retinitis pigmentosa, polydactyly, obesity, kidney disease and cognitive impairment. The cognitive functioning, behavioral phenotype, prevalence of psychiatric diseases and memory performances of a cohort of 34 patients with BBS were evaluated and a systemic brain magnetic resonance imaging (MRI) was performed. The patients' cognitive functioning was of marked variable efficiency ranging from normal to disabling performances. Neuropsychological disorders such as slow thought process, attention difficulties and obsessive-compulsive traits were observed. Our main finding was hippocampal dysgenesis, diagnosed by MRI, found in 42.31% of the patients in this cohort. Moreover, we show that BBS proteins are expressed in the human hippocampus and in the human brain in the normal subject. Recent literature in the murine model shows that hippocampal neurogenesis, in particular in the adult mouse, requires an intact primary cilia. These results encourage us to further investigate the possible role of BBS proteins in the hippocampus and related central nervous system structures.
Collapse
Affiliation(s)
- V Bennouna-Greene
- Laboratoire de Génétique Médicale, Equipe Avenir-INSERM EA3949, Université de Strasbourg, F67085, France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
79
|
Abstract
Disrupted-in-Schizophrenia 1 (DISC1) is a strong candidate gene for schizophrenia and other mental disorders. DISC1 regulates neurodevelopmental processes including neurogenesis, neuronal migration, neurite outgrowth, and neurotransmitter signaling. Abnormal neuronal morphology and cortical architecture are seen in human postmortem brain from patients with schizophrenia. However, the etiology and development of these histological abnormalities remain unclear. We analyzed the histology of two Disc1 mutant mice with point mutations (Q31L and L100P) and found a relative reduction in neuron number, decreased neurogenesis, and altered neuron distribution compared to wild-type littermates. Frontal cortical neurons have shorter dendrites and decreased surface area and spine density. Overall, the histology of Disc1 mutant mouse cortex is reminiscent of the findings in schizophrenia. These results provide further evidence that Disc1 participates in cortical development, including neurogenesis and neuron migration.
Collapse
|
80
|
Taniguchi Y, Young-Pearse T, Sawa A, Kamiya A. In utero electroporation as a tool for genetic manipulation in vivo to study psychiatric disorders: from genes to circuits and behaviors. Neuroscientist 2011; 18:169-79. [PMID: 21551077 DOI: 10.1177/1073858411399925] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Many genetic risk factors for major mental disorders have key roles in brain development. Thus, exploring the roles for these genetic factors for brain development at the molecular, cellular, and neuronal circuit level is crucial for discovering how genetic disturbances affect high brain functions, which ultimately lead to disease pathologies. However, it is a tremendously difficult task, given that most mental disorders have genetic complexities in which many genetic risk factors have multiple roles in different cell types and brain regions over a time-course dependent manner. Furthermore, some genetic risk factors are likely to act epistatically in common molecular pathways. For this reason, a technique for spatial and temporal manipulation of multiple genes is necessary for understanding how genetic disturbances contribute to disease etiology. Here, the authors will review the said technique, in utero electroporation, which investigates the molecular disease pathways in rodent models for major mental disorders. This technique is also useful to examine the effect of genetic risks at the behavioral level. Furthermore, the authors will discuss the recent progress of this technology, such as inducible and cell type-specific targeting, as well as nonepisomal genetic manipulation, which provide further availability of this technique for research on major mental disorders.
Collapse
Affiliation(s)
- Yu Taniguchi
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA
| | | | | | | |
Collapse
|
81
|
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: 4.0] [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
|
82
|
DISC1-dependent switch from progenitor proliferation to migration in the developing cortex. Nature 2011; 473:92-6. [PMID: 21471969 PMCID: PMC3088774 DOI: 10.1038/nature09859] [Citation(s) in RCA: 154] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2010] [Accepted: 01/21/2011] [Indexed: 12/11/2022]
Abstract
Regulatory mechanisms governing the sequence from progenitor cell proliferation to neuronal migration during corticogenesis are poorly understood1–10. Here we report that phosphorylation of DISC1, a major susceptibility factor for several mental disorders, acts as a molecular switch from maintaining proliferation of mitotic progenitor cells to activating migration of postmitotic neurons. Unphosphorylated DISC1 regulates canonical Wnt signaling via an interaction with GSK3β, whereas specific phosphorylation at Serine 710 (S710) triggers the recruitment of Bardet-Biedl-Syndrome (BBS) proteins to the centrosome. In support of this model, loss of BBS1 leads to defects in migration, but not proliferation, while DISC1 knockdown leads to deficits in both. A phospho-dead mutant can only rescue proliferation, while a phospho-mimic mutant rescues exclusively migration defects. These data highlight a dual role for DISC1 in corticogenesis and suggest that phosphorylation of this protein at S710 activates a key developmental switch.
Collapse
|
83
|
Fallin MD, Lasseter VK, Liu Y, Avramopoulos D, McGrath J, Wolyniec PS, Nestadt G, Liang KY, Chen PL, Valle D, Pulver AE. Linkage and association on 8p21.2-p21.1 in schizophrenia. Am J Med Genet B Neuropsychiatr Genet 2011; 156:188-97. [PMID: 21302347 DOI: 10.1002/ajmg.b.31154] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2010] [Accepted: 11/17/2010] [Indexed: 11/07/2022]
Abstract
In the past decade, we and others have consistently reported linkage to a schizophrenia (SZ) susceptibility region on chromosome 8p21. Most recently, in the largest SZ linkage sample to date, a multi-site international collaboration performed a SNP-based linkage scan (~6,000 SNPs; 831 pedigrees; 121 from Johns Hopkins (JHU)), that showed the strongest evidence for linkage in a 1 Mb region of chr 8p21 from rs1561817 to rs9797 (Z(max) = 3.22, P = 0.0004) [Holmans et al. 2009. Mol Psychiatry]. We have investigated this 8p21 peak region further in two ways: first by linkage and family-based association in 106 8p-linked European-Caucasian (EUC) JHU pedigrees using 1,402 SNPs across a 4.4 Mb region surrounding the peak; second, by an independent case-control association study in the genetically more homogeneous Ashkenazim (AJ) (709 cases, 1,547 controls) using 970 SNPs in a further narrowed 2.8 Mb region. Family-based association analyses in EUC pedigrees and case-control analyses in AJ samples reveal significant associations for SNPs in and around DPYSL2 and ADRA1A, candidate genes previously associated with SZ in our work and others. Further, several independent gene expression studies have shown that DPYSL2 is differentially expressed in SZ brains [Beasley et al. 2006. Proteomics 6(11):3414–3425; Edgar et al. 2000. Mol Psychiatry 5(1):85–90; Johnston-Wilson et al. 2000. Mol Psychiatry 5(2):142–149] or in response to psychosis-inducing pharmaceuticals [Iwazaki et al. 2007. Proteomics 7(7):1131–1139; Paulson et al. 2004. Proteomics 4(3):819–825]. Taken together, this work further supports DPYSL2 and the surrounding genomic region as a susceptibility locus for SZ.
Collapse
Affiliation(s)
- M Daniele Fallin
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
84
|
Kleinman JE, Law AJ, Lipska BK, Hyde TM, Ellis JK, Harrison PJ, Weinberger DR. Genetic neuropathology of schizophrenia: new approaches to an old question and new uses for postmortem human brains. Biol Psychiatry 2011; 69:140-5. [PMID: 21183009 PMCID: PMC4351748 DOI: 10.1016/j.biopsych.2010.10.032] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2010] [Revised: 10/06/2010] [Accepted: 10/14/2010] [Indexed: 12/13/2022]
Abstract
Human postmortem brain studies are critical for elucidating the pathophysiology and etiology of schizophrenia and other major mental illnesses. The traditional approach compares patients and control subjects but is potentially confounded by a number of artifacts, including medication, substance misuse, and other secondary effects of illness. Genetic advances now make possible a novel approach that focuses on how allelic variation in risk-associated genes affects expression and function of transcripts and proteins. These questions can be addressed in normal brain, overcoming to some extent the confounding effects of studying brains from subjects with schizophrenia; equally, extension of the studies to include cases also has advantages. Conceptually, the approach may be seen as the neuropathologic counterpart of genetic neuroimaging, representing a potentially powerful intermediate phenotype. For several schizophrenia susceptibility genes, the data show that risk-associated polymorphisms do affect gene expression or the function of the encoded protein; in some instances, expression of downstream or interacting partners of the gene are also altered. A further striking finding is that the implicated transcripts often appear to be enriched in, or specific to, human brain. Some also show enhanced expression in fetal brain. These considerations give unique importance to postmortem human brain tissue in elucidating the genetic mechanisms underlying schizophrenia and probably other neurodevelopmental disorders as well. Studies of this kind can provide clues as to the biological mechanisms of genetic association, especially when carried out in conjunction with experimental studies. Moreover, the data, interpreted judiciously, can strengthen the plausibility of the association itself.
Collapse
Affiliation(s)
- Joel E Kleinman
- Section on Neuropathology, Clinical Brain Disorders Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | |
Collapse
|
85
|
Lu L, Mamiya T, Koseki T, Mouri A, Nabeshima T. Genetic Animal Models of Schizophrenia Related with the Hypothesis of Abnormal Neurodevelopment. Biol Pharm Bull 2011; 34:1358-63. [DOI: 10.1248/bpb.34.1358] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Lingling Lu
- The Academic Frontier Project for Private University, Comparative Cognitive Science Institute, Meijo University
- Department of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Meijo University
| | - Takayoshi Mamiya
- The Academic Frontier Project for Private University, Comparative Cognitive Science Institute, Meijo University
- Department of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Meijo University
- Japanese Drug Organization for Appropriate Use and Research
| | - Takenao Koseki
- The Academic Frontier Project for Private University, Comparative Cognitive Science Institute, Meijo University
- Department of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Meijo University
| | - Akihiro Mouri
- The Academic Frontier Project for Private University, Comparative Cognitive Science Institute, Meijo University
- Department of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Meijo University
- Japanese Drug Organization for Appropriate Use and Research
| | - Toshitaka Nabeshima
- The Academic Frontier Project for Private University, Comparative Cognitive Science Institute, Meijo University
- Department of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Meijo University
- Japanese Drug Organization for Appropriate Use and Research
| |
Collapse
|
86
|
Abstract
BACKGROUND For many years, the prevailing paradigm has stated that in each individual with schizophrenia (SZ) the genetic risk is due to a combination of many genetic variants, individually of small effect. Recent empirical data are prompting a re-evaluation of this polygenic, common disease-common variant (CDCV) model. Evidence includes a lack of the expected strong positive findings from genome-wide association studies and the concurrent discovery of many different mutations that individually strongly predispose to SZ and other psychiatric disorders. This has led some to adopt a mixed model wherein some cases are caused by polygenic mechanisms and some by single mutations. This model runs counter to a substantial body of theoretical literature that had supposedly conclusively rejected Mendelian inheritance with genetic heterogeneity. Here we ask how this discrepancy between theory and data arose and propose a rationalization of the recent evidence base. METHOD In light of recent empirical findings, we reconsider the methods and conclusions of early theoretical analyses and the explicit assumptions underlying them. RESULTS We show that many of these assumptions can now be seen to be false and that the model of genetic heterogeneity is consistent with observed familial recurrence risks, endophenotype studies and other population-wide parameters. CONCLUSIONS We argue for a more biologically consilient mixed model that involves interactions between disease-causing and disease-modifying variants in each individual. We consider the implications of this model for moving SZ research beyond statistical associations to pathogenic mechanisms.
Collapse
Affiliation(s)
- K J Mitchell
- Smurfit Institute of Genetics, Trinity College Dublin, Ireland.
| | | |
Collapse
|
87
|
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: 126] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [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
|
88
|
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: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [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
|
89
|
Fukuda T, Sugita S, Inatome R, Yanagi S. CAMDI, a novel disrupted in schizophrenia 1 (DISC1)-binding protein, is required for radial migration. J Biol Chem 2010; 285:40554-61. [PMID: 20956536 DOI: 10.1074/jbc.m110.179481] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Centrosomes play a crucial role in the directed migration of developing neurons. However, the underlying mechanism is poorly understood. This study has identified a novel disrupted in schizophrenia 1 (DISC1)-interacting protein, named CAMDI after coiled-coil protein associated with myosin II and DISC1, which translocates to the centrosome in a DISC1-dependent manner. Knockdown of CAMDI by shRNA revealed severely impaired radial migration with disoriented centrosomes. A yeast two-hybrid screen identified myosin II as a binding protein of CAMDI. CAMDI interacts preferentially with phosphomyosin II and induces an accumulation of phosphomyosin II at the centrosome in a DISC1-dependent manner. Interestingly, one single nucleotide polymorphism of the CAMDI gene (R828W) is identified, and its gene product was found to reduce the binding ability to phosphomyosin II. Furthermore, mice with overexpression of R828W in neurons exhibit an impaired radial migration. Our findings indicate that CAMDI is required for radial migration probably through DISC1 and myosin II-mediated centrosome positioning during neuronal development.
Collapse
Affiliation(s)
- Toshifumi Fukuda
- Laboratory of Molecular Biochemistry, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan.
| | | | | | | |
Collapse
|
90
|
Moens LN, Ceulemans S, Alaerts M, Van Den Bossche MJA, Lenaerts AS, De Zutter S, Norrback KF, Adolfsson R, Del-Favero J. PCM1 and schizophrenia: a replication study in the Northern Swedish population. Am J Med Genet B Neuropsychiatr Genet 2010; 153B:1240-3. [PMID: 20468070 DOI: 10.1002/ajmg.b.31088] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Previous studies implicated centrosomal dysfunction as a source of various neuropsychiatric disorders, including schizophrenia (SZ). Two recent reports [Gurling et al., 2006; Datta et al., 2008. Mol Psychiatry] described an association between polymorphisms in the PCM1 gene and SZ in a UK/Scottish population. In this study, we aimed to replicate these findings in a Northern Swedish association sample of 486 research subjects with SZ and 512 unrelated control individuals. We genotyped 12 previously described SNP markers and carried out haplotype analyses using the same multi-marker haplotypes previously reported. Though we could not replicate the association with SNPs rs445422 and rs208747, we did observe a significant protective association with intronic SNP rs13276297. Furthermore, we performed a meta-analysis comprising 1,794 SZ patients and 1,553 controls, which confirmed the previously reported association with rs445422 and rs208747. These data provide further evidence that PCM1-though certainly not a major risk factor in the Northern Swedish population-cannot be ruled out as a contributor to SZ risk and/or protection, and deserves further replication in larger populations to elucidate its role in disease etiology.
Collapse
Affiliation(s)
- Lotte N Moens
- Applied Molecular Genomics Group, Department of Molecular Genetics, VIB, Universiteitsplein 1, Antwerp, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
91
|
Kubo KI, Tomita K, Uto A, Kuroda K, Seshadri S, Cohen J, Kaibuchi K, Kamiya A, Nakajima K. Migration defects by DISC1 knockdown in C57BL/6, 129X1/SvJ, and ICR strains via in utero gene transfer and virus-mediated RNAi. Biochem Biophys Res Commun 2010; 400:631-7. [PMID: 20807500 DOI: 10.1016/j.bbrc.2010.08.117] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2010] [Accepted: 08/26/2010] [Indexed: 02/01/2023]
Abstract
Disrupted-in-Schizophrenia 1 (DISC1) is a promising genetic risk factor for major mental disorders. Many groups repeatedly reported a role for DISC1 in brain development in various strains of mice and rats by using RNA interference (RNAi) approach. Nonetheless, due to the complexity of its molecular disposition, such as many splice variants and a spontaneous deletion in a coding exon of the DISC1 gene in some mouse strains, there have been debates on the interpretation on these published data. Thus, in this study, we address this question by DISC1 knockdown via short-hairpin RNAs (shRNAs) against several distinct target sequences with more than one delivery methodologies into several mouse strains, including C57BL/6, ICR, and 129X1/SvJ. Here, we show that DISC1 knockdown by in utero electroporation of shRNA against exons 2, 6, and 10 consistently results in neuronal migration defects in the developing cerebral cortex, which are successfully rescued by co-expression of full-length DISC1. Furthermore, lentivirus-mediated shRNA also led to migration defects, which is consistent with two other methodologies already published, such as plasmid-mediated and retrovirus-mediated ones. The previous study by Song's group also reported that, in the adult hippocampus, the phenotype elicited by DISC1 knockdown with shRNA targeting exon 2 was consistently seen in both C57BL/6 and 129S6 mice. Taken together, we propose that some of DISC1 isoforms that are feasible to be knocked down by shRNAs to exon 2, 6, and 10 of the DISC1 gene play a key role for neuronal migration commonly in various mouse strains and rats.
Collapse
Affiliation(s)
- Ken-Ichiro Kubo
- Department of Anatomy, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
92
|
Hattori T, Shimizu S, Koyama Y, Yamada K, Kuwahara R, Kumamoto N, Matsuzaki S, Ito A, Katayama T, Tohyama M. DISC1 regulates cell-cell adhesion, cell-matrix adhesion and neurite outgrowth. Mol Psychiatry 2010; 15:778, 798-809. [PMID: 20479754 DOI: 10.1038/mp.2010.60] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Disrupted-in-schizophrenia 1 (DISC1) is a promising susceptibility gene for major mental illness. Recent studies have implicated DISC1 in key neurodevelopmental processes, including neurite outgrowth, neuronal migration and proliferation. Here, we report that DISC1 regulates cell-cell and cell-matrix adhesion and neurite outgrowth. DISC1 overexpression increased expression of the adherence junction protein N-cadherin and enhanced cell-cell adhesion. The increased N-cadherin accumulated in the areas of cell-cell contact. DISC1 overexpression also enhanced cell-matrix adhesion by inducing expression of beta1-integrin protein. In the presence of nerve growth factor (NGF), DISC1 overexpression increased beta1-integrin expression at the cell membrane and growth cone. NGF-induced neurite extension was enhanced by DISC1, and anti-beta1-integrin antibody reduced the neurite outgrowth of DISC1-overexpressing cells to the control level. Furthermore, DISC1 also regulated N-cadherin and beta1-integrin expression at the cell membrane in primary neurons. We conclude that DISC1 regulates cell-cell adhesion and cell-matrix adhesion by regulating the expression of adhesion molecules.
Collapse
Affiliation(s)
- T Hattori
- Department of Molecular Neuropsychiatry, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
93
|
Datta SR, McQuillin A, Rizig M, Blaveri E, Thirumalai S, Kalsi G, Lawrence J, Bass NJ, Puri V, Choudhury K, Pimm J, Crombie C, Fraser G, Walker N, Curtis D, Zvelebil M, Pereira A, Kandaswamy R, St Clair D, Gurling HMD. A threonine to isoleucine missense mutation in the pericentriolar material 1 gene is strongly associated with schizophrenia. Mol Psychiatry 2010; 15:615-28. [PMID: 19048012 DOI: 10.1038/mp.2008.128] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Markers at the pericentriolar material 1 gene (PCM1) have shown genetic association with schizophrenia in both a University College London (UCL) and a USA-based case-control sample. In this paper we report a statistically significant replication of the PCM1 association in a large Scottish case-control sample from Aberdeen. Resequencing of the genomic DNA from research volunteers who had inherited haplotypes associated with schizophrenia showed a threonine to isoleucine missense mutation in exon 24 which was likely to change the structure and function of PCM1 (rs370429). This mutation was found only as a heterozygote in 98 schizophrenic research subjects and controls out of 2246 case and control research subjects. Among the 98 carriers of rs370429, 67 were affected with schizophrenia. The same alleles and haplotypes were associated with schizophrenia in both the London and Aberdeen samples. Another potential aetiological base pair change in PCM1 was rs445422, which altered a splice site signal. A further mutation, rs208747, was shown by electrophoretic mobility shift assays to create or destroy a promoter transcription factor site. Five further non-synonymous changes in exons were also found. Genotyping of the new variants discovered in the UCL case-control sample strengthened the evidence for allelic and haplotypic association (P=0.02-0.0002). Given the number and identity of the haplotypes associated with schizophrenia, further aetiological base pair changes must exist within and around the PCM1 gene. PCM1 protein has been shown to interact directly with the disrupted-in-schizophrenia 1 (DISC1) protein, Bardet-Biedl syndrome 4, and Huntingtin-associated protein 1, and is important in neuronal cell growth. In a separate study we found that clozapine but not haloperidol downregulated PCM1 expression in the mouse brain. We hypothesize that mutant PCM1 may be responsible for causing a subtype of schizophrenia through abnormal cell division and abnormal regeneration in dividing cells in the central nervous system. This is supported by our previous finding of orbitofrontal volumetric deficits in PCM1-associated schizophrenia patients as opposed to temporal pole deficits in non-PCM1-associated schizophrenia patients. Caution needs to be exercised in interpreting the actual biological effects of the mutations we have found without further cell biology. However, the DNA changes we have found deserve widespread genotyping in multiple case-control populations.
Collapse
Affiliation(s)
- S R Datta
- Molecular Psychiatry Laboratory, Research Department of Mental Health Sciences, University College London Medical School, Windeyer Institute of Medical Sciences, London, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
94
|
DISC1 regulates primary cilia that display specific dopamine receptors. PLoS One 2010; 5:e10902. [PMID: 20531939 PMCID: PMC2878344 DOI: 10.1371/journal.pone.0010902] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2009] [Accepted: 05/10/2010] [Indexed: 12/11/2022] Open
Abstract
Background Mutations in the DISC1 gene are strongly associated with major psychiatric syndromes such as schizophrenia. DISC1 encodes a cytoplasmic protein with many potential interaction partners, but its cellular functions remain poorly understood. We identified a role of DISC1 in the cell biology of primary cilia that display disease-relevant dopamine receptors. Methodology/Principal Findings A GFP-tagged DISC1 construct expressed in NIH3T3 cells and rat striatal neurons localized near the base of primary cilia. RNAi-mediated knockdown of endogenous DISC1 resulted in a marked reduction in the number of cells expressing a primary cilium. FLAG-tagged versions of the cloned human D1, D2 and D5 dopamine receptors concentrated highly on the ciliary surface, and this reflects a specific targeting mechanism specific because D3 and D4 receptors localized to the plasma membrane but were not concentrated on cilia. Conclusions/Significance These results identify a role of DISC1 in regulating the formation and/or maintenance of primary cilia, and establish subtype-specific targeting of dopamine receptors to the ciliary surface. Our findings provide new insight to receptor cell biology and suggest a relationship between DISC1 and neural dopamine signaling.
Collapse
|
95
|
Hayashi-Takagi A, Sawa A. Disturbed synaptic connectivity in schizophrenia: convergence of genetic risk factors during neurodevelopment. Brain Res Bull 2010; 83:140-6. [PMID: 20433911 DOI: 10.1016/j.brainresbull.2010.04.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2009] [Revised: 02/07/2010] [Accepted: 04/19/2010] [Indexed: 12/13/2022]
Abstract
The pathological mechanisms underlying schizophrenia are unclear. Although genetic susceptibility factors for schizophrenia likely influence neurodevelopmental processes, the onset of the disease is in adolescence and young adulthood. Here we review recent literatures implicating neurodevelopmental deficits in schizophrenia and discuss how genetic factors are involved in the processes toward onset of the disease. We emphasize the importance of postnatal glutamate synapse development in the pathology of the disorder. These genetic risk factors contribute to the process possibly in a synergistic manner. The notion of signal pathways involving more than one genetic factor is in accord with the multifactorial nature of schizophrenia.
Collapse
Affiliation(s)
- Akiko Hayashi-Takagi
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States.
| | | |
Collapse
|
96
|
Eastwood SL, Walker M, Hyde TM, Kleinman JE, Harrison PJ. The DISC1 Ser704Cys substitution affects centrosomal localization of its binding partner PCM1 in glia in human brain. Hum Mol Genet 2010; 19:2487-96. [PMID: 20360304 DOI: 10.1093/hmg/ddq130] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Disrupted-in-schizophrenia 1 (DISC1) has been genetically associated with schizophrenia, and with brain phenotypes including grey matter volume and working memory performance. However, the molecular and cellular basis for these associations remains to be elucidated. One potential mechanism may be via an altered interaction of DISC1 with its binding partners. In this context, we previously demonstrated that one DISC1 variant, Leu607Phe, influenced the extent of centrosomal localization of pericentriolar material 1 (PCM1) in SH-SY5Y cells. The current study extends this work to human brain, and includes another DISC1 coding variant, Ser704Cys. Using immunohistochemistry, we first characterized the distribution of PCM1 in human superior temporal gyrus (STG). PCM1 immunoreactivity was localized to the centrosome in glia, but not in neurons, which showed widespread immunoreactivity. We quantified centrosomal PCM1 immunoreactivity in STG glia of 81 controls and 67 subjects with schizophrenia, genotyped for the two polymorphisms. Centrosomal PCM1 immunoreactive area was smaller in Cys704 carriers than in Ser704 homozygotes, with a similar trend in Phe607 homozygotes compared with Leu607 carriers, replicating the finding in SH-SY5Y cells. No differences were seen between controls and subjects with schizophrenia. These findings confirm in vivo that DISC1 coding variants modulate centrosomal PCM1 localization, highlight a role for DISC1 in glial function and provide a possible cellular mechanism contributing to the association of these DISC1 variants with psychiatric phenotypes. Whether this influence of DISC1 genotype extends to other centrosomal proteins and DISC1 binding partners remains to be determined.
Collapse
Affiliation(s)
- Sharon L Eastwood
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford OX3 7JX, UK.
| | | | | | | | | |
Collapse
|
97
|
Niwa M, Kamiya A, Murai R, Kubo KI, Gruber AJ, Tomita K, Lu L, Tomisato S, Jaaro-Peled H, Seshadri S, Hiyama H, Huang B, Kohda K, Noda Y, O'Donnell P, Nakajima K, Sawa A, Nabeshima T. Knockdown of DISC1 by in utero gene transfer disturbs postnatal dopaminergic maturation in the frontal cortex and leads to adult behavioral deficits. Neuron 2010; 65:480-9. [PMID: 20188653 DOI: 10.1016/j.neuron.2010.01.019] [Citation(s) in RCA: 236] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/14/2010] [Indexed: 01/28/2023]
Abstract
Adult brain function and behavior are influenced by neuronal network formation during development. Genetic susceptibility factors for adult psychiatric illnesses, such as Neuregulin-1 and Disrupted-in-Schizophrenia-1 (DISC1), influence adult high brain functions, including cognition and information processing. These factors have roles during neurodevelopment and are likely to cooperate, forming pathways or "signalosomes." Here we report the potential to generate an animal model via in utero gene transfer in order to address an important question of how nonlethal deficits in early development may affect postnatal brain maturation and high brain functions in adulthood, which are impaired in various psychiatric illnesses such as schizophrenia. We show that transient knockdown of DISC1 in the pre- and perinatal stages, specifically in a lineage of pyramidal neurons mainly in the prefrontal cortex, leads to selective abnormalities in postnatal mesocortical dopaminergic maturation and behavioral abnormalities associated with disturbed cortical neurocircuitry after puberty.
Collapse
Affiliation(s)
- Minae Niwa
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
98
|
Ge X, Frank CL, Calderon de Anda F, Tsai LH. Hook3 interacts with PCM1 to regulate pericentriolar material assembly and the timing of neurogenesis. Neuron 2010; 65:191-203. [PMID: 20152126 DOI: 10.1016/j.neuron.2010.01.011] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/30/2009] [Indexed: 01/22/2023]
Abstract
Centrosome functions are important in multiple brain developmental processes. Proper functioning of the centrosome relies on assembly of protein components into the pericentriolar material. This dynamic assembly is mediated by the trafficking of pericentriolar satellites, which are comprised of centrosomal proteins. Here we demonstrate that trafficking of pericentriolar satellites requires the interaction between Hook3 and Pericentriolar Material 1 (PCM1). Hook3, previously shown to link the centrosome and the nucleus in C. elegans, is recruited to pericentriolar satellites through interaction with PCM1, a protein associated with schizophrenia. Disruption of the Hook3-PCM1 interaction in vivo impairs interkinetic nuclear migration, a featured behavior of embryonic neural progenitors. This in turn leads to overproduction of neurons and premature depletion of the neural progenitor pool in the developing neocortex. These results underscore the importance of centrosomal assembly in neurogenesis and provide potential insights into the etiology of brain developmental diseases related to the centrosome dysfunction.
Collapse
Affiliation(s)
- Xuecai Ge
- Picower Institute for Learning and Memory, Department of Brain and Cognitive, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Building 46, Room 4235A, Cambridge, MA 02139, USA
| | | | | | | |
Collapse
|
99
|
Han YG, Alvarez-Buylla A. Role of primary cilia in brain development and cancer. Curr Opin Neurobiol 2010; 20:58-67. [PMID: 20080044 DOI: 10.1016/j.conb.2009.12.002] [Citation(s) in RCA: 117] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2009] [Revised: 12/02/2009] [Accepted: 12/03/2009] [Indexed: 12/11/2022]
Abstract
The primary cilium, a hair-like extension from a cell's surface, acts as a sensory organelle to receive signals that regulate cellular behavior and physiology. Like most mammalian cells, neural progenitors and neurons have primary cilia. Recent studies show that this tiny projection plays important roles in brain development and diseases. Ciliary mutant mice show defects in brain patterning, progenitor proliferation, and specification of adult neural stem cells. Primary cilia also have dual opposing functions in the development of brain tumors. Ciliary defects are associated with genetic syndromes that frequently have neurological symptoms. Understanding the multifaceted roles that primary cilia have in brain development will provide important insights into the mechanism of brain development and diseases.
Collapse
Affiliation(s)
- Young-Goo Han
- Department of Neurological Surgery, The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, 505 Parnassus Ave., San Francisco, CA 94143-0112, USA
| | | |
Collapse
|
100
|
Abstract
Pericentrin is an integral component of the centrosome that serves as a multifunctional scaffold for anchoring numerous proteins and protein complexes. Through these interactions, pericentrin contributes to a diversity of fundamental cellular processes. Recent studies link pericentrin to a growing list of human disorders. Studies on pericentrin at the cellular, molecular, and, more recently, organismal level, provide a platform for generating models to elucidate the etiology of these disorders. Although the complexity of phenotypes associated with pericentrin-mediated disorders is somewhat daunting, insights into the cellular basis of disease are beginning to come into focus. In this review, we focus on human conditions associated with loss or elevation of pericentrin and propose cellular and molecular models that might explain them.
Collapse
Affiliation(s)
- Benedicte Delaval
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | |
Collapse
|