1
|
Zakaria WNA, Sasongko TH, Al-Rahbi B, Al-Sowayan N, Ahmad AH, Zakaria R, Ahmi A, Othman Z. Gene and schizophrenia in the pregenome and postgenome-wide association studies era: a bibliometric analysis and network visualization. Psychiatr Genet 2023; 33:37-49. [PMID: 36825838 DOI: 10.1097/ypg.0000000000000336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
This study aimed to perform a bibliometric analysis on genetic studies in schizophrenia in the pregenome-wide association studies (GWAS) and post-GWAS era. We searched the literature on genes and schizophrenia using the Scopus database. The documents increased with time, especially after the human genome project and International HapMap Project, with the highest citation in 2008. The top occurrence author keywords were discovered to be different in the pre-GWAS and post-GWAS eras, reflecting the progress of genetic studies connected to schizophrenia. Emerging keywords highlighted a trend towards an application of precision medicine, showing an interplay of environmental exposures as well as genetic factors in schizophrenia pathogenesis, progression, and response to therapy. In conclusion, the gene and schizophrenia literature has grown rapidly after the human genome project, and the temporal variation in the author keywords pattern reflects the trend of genetic studies related to schizophrenia in the pre-GWAS and post-GWAS era.
Collapse
Affiliation(s)
- Wan Nur Amalina Zakaria
- Human Genome Centre, School of Medical Sciences, Universiti Sains Malaysia Health Campus, Kota Bharu, Kelantan, Malaysia
| | - Teguh Haryo Sasongko
- Department of Physiology, School of Medicine, and Institute for Research, Development and Innovation, International Medical University, Kuala Lumpur, Malaysia
| | | | - Noorah Al-Sowayan
- Department of Biology, College of Science, Qassim University, Saudi Arabia
| | - Asma Hayati Ahmad
- Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Rahimah Zakaria
- Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Aidi Ahmi
- Tunku Puteri Intan Safinaz School of Accountancy, Universiti Utara Malaysia 06010 UUM Sintok, Kedah
| | - Zahiruddin Othman
- Department of Psychiatry, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| |
Collapse
|
2
|
Mapping the expression of an ANK3 isoform associated with bipolar disorder in the human brain. Transl Psychiatry 2022; 12:45. [PMID: 35091539 PMCID: PMC8799726 DOI: 10.1038/s41398-022-01784-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 12/09/2021] [Accepted: 01/04/2022] [Indexed: 11/08/2022] Open
Abstract
The gene ankyrin-3 (ANK3) has been consistently associated with bipolar disorder (BD) in several genome-wide association studies (GWAS). The exact molecular mechanisms underlying this genetic association remain unknown. The discovery of a loss-of-function variant (rs41283526*G) in an alternatively spliced exon (ENSE00001786716) with a protective effect, suggested that elevated expression of this particular isoform could be a risk factor for developing the disorder. We developed a novel approach for measuring the expression level of all splice forms at a challenging genetic locus using a combination of droplet digital PCR and high-throughput sequencing of indexed PCR amplicons. The combined method was performed on a large collection of 568 postmortem brain samples of BD and SCZ cases and controls. We also studied the expression of the splice forms in a child-development cohort of 41 healthy males. We found that our approach can quantify the splice forms in brain samples, although with less precision than ddPCR. We detected highly significant differences in expression of splice forms and transcription start sites between brain regions, notably with higher expression of the BD-associated isoform in the corpus callosum compared to frontal tissue (mean fold change = 1.80, p < 1e-4). Although the patients in our sample expressed the BD-associated splice form at a similar level to controls, adolescents in our child-development cohort had a clearly higher expression level than younger children (mean fold change = 1.97, p = 5e-3). These results suggest that this ANK3 splice form may play a role in the myelin maturation of the human brain.
Collapse
|
3
|
Gupta R, Bigdeli TB, Buckley PF, Fanous AH. Genetics of Schizophrenia and Bipolar Disorder: Potential Clinical Applications. Psychiatr Ann 2021. [DOI: 10.3928/00485713-20210310-01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
4
|
The "missing heritability"-Problem in psychiatry: Is the interaction of genetics, epigenetics and transposable elements a potential solution? Neurosci Biobehav Rev 2021; 126:23-42. [PMID: 33757815 DOI: 10.1016/j.neubiorev.2021.03.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/15/2021] [Accepted: 03/17/2021] [Indexed: 02/07/2023]
Abstract
Psychiatric disorders exhibit an enormous burden on the health care systems worldwide accounting for around one-third of years lost due to disability among adults. Their etiology is largely unknown and diagnostic classification is based on symptomatology and course of illness and not on objective biomarkers. Most psychiatric disorders are moderately to highly heritable. However, it is still unknown what mechanisms may explain the discrepancy between heritability estimates and the present data from genetic analysis. In addition to genetic differences also epigenetic modifications are considered as potentially relevant in the transfer of susceptibility to psychiatric diseases. Though, whether or not epigenetic alterations can be inherited for many generations is highly controversial. In the present article, we will critically summarize both the genetic findings and the results from epigenetic analyses, including also those of noncoding RNAs. We will argue that one possible solution to the "missing heritability" problem in psychiatry is a potential role of retrotransposons, the exploration of which is presently only in its beginnings.
Collapse
|
5
|
Poltavskaya EG, Fedorenko OY, Vyalova NM, Kornetova EG, Bokhan NA, Loonen AJM, Ivanova SA. Genetic polymorphisms of PIP5K2A and course of schizophrenia. BMC MEDICAL GENETICS 2020; 21:171. [PMID: 33092542 PMCID: PMC7579868 DOI: 10.1186/s12881-020-01107-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 08/11/2020] [Indexed: 11/30/2022]
Abstract
Background Schizophrenia is a severe highly heritable mental disorder. The clinical heterogeneity of schizophrenia is expressed in the difference in the leading symptoms and course of the disease. Identifying the genetic variants that affect clinical heterogeneity may ultimately reveal the genetic basis of the features of schizophrenia and suggest novel treatment targets. PIP5K2A (Phosphatidylinositol-4-Phosphate 5-Kinase Type II Alpha) has been investigated as a potential susceptibility gene for schizophrenia. Methods In this work, we studied the possible association between eleven polymorphic variants of PIP5K2A and the clinical features of schizophrenia in a population of 384 white Siberian patients with schizophrenia. Genotyping was carried out on QuantStudio 5 Real-Time PCR System with a TaqMan Validate SNP Genotyping Assay (Applied Biosystems, USA). Results PIP5K2A rs8341 (χ2 = 6.559, p = 0.038) and rs946961 (χ2 = 5.976, p = 0.049) showed significant association with course of schizophrenia (continuous or episodic). The rs8341*CT (OR = 1.63, 95% CI: 1.04–2.54) and rs946961*CC (OR = 5.17, 95% CI: 1.20–22.21) genotypes were associated with a continuous type of course, while the rs8341*TT genotype (OR = 0.53, 95% CI: 0.29–0.97) was associated with an episodic type of course of schizophrenia. Therefore rs8341*TT genotype presumably has protective effect against the more severe continuous course of schizophrenia compared to the episodic one. Conclusions Our experimental data confirm that PIP5K2A is a genetic factor influencing the type of course of schizophrenia in Siberian population. Disturbances in the phosphatidylinositol pathways may be a possible reason for the transition to a more severe continuous course of schizophrenia.
Collapse
Affiliation(s)
- Evgeniya G Poltavskaya
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Aleutskaya str., 4, Tomsk, Russian Federation, 634014.
| | - Olga Yu Fedorenko
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Aleutskaya str., 4, Tomsk, Russian Federation, 634014.,National Research Tomsk Polytechnic University, Tomsk, Russian Federation
| | - Natalya M Vyalova
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Aleutskaya str., 4, Tomsk, Russian Federation, 634014
| | - Elena G Kornetova
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Aleutskaya str., 4, Tomsk, Russian Federation, 634014
| | - Nikolay A Bokhan
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Aleutskaya str., 4, Tomsk, Russian Federation, 634014.,National Research Tomsk State University, Tomsk, Russian Federation.,Siberian State Medical University Hospital, Moscowsky Trakt, 2, Tomsk, Russian Federation
| | - Anton J M Loonen
- Groningen Research Institute of Pharmacy, PharmacoTherapy, Epidemiology & Economics, University of Groningen, Antonius Deusinglaan 1, 9713, AV, Groningen, The Netherlands.,GGZ Westelijk Noord-Brabant, Hoofdlaan 8, 4661 AA, Halsteren, The Netherlands
| | - Svetlana A Ivanova
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Aleutskaya str., 4, Tomsk, Russian Federation, 634014.,National Research Tomsk Polytechnic University, Tomsk, Russian Federation.,Siberian State Medical University Hospital, Moscowsky Trakt, 2, Tomsk, Russian Federation
| |
Collapse
|
6
|
Charney AW, Mullins N, Park YJ, Xu J. On the diagnostic and neurobiological origins of bipolar disorder. Transl Psychiatry 2020; 10:118. [PMID: 32327632 PMCID: PMC7181677 DOI: 10.1038/s41398-020-0796-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 03/11/2020] [Accepted: 04/01/2020] [Indexed: 11/22/2022] Open
Abstract
Psychiatry is constructed around a taxonomy of several hundred diagnoses differentiated by nuances in the timing, co-occurrence, and severity of symptoms. Bipolar disorder (BD) is notable among these diagnoses for manic, depressive, and psychotic symptoms all being core features. Here, we trace current understanding of the neurobiological origins of BD and related diagnoses. To provide context, we begin by exploring the historical origins of psychiatric taxonomy. We then illustrate how key discoveries in pharmacology and neuroscience gave rise to a generation of neurobiological hypotheses about the origins of these disorders that facilitated therapeutic innovation but failed to explain disease pathogenesis. Lastly, we examine the extent to which genetics has succeeded in filling this void and contributing to the construction of an objective classification of psychiatric disturbance.
Collapse
Affiliation(s)
- Alexander W Charney
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA.
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA.
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA.
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA.
- Mental Illness Research, Education, and Clinical Center (VISN 2 South), James J. Peters Veterans Affairs Medical Center, Bronx, NY, 10468, USA.
| | - Niamh Mullins
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - You Jeong Park
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Jonathan Xu
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| |
Collapse
|
7
|
Huang H, Li Y, Liang H, Wu CO. Decomposition feature selection with applications in detecting correlated biomarkers of bipolar disorders. Stat Med 2019; 38:4574-4582. [PMID: 31304613 DOI: 10.1002/sim.8317] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 04/24/2019] [Accepted: 06/14/2019] [Indexed: 12/16/2022]
Abstract
Feature selection is an important initial step of exploratory analysis in biomedical studies. Its main objective is to eliminate the covariates that are uncorrelated with the outcome. For highly correlated covariates, traditional feature selection methods, such as the Lasso, tend to select one of them and eliminate the others, although some of the eliminated ones are still scientifically valuable. To alleviate this drawback, we propose a feature selection method based on covariate space decomposition, referred herein as the "Decomposition Feature Selection" (DFS), and show that this method can lead to scientifically meaningful results in studies with correlated high dimensional data. The DFS consists of two steps: (i) decomposing the covariate space into disjoint subsets such that each of the subsets contains only uncorrelated covariates and (ii) identifying significant predictors by traditional feature selection within each covariate subset. We demonstrate through simulation studies that the DFS has superior practical performance over the Lasso type methods when multiple highly correlated covariates need to be retained. Application of the DFS is demonstrated through a study of bipolar disorders with correlated biomarkers.
Collapse
Affiliation(s)
- Hailin Huang
- Department of Statistics, The George Washington University, Washington, District of Columbia
| | - Yuanzhang Li
- Division of Preventive Medicine, Walter Reed Army Institute of Research, Washington, District of Columbia
| | - Hua Liang
- Department of Statistics, The George Washington University, Washington, District of Columbia
| | - Colin O Wu
- Office of Biostatistics Research, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
8
|
Schanze I, Bunt J, Lim JWC, Schanze D, Dean RJ, Alders M, Blanchet P, Attié-Bitach T, Berland S, Boogert S, Boppudi S, Bridges CJ, Cho MT, Dobyns WB, Donnai D, Douglas J, Earl DL, Edwards TJ, Faivre L, Fregeau B, Genevieve D, Gérard M, Gatinois V, Holder-Espinasse M, Huth SF, Izumi K, Kerr B, Lacaze E, Lakeman P, Mahida S, Mirzaa GM, Morgan SM, Nowak C, Peeters H, Petit F, Pilz DT, Puechberty J, Reinstein E, Rivière JB, Santani AB, Schneider A, Sherr EH, Smith-Hicks C, Wieland I, Zackai E, Zhao X, Gronostajski RM, Zenker M, Richards LJ. NFIB Haploinsufficiency Is Associated with Intellectual Disability and Macrocephaly. Am J Hum Genet 2018; 103:752-768. [PMID: 30388402 PMCID: PMC6218805 DOI: 10.1016/j.ajhg.2018.10.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 10/03/2018] [Indexed: 12/19/2022] Open
Abstract
The nuclear factor I (NFI) family of transcription factors play an important role in normal development of multiple organs. Three NFI family members are highly expressed in the brain, and deletions or sequence variants in two of these, NFIA and NFIX, have been associated with intellectual disability (ID) and brain malformations. NFIB, however, has not previously been implicated in human disease. Here, we present a cohort of 18 individuals with mild ID and behavioral issues who are haploinsufficient for NFIB. Ten individuals harbored overlapping microdeletions of the chromosomal 9p23-p22.2 region, ranging in size from 225 kb to 4.3 Mb. Five additional subjects had point sequence variations creating a premature termination codon, and three subjects harbored single-nucleotide variations resulting in an inactive protein as determined using an in vitro reporter assay. All individuals presented with additional variable neurodevelopmental phenotypes, including muscular hypotonia, motor and speech delay, attention deficit disorder, autism spectrum disorder, and behavioral abnormalities. While structural brain anomalies, including dysgenesis of corpus callosum, were variable, individuals most frequently presented with macrocephaly. To determine whether macrocephaly could be a functional consequence of NFIB disruption, we analyzed a cortex-specific Nfib conditional knockout mouse model, which is postnatally viable. Utilizing magnetic resonance imaging and histology, we demonstrate that Nfib conditional knockout mice have enlargement of the cerebral cortex but preservation of overall brain structure and interhemispheric connectivity. Based on our findings, we propose that haploinsufficiency of NFIB causes ID with macrocephaly.
Collapse
Affiliation(s)
- Ina Schanze
- Institute of Human Genetics, University Hospital Magdeburg, Otto-von-Guericke University, Magdeburg 39120, Germany
| | - Jens Bunt
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia.
| | - Jonathan W C Lim
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Denny Schanze
- Institute of Human Genetics, University Hospital Magdeburg, Otto-von-Guericke University, Magdeburg 39120, Germany
| | - Ryan J Dean
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Marielle Alders
- Department of Clinical Genetics, Academic Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands
| | - Patricia Blanchet
- INSERM U1183, Département de Génétique Médicale, Maladies Rares et Médecine Personnalisée, Génétique clinique, CHU Montpellier, Université Montpellier, Centre de référence anomalies du développement SORO, Montpellier 34295, France
| | - Tania Attié-Bitach
- INSERM U1163, Laboratory of Embryology and Genetics of Congenital Malformations, Paris Descartes University, Sorbonne Paris Cité and Imagine Institute, Paris 75015, France
| | - Siren Berland
- Department of Medical Genetics, Haukeland University Hospital, Bergen 5021, Norway
| | - Steven Boogert
- Institute of Human Genetics, University Hospital Magdeburg, Otto-von-Guericke University, Magdeburg 39120, Germany
| | - Sangamitra Boppudi
- Institute of Human Genetics, University Hospital Magdeburg, Otto-von-Guericke University, Magdeburg 39120, Germany
| | - Caitlin J Bridges
- Institute of Human Genetics, University Hospital Magdeburg, Otto-von-Guericke University, Magdeburg 39120, Germany
| | | | - William B Dobyns
- Department of Pediatrics (Genetics), University of Washington and Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Dian Donnai
- Manchester Centre for Genomic Medicine, Manchester Academic Health Science Centre, Central Manchester University Hospitals NHS Foundation Trust; Division of Evolution and Genomic Sciences School of Biological Sciences, and University of Manchester, Manchester M13 9WL, UK
| | - Jessica Douglas
- Boston Children's Hospital - The Feingold Center, Waltham, MA 02115, USA
| | - Dawn L Earl
- Division of Genetic Medicine, Seattle Children's Hospital, Seattle, WA 98105, USA
| | - Timothy J Edwards
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia; The Faculty of Medicine Brisbane, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Laurence Faivre
- UMR1231, Génétique des Anomalies du Développement, Université de Bourgogne, Dijon 21079, France; Centre de Génétique et Centre de Référence Anomalies du Développement et Syndromes Malformatifs de l'Interrégion Est et FHU TRANSLAD, Centre Hospitalier Universitaire Dijon, Dijon 21079, France
| | - Brieana Fregeau
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - David Genevieve
- INSERM U1183, Département de Génétique Médicale, Maladies Rares et Médecine Personnalisée, Génétique clinique, CHU Montpellier, Université Montpellier, Centre de référence anomalies du développement SORO, Montpellier 34295, France
| | - Marion Gérard
- Service de Génétique, CHU de Caen - Hôpital Clémenceau, Caen Cedex 14000, France
| | - Vincent Gatinois
- INSERM U1183, Département de Génétique Médicale, Maladies Rares et Médecine Personnalisée, Génétique clinique, CHU Montpellier, Université Montpellier, Centre de référence anomalies du développement SORO, Montpellier 34295, France
| | - Muriel Holder-Espinasse
- Service de Génétique Clinique, Hôpital Jeanne de Flandre, CHU Lille, Lille 59000, France; Department of Clinical Genetics, Guy's Hospital, London SE1 9RT, UK
| | - Samuel F Huth
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Kosuke Izumi
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Division of Genetics, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Bronwyn Kerr
- Manchester Centre for Genomic Medicine, Manchester Academic Health Science Centre, Central Manchester University Hospitals NHS Foundation Trust; Division of Evolution and Genomic Sciences School of Biological Sciences, and University of Manchester, Manchester M13 9WL, UK
| | - Elodie Lacaze
- Department of genetics, Le Havre Hospital, 76600 Le Havre, France
| | - Phillis Lakeman
- Department of Clinical Genetics, Academic Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands
| | - Sonal Mahida
- Department of Neurogenetics, Kennedy Krieger Institute, Baltimore, MD 21205, USA
| | - Ghayda M Mirzaa
- Department of Pediatrics (Genetics), University of Washington and Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Sian M Morgan
- All Wales Genetics Laboratory, Institute of Medical Genetics, University Hospital of Wales, Cardiff CF14 4XW, UK
| | - Catherine Nowak
- Boston Children's Hospital - The Feingold Center, Waltham, MA 02115, USA
| | - Hilde Peeters
- Center for Human Genetics, University Hospital Leuven, KU Leuven, Leuven 3000, Belgium
| | - Florence Petit
- Service de Génétique Clinique, Hôpital Jeanne de Flandre, CHU Lille, Lille 59000, France
| | - Daniela T Pilz
- West of Scotland Genetics Service, Queen Elizabeth University Hospital, Glasgow G51 4TF, UK
| | - Jacques Puechberty
- INSERM U1183, Département de Génétique Médicale, Maladies Rares et Médecine Personnalisée, Génétique clinique, CHU Montpellier, Université Montpellier, Centre de référence anomalies du développement SORO, Montpellier 34295, France
| | - Eyal Reinstein
- Medical Genetics Institute, Meir Medical Center, Kfar-Saba 4428164, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Jean-Baptiste Rivière
- UMR1231, Génétique des Anomalies du Développement, Université de Bourgogne, Dijon 21079, France; Centre de Génétique et Centre de Référence Anomalies du Développement et Syndromes Malformatifs de l'Interrégion Est et FHU TRANSLAD, Centre Hospitalier Universitaire Dijon, Dijon 21079, France; Child Health and Human Development Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Avni B Santani
- Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Anouck Schneider
- INSERM U1183, Département de Génétique Médicale, Maladies Rares et Médecine Personnalisée, Génétique clinique, CHU Montpellier, Université Montpellier, Centre de référence anomalies du développement SORO, Montpellier 34295, France
| | - Elliott H Sherr
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| | | | - Ilse Wieland
- Institute of Human Genetics, University Hospital Magdeburg, Otto-von-Guericke University, Magdeburg 39120, Germany
| | - Elaine Zackai
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xiaonan Zhao
- Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Richard M Gronostajski
- Department of Biochemistry, Program in Genetics, Genomics and Bioinformatics, Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Martin Zenker
- Institute of Human Genetics, University Hospital Magdeburg, Otto-von-Guericke University, Magdeburg 39120, Germany.
| | - Linda J Richards
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia; School of Biomedical Sciences, The Faculty of Medicine Brisbane, The University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
9
|
Identification of rare nonsynonymous variants in SYNE1/CPG2 in bipolar affective disorder. Psychiatr Genet 2018; 27:81-88. [PMID: 28178086 DOI: 10.1097/ypg.0000000000000166] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Bipolar affective disorder (BPD) is a severe mood disorder with a prevalence of ∼1.5% in the population. The pathogenesis of BPD is poorly understood; however, a strong heritable component has been identified. Previous genome-wide association studies have indicated a region on 6q25, coding for the SYNE1 gene, which increases disease susceptibility. SYNE1 encodes the synaptic nuclear envelope protein-1, nesprin-1. A brain-specific splice variant of SYNE1, CPG2 encoding candidate plasticity gene 2, has been identified. The intronic single-nucleotide polymorphism with the strongest genome-wide significant association in BPD, rs9371601, is present in both SYNE1 and CPG2. METHODS We screened 937 BPD samples for genetic variation in SYNE1 exons 14-33, which covers the CPG2 region, using high-resolution melt analysis. In addition, we screened two regions of increased transcriptional activity, one of them proposed to be the CPG2 promoter region. RESULTS AND CONCLUSION We identified six nonsynonymous and six synonymous variants. We genotyped three rare nonsynonymous variants, rs374866393, rs148346599 and rs200629713, in a total of 1099 BPD samples and 1056 controls. Burden analysis of these rare variants did not show a significant association with BPD. However, nine patients are compound heterozygotes for variants in SYNE1/CPG2, suggesting that rare coding variants may contribute significantly towards the complex genetic architecture underlying BPD. Imputation analysis in our own whole-genome sequencing sample of 99 BPD individuals identified an additional eight risk variants in the CPG2 region of SYNE1.
Collapse
|
10
|
Yamamuro K, Kimoto S, Iida J, Kishimoto N, Tanaka S, Toritsuka M, Ikawa D, Yamashita Y, Ota T, Makinodan M, Yoshino H, Kishimoto T. Distinct patterns of blood oxygenation in the prefrontal cortex in clinical phenotypes of schizophrenia and bipolar disorder. J Affect Disord 2018. [PMID: 29522943 DOI: 10.1016/j.jad.2018.02.065] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND Schizophrenia (SZ) and bipolar disorder (BD) are characterized by different clinical symptoms, and have previously been considered as categorically separate. However, several lines of evidence controversially suggest that these two disorders may run on a continuum. While it is therefore important to evaluate the subtle differences between SZ and BD, few studies have investigated the difference of brain functioning between the two by focusing on the common symptoms of cognitive functioning and impulsivity, rather than positive/negative and mood symptoms. Recent developments in near-infrared spectroscopy (NIRS) technology have enabled noninvasive assessment of brain function in people with psychiatric disorders. METHODS Near-infrared spectroscopy (NIRS) using 24-channels was conducted during the verbal fluency task (VFT) and Stroop color-word task (SCWT) in 38 patients diagnosed with SZ, 34 patients with BD, and 26 age- and sex-matched healthy controls. RESULTS Oxyhemoglobin changes in the prefrontal cortex (PFC) were significantly lower particularly in the SZ compared to control group during the VFT. On the other hand, these were significantly lower particularly in the BD and SZ group to control group during the SCWT. Regression analysis showed that hemodynamic changes were significantly correlated with verbal memory and impulsivity in both disorders. CONCLUSION These findings suggest that different hemodynamic responses in the prefrontal cortex might reflect cognitive functioning and impulsivity, providing a greater insight into SZ and BD pathophysiology.
Collapse
Affiliation(s)
- Kazuhiko Yamamuro
- Department of Psychiatry, Nara Medical University School of Medicine, Kashihara, Japan.
| | - Sohei Kimoto
- Department of Psychiatry, Nara Medical University School of Medicine, Kashihara, Japan
| | - Junzo Iida
- Faculty of Nursing, Nara Medical University School of Medicine, Kashihara, Japan
| | - Naoko Kishimoto
- Department of Psychiatry, Nara Medical University School of Medicine, Kashihara, Japan
| | - Shohei Tanaka
- Department of Psychiatry, Nara Medical University School of Medicine, Kashihara, Japan
| | - Michihiro Toritsuka
- Department of Psychiatry, Nara Medical University School of Medicine, Kashihara, Japan
| | - Daisuke Ikawa
- Department of Psychiatry, Nara Medical University School of Medicine, Kashihara, Japan
| | - Yasunori Yamashita
- Department of Psychiatry, Nara Medical University School of Medicine, Kashihara, Japan
| | - Toyosaku Ota
- Department of Psychiatry, Nara Medical University School of Medicine, Kashihara, Japan
| | - Manabu Makinodan
- Department of Psychiatry, Nara Medical University School of Medicine, Kashihara, Japan
| | - Hiroki Yoshino
- Department of Psychiatry, Nara Medical University School of Medicine, Kashihara, Japan
| | - Toshifumi Kishimoto
- Department of Psychiatry, Nara Medical University School of Medicine, Kashihara, Japan
| |
Collapse
|
11
|
Weißflog L, Becker N, Bossert N, Freudenberg F, Kittel-Schneider S, Reif A. Expressional profile of the diacylglycerol kinase eta gene DGKH. Eur Arch Psychiatry Clin Neurosci 2017; 267:445-454. [PMID: 27085324 DOI: 10.1007/s00406-016-0695-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 04/09/2016] [Indexed: 11/25/2022]
Abstract
Bipolar disorder (BPD) is a genetically complex mental disorder, which is characterized by recurrent depressive and manic episodes, occurring with a typical cyclical course. In a recent study, we were able to identify a risk haplotype for BPD, as well as for unipolar depression and adult attention-deficit/hyperactivity disorder (ADHD), within the DGKH gene. DGKH codes for the eta (η) isoform of diacylglycerol kinase, which is involved in the phosphoinositol pathway. In the present study, we determined the expressional profile of Dgkh using quantitative real-time PCR (qPCR), in situ hybridization and immunohistological staining in the human and in the mouse brain. Expression studies showed that two different Dgkh transcripts exhibited distinct occurrence in a variety of murine tissues and also differed in their expression levels. The proteins encoded by those transcripts differ in functional protein domains suggesting distinct biochemical and cell biological properties and functions. qPCR analyses revealed an increase in Dgkh expression during mouse brain development indicating a possible role of this kinase in late developmental stages. Immunostainings revealed strong Dgkh expression in neurons of the hippocampus and the cerebellum of the murine brain, whereas highest expression levels of DGKH in the human brain were found in the striatum. Taken together, our studies revealed expressional changes during mouse brain development and occurrence of Dgkη in neurons of regions that have been linked to BPD as well as ADHD in humans providing evidence for the implication of DGKH in those disorders.
Collapse
Affiliation(s)
- Lena Weißflog
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital of Frankfurt, Heinrich-Hoffmann-Str. 10, 60528, Frankfurt, Germany.
| | - Nils Becker
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital of Würzburg, Füchsleinstr. 15, 97080, Würzburg, Germany
- Department of Behavioral Physiology and Sociobiology, Theodor-Boveri-Institute of Bioscience, University of Würzburg, Würzburg, Germany
| | - Nelli Bossert
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital of Würzburg, Füchsleinstr. 15, 97080, Würzburg, Germany
- Leiden Institute of Physics, Leiden University, Leiden, Netherlands
| | - Florian Freudenberg
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital of Frankfurt, Heinrich-Hoffmann-Str. 10, 60528, Frankfurt, Germany
| | - Sarah Kittel-Schneider
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital of Frankfurt, Heinrich-Hoffmann-Str. 10, 60528, Frankfurt, Germany
| | - Andreas Reif
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital of Frankfurt, Heinrich-Hoffmann-Str. 10, 60528, Frankfurt, Germany
| |
Collapse
|
12
|
van de Leemput J, Hess JL, Glatt SJ, Tsuang MT. Genetics of Schizophrenia: Historical Insights and Prevailing Evidence. ADVANCES IN GENETICS 2016; 96:99-141. [PMID: 27968732 DOI: 10.1016/bs.adgen.2016.08.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Schizophrenia's (SZ's) heritability and familial transmission have been known for several decades; however, despite the clear evidence for a genetic component, it has been very difficult to pinpoint specific causative genes. Even so genetic studies have taught us a lot, even in the pregenomic era, about the molecular underpinnings and disease-relevant pathways. Recurring themes emerged revealing the involvement of neurodevelopmental processes, glutamate regulation, and immune system differential activation in SZ etiology. The recent emergence of epigenetic studies aimed at shedding light on the biological mechanisms underlying SZ has provided another layer of information in the investigation of gene and environment interactions. However, this epigenetic insight also brings forth another layer of complexity to the (epi)genomic landscape such as interactions between genetic variants, epigenetic marks-including cross-talk between DNA methylation and histone modification processes-, gene expression regulation, and environmental influences. In this review, we seek to synthesize perspectives, including limitations and obstacles yet to overcome, from genetic and epigenetic literature on SZ through a qualitative review of risk factors and prevailing hypotheses. Encouraged by the findings of both genetic and epigenetic studies to date, as well as the continued development of new technologies to collect and interpret large-scale studies, we are left with a positive outlook for the future of elucidating the molecular genetic mechanisms underlying SZ and other complex neuropsychiatric disorders.
Collapse
Affiliation(s)
- J van de Leemput
- University of California, San Diego, La Jolla, CA, United States
| | - J L Hess
- SUNY Upstate Medical University, Syracuse, NY, United States
| | - S J Glatt
- SUNY Upstate Medical University, Syracuse, NY, United States
| | - M T Tsuang
- University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
13
|
Abstract
Mitochondrial diseases are a clinically heterogeneous group of disorders that ultimately result from dysfunction of the mitochondrial respiratory chain. There is some evidence to suggest that mitochondrial dysfunction plays a role in neuropsychiatric illness; however, the data are inconclusive. This article summarizes the available literature published in the area of neuropsychiatric manifestations in both children and adults with primary mitochondrial disease, with a focus on autism spectrum disorder in children and mood disorders and schizophrenia in adults.
Collapse
Affiliation(s)
- Samantha E Marin
- Department of Neurosciences, University of California, San Diego (UCSD), 9500 Gilman Drive #0935, La Jolla, CA 92093-0935, USA
| | - Russell P Saneto
- Department of Neurology, Seattle Children's Hospital, University of Washington, 4800 Sand Point Way Northeast, Seattle, WA 98105, USA; Department of Pediatrics, Seattle Children's Hospital, University of Washington, 4800 Sand Point Way Northeast, Seattle, WA 98105, USA.
| |
Collapse
|
14
|
Plant N. Can a systems approach produce a better understanding of mood disorders? Biochim Biophys Acta Gen Subj 2016; 1861:3335-3344. [PMID: 27565355 DOI: 10.1016/j.bbagen.2016.08.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 07/29/2016] [Accepted: 08/22/2016] [Indexed: 10/21/2022]
Abstract
BACKGROUND One in twenty-five people suffer from a mood disorder. Current treatments are sub-optimal with poor patient response and uncertain modes-of-action. There is thus a need to better understand underlying mechanisms that determine mood, and how these go wrong in affective disorders. Systems biology approaches have yielded important biological discoveries for other complex diseases such as cancer, and their potential in affective disorders will be reviewed. SCOPE OF REVIEW This review will provide a general background to affective disorders, plus an outline of experimental and computational systems biology. The current application of these approaches in understanding affective disorders will be considered, and future recommendations made. MAJOR CONCLUSIONS Experimental systems biology has been applied to the study of affective disorders, especially at the genome and transcriptomic levels. However, data generation has been slowed by a lack of human tissue or suitable animal models. At present, computational systems biology has only be applied to understanding affective disorders on a few occasions. These studies provide sufficient novel biological insight to motivate further use of computational biology in this field. GENERAL SIGNIFICANCE In common with many complex diseases much time and money has been spent on the generation of large-scale experimental datasets. The next step is to use the emerging computational approaches, predominantly developed in the field of oncology, to leverage the most biological insight from these datasets. This will lead to the critical breakthroughs required for more effective diagnosis, stratification and treatment of affective disorders.
Collapse
Affiliation(s)
- Nick Plant
- School of Bioscience and Medicine, Faculty of Health and Medical Science, University of Surrey, Guildford GU2 7XH, UK.
| |
Collapse
|
15
|
Hollins SL, Cairns MJ. MicroRNA: Small RNA mediators of the brains genomic response to environmental stress. Prog Neurobiol 2016; 143:61-81. [PMID: 27317386 DOI: 10.1016/j.pneurobio.2016.06.005] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Revised: 05/24/2016] [Accepted: 06/11/2016] [Indexed: 01/09/2023]
Abstract
The developmental processes that establish the synaptic architecture of the brain while retaining capacity for activity-dependent remodeling, are complex and involve a combination of genetic and epigenetic influences. Dysregulation of these processes can lead to problems with neural circuitry which manifest in humans as a range of neurodevelopmental syndromes, such as schizophrenia, bipolar disorder and fragile X mental retardation. Recent studies suggest that prenatal, postnatal and intergenerational environmental factors play an important role in the aetiology of stress-related psychopathology. A number of these disorders have been shown to display epigenetic changes in the postmortem brain that reflect early life experience. These changes affect the regulation of gene expression though chromatin remodeling (transcriptional) and post-transcriptional influences, especially small noncoding microRNA (miRNA). These dynamic and influential molecules appear to play an important function in both brain development and its adaption to stress. In this review, we examine the role of miRNA in mediating the brain's response to both prenatal and postnatal environmental perturbations and explore how stress- induced alterations in miRNA expression can regulate the stress response via modulation of the immune system. Given the close relationship between environmental stress, miRNA, and brain development/function, we assert that miRNA hold a significant position at the molecular crossroads between neural development and adaptations to environmental stress. A greater understanding of the dynamics that mediate an individual's predisposition to stress-induced neuropathology has major human health benefits and is an important area of research.
Collapse
Affiliation(s)
- Sharon L Hollins
- School of Biomedical Sciences and Pharmacy and the Hunter Medical Research Institute, the University of Newcastle, Callaghan, NSW 2308, Australia
| | - Murray J Cairns
- School of Biomedical Sciences and Pharmacy and the Hunter Medical Research Institute, the University of Newcastle, Callaghan, NSW 2308, Australia; Schizophrenia Research Institute, Sydney, NSW, Australia.
| |
Collapse
|
16
|
Safari R, Salimi R, Tunca Z, Ozerdem A, Ceylan D, Sakizli M. Mutation/SNP analysis in EF-hand calcium binding domain of mitochondrial Ca2+ uptake 1 gene in bipolar disorder patients. J Integr Neurosci 2016; 15:163-73. [DOI: 10.1142/s0219635216500096] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
17
|
Chen J, Calhoun VD, Perrone-Bizzozero NI, Pearlson GD, Sui J, Du Y, Liu J. A pilot study on commonality and specificity of copy number variants in schizophrenia and bipolar disorder. Transl Psychiatry 2016; 6:e824. [PMID: 27244233 PMCID: PMC5545651 DOI: 10.1038/tp.2016.96] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 03/17/2016] [Indexed: 12/11/2022] Open
Abstract
Schizophrenia (SZ) and bipolar disorder (BD) are known to share genetic risks. In this work, we conducted whole-genome scanning to identify cross-disorder and disorder-specific copy number variants (CNVs) for these two disorders. The Database of Genotypes and Phenotypes (dbGaP) data were used for discovery, deriving from 2416 SZ patients, 592 BD patients and 2393 controls of European Ancestry, as well as 998 SZ patients, 121 BD patients and 822 controls of African Ancestry. PennCNV and Birdsuite detected high-confidence CNVs that were aggregated into CNV regions (CNVRs) and compared with the database of genomic variants for confirmation. Then, large (size⩾500 kb) and small common CNVRs (size <500 kb, frequency⩾1%) were examined for their associations with SZ and BD. Particularly for the European Ancestry samples, the dbGaP findings were further evaluated in the Wellcome Trust Case Control Consortium (WTCCC) data set for replication. Previously implicated variants (1q21.1, 15q13.3, 16p11.2 and 22q11.21) were replicated. Some cross-disorder variants were noted to differentially affect SZ and BD, including CNVRs in chromosomal regions encoding immunoglobulins and T-cell receptors that were associated more with SZ, and the 10q11.21 small CNVR (GPRIN2) associated more with BD. Disorder-specific CNVRs were also found. The 22q11.21 CNVR (COMT) and small CNVRs in 11p15.4 (TRIM5) and 15q13.2 (ARHGAP11B and FAN1) appeared to be SZ-specific. CNVRs in 17q21.2, 9p21.3 and 9q21.13 might be BD-specific. Overall, our primary findings in individual disorders largely echo previous reports. In addition, the comparison between SZ and BD reveals both specific and common risk CNVs. Particularly for the latter, differential involvement is noted, motivating further comparative studies and quantitative models.
Collapse
Affiliation(s)
- J Chen
- The Mind Research Network, Albuquerque, NM, USA
| | - V D Calhoun
- The Mind Research Network, Albuquerque, NM, USA
- Department of Electrical Engineering, University of New Mexico, Albuquerque, NM, USA
| | - N I Perrone-Bizzozero
- Departments of Neurosciences and Psychiatry, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - G D Pearlson
- Olin Neuropsychiatry Research Center, Institute of Living, Hartford, CT, USA
- Departments of Psychiatry and Neurobiology, Yale University, New Haven, CT, USA
| | - J Sui
- The Mind Research Network, Albuquerque, NM, USA
- Brainnetome Center and National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Y Du
- The Mind Research Network, Albuquerque, NM, USA
| | - J Liu
- The Mind Research Network, Albuquerque, NM, USA
- Department of Electrical Engineering, University of New Mexico, Albuquerque, NM, USA
| |
Collapse
|
18
|
Staiger EA, Albright JD, Brooks SA. Genome‐wide association mapping of heritable temperament variation in the
T
ennessee
W
alking
H
orse. GENES BRAIN AND BEHAVIOR 2016; 15:514-26. [DOI: 10.1111/gbb.12290] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 03/03/2016] [Accepted: 03/11/2016] [Indexed: 12/26/2022]
Affiliation(s)
- E. A. Staiger
- Department of Animal Science Cornell University Ithaca NY
| | - J. D. Albright
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine University of Tennessee Knoxville TN
| | - S. A. Brooks
- Department of Animal Science University of Florida Gainesville FL USA
| |
Collapse
|
19
|
Forero DA, Herteleer L, De Zutter S, Norrback KF, Nilsson LG, Adolfsson R, Callaerts P, Del-Favero J. A network of synaptic genes associated with schizophrenia and bipolar disorder. Schizophr Res 2016; 172:68-74. [PMID: 26899345 DOI: 10.1016/j.schres.2016.02.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 02/04/2016] [Accepted: 02/05/2016] [Indexed: 11/16/2022]
Abstract
Identification of novel candidate genes for schizophrenia (SZ) and bipolar disorder (BP), two psychiatric disorders with large epidemiological impacts, is a key research area in neurosciences and psychiatric genetics. Previous evidence from genome-wide studies suggests an important role for genes involved in synaptic plasticity in the risk for SZ and BP. We used a convergent genomics approach, combining different lines of biological evidence, to identify genes involved in the cAMP/PKA/CREB functional pathway that could be novel candidates for BP and SZ: CREB1, CREM, GRIN2C, NPY2R, NF1, PPP3CB and PRKAR1A. These 7 genes were analyzed in a HapMap based association study comprising 48 common SNPs in 486 SZ, 351 BP patients and 514 control individuals recruited from an isolated population in Northern Sweden. Genetic analysis showed significant allelic associations of SNPs in PRKAR1A with SZ and of PPP3CB and PRKAR1A with BP. Our results highlight the feasibility and the importance of convergent genomic data analysis for the identification of candidate genes and our data provide support for the role of common inherited variants in synaptic genes and their involvement in the etiology of BP and SZ.
Collapse
Affiliation(s)
- Diego A Forero
- Applied Molecular Genomics Unit, Department of Molecular Genetics, VIB, Belgium; University of Antwerp, Antwerp, Belgium; Laboratory of Behavioral and Developmental Genetics, VIB, Belgium; Catholic University of Leuven, Leuven, Belgium; Laboratory of NeuroPsychiatric Genetics, School of Medicine, Universidad Antonio Nariño, Bogotá, Colombia
| | - Liesbet Herteleer
- Laboratory of Behavioral and Developmental Genetics, VIB, Belgium; Catholic University of Leuven, Leuven, Belgium
| | - Sonia De Zutter
- Applied Molecular Genomics Unit, Department of Molecular Genetics, VIB, Belgium; University of Antwerp, Antwerp, Belgium
| | - Karl-Fredrik Norrback
- Department of Clinical Sciences, Division of Psychiatry, University of Umeå, Umeå, Sweden; Sunderby Hospital, Sweden
| | | | - Rolf Adolfsson
- Department of Clinical Sciences, Division of Psychiatry, University of Umeå, Umeå, Sweden; Sunderby Hospital, Sweden
| | - Patrick Callaerts
- Laboratory of Behavioral and Developmental Genetics, VIB, Belgium; Catholic University of Leuven, Leuven, Belgium.
| | - Jurgen Del-Favero
- Applied Molecular Genomics Unit, Department of Molecular Genetics, VIB, Belgium; University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
20
|
Novel, primate-specific PDE10A isoform highlights gene expression complexity in human striatum with implications on the molecular pathology of bipolar disorder. Transl Psychiatry 2016; 6:e742. [PMID: 26905414 PMCID: PMC4872433 DOI: 10.1038/tp.2016.3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Revised: 12/08/2015] [Accepted: 12/21/2015] [Indexed: 12/12/2022] Open
Abstract
Bipolar disorder is a highly heritable neuropsychiatric disorder affecting nearly 2.5% of the population. Prior genetic studies identified a panel of common and rare single-nucleotide polymorphisms associated with the disease that map to the first intron of the PDE10A gene. RNA sequencing of striatal brain tissue from bipolar and healthy control subjects identified a novel transcript of PDE10A, named PDE10A19, that codes for a PDE10A isoform with a unique N terminus. Genomic sequences that can encode the novel N terminus were conserved in other primates but not rodents. The RNA transcript was expressed at equal or greater levels in the human striatum compared with the two annotated transcripts, PDE10A1 and PDE10A2. The PDE10A19 transcript was detected in polysomal fractions; western blotting experiments confirmed that the RNA transcript is translated into protein. Immunocytochemistry studies using transfected mouse striatal and cortical neurons demonstrated that the PDE10A19 protein distributes to the cytosol, like PDE10A1, and unlike PDE10A2, which is associated with plasma membranes. Immunoprecipitation and immunocytochemical experiments revealed that the PDE10A19 isoform interacts physically with PDE10A2 and, when expressed at elevated levels, interferes with the plasma membrane localization of PDE10A2. These studies illustrate the complexity of PDE10A gene expression in the human brain and highlight the need to unravel the gene's complex and complete coding capabilities along with its transcriptional and translational regulation to guide the development of therapeutic agents that target the protein for the treatment of neuropsychiatric illness.
Collapse
|
21
|
Acikel C, Aydin Son Y, Celik C, Gul H. Evaluation of potential novel variations and their interactions related to bipolar disorders: analysis of genome-wide association study data. Neuropsychiatr Dis Treat 2016; 12:2997-3004. [PMID: 27920536 PMCID: PMC5127431 DOI: 10.2147/ndt.s112558] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Multifactor dimensionality reduction (MDR) is a nonparametric approach that can be used to detect relevant interactions between single-nucleotide polymorphisms (SNPs). The aim of this study was to build the best genomic model based on SNP associations and to identify candidate polymorphisms that are the underlying molecular basis of the bipolar disorders. METHODS This study was performed on Whole-Genome Association Study of Bipolar Disorder (dbGaP [database of Genotypes and Phenotypes] study accession number: phs000017.v3.p1) data. After preprocessing of the genotyping data, three classification-based data mining methods (ie, random forest, naïve Bayes, and k-nearest neighbor) were performed. Additionally, as a nonparametric, model-free approach, the MDR method was used to evaluate the SNP profiles. The validity of these methods was evaluated using true classification rate, recall (sensitivity), precision (positive predictive value), and F-measure. RESULTS Random forests, naïve Bayes, and k-nearest neighbors identified 16, 13, and ten candidate SNPs, respectively. Surprisingly, the top six SNPs were reported by all three methods. Random forests and k-nearest neighbors were more successful than naïve Bayes, with recall values >0.95. On the other hand, MDR generated a model with comparable predictive performance based on five SNPs. Although different SNP profiles were identified in MDR compared to the classification-based models, all models mapped SNPs to the DOCK10 gene. CONCLUSION Three classification-based data mining approaches, random forests, naïve Bayes, and k-nearest neighbors, have prioritized similar SNP profiles as predictors of bipolar disorders, in contrast to MDR, which has found different SNPs through analysis of two-way and three-way interactions. The reduced number of associated SNPs discovered by MDR, without loss in the classification performance, would facilitate validation studies and decision support models, and would reduce the cost to develop predictive and diagnostic tests. Nevertheless, we need to emphasize that translation of genomic models to the clinical setting requires models with higher classification performance.
Collapse
Affiliation(s)
| | - Yesim Aydin Son
- Department of Health Informatics, Graduate School of Informatics, Middle East Technical University
| | | | - Husamettin Gul
- Department of Medical Informatics, Gulhane Military Medical Academy, Ankara, Turkey
| |
Collapse
|
22
|
O'Shea KS, McInnis MG. Neurodevelopmental origins of bipolar disorder: iPSC models. Mol Cell Neurosci 2015; 73:63-83. [PMID: 26608002 DOI: 10.1016/j.mcn.2015.11.006] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Revised: 10/14/2015] [Accepted: 11/18/2015] [Indexed: 12/22/2022] Open
Abstract
Bipolar disorder (BP) is a chronic neuropsychiatric condition characterized by pathological fluctuations in mood from mania to depression. Adoption, twin and family studies have consistently identified a significant hereditary component to BP, yet there is no clear genetic event or consistent neuropathology. BP has been suggested to have a developmental origin, although this hypothesis has been difficult to test since there are no viable neurons or glial cells to analyze, and research has relied largely on postmortem brain, behavioral and imaging studies, or has examined proxy tissues including saliva, olfactory epithelium and blood cells. Neurodevelopmental factors, particularly pathways related to nervous system development, cell migration, extracellular matrix, H3K4 methylation, and calcium signaling have been identified in large gene expression and GWAS studies as altered in BP. Recent advances in stem cell biology, particularly the ability to reprogram adult somatic tissues to a pluripotent state, now make it possible to interrogate these pathways in viable cell models. A number of induced pluripotent stem cell (iPSC) lines from BP patient and healthy control (C) individuals have been derived in several laboratories, and their ability to form cortical neurons examined. Early studies suggest differences in activity, calcium signaling, blocks to neuronal differentiation, and changes in neuronal, and possibly glial, lineage specification. Initial observations suggest that differentiation of BP patient-derived neurons to dorsal telencephalic derivatives may be impaired, possibly due to alterations in WNT, Hedgehog or Nodal pathway signaling. These investigations strongly support a developmental contribution to BP and identify novel pathways, mechanisms and opportunities for improved treatments.
Collapse
Affiliation(s)
- K Sue O'Shea
- Department of Cell and Developmental Biology, University of Michigan, 3051 BSRB, 109 Zina Pitcher PL, Ann Arbor, MI 48109-2200, United States; Department of Psychiatry, University of Michigan, 4250 Plymouth Rd, Ann Arbor, MI 48109-5765, United States.
| | - Melvin G McInnis
- Department of Psychiatry, University of Michigan, 4250 Plymouth Rd, Ann Arbor, MI 48109-5765, United States
| |
Collapse
|
23
|
Waters RP, Rivalan M, Bangasser DA, Deussing JM, Ising M, Wood SK, Holsboer F, Summers CH. Evidence for the role of corticotropin-releasing factor in major depressive disorder. Neurosci Biobehav Rev 2015; 58:63-78. [PMID: 26271720 DOI: 10.1016/j.neubiorev.2015.07.011] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 06/24/2015] [Accepted: 07/24/2015] [Indexed: 01/05/2023]
Abstract
Major depressive disorder (MDD) is a devastating disease affecting over 300 million people worldwide, and costing an estimated 380 billion Euros in lost productivity and health care in the European Union alone. Although a wealth of research has been directed toward understanding and treating MDD, still no therapy has proved to be consistently and reliably effective in interrupting the symptoms of this disease. Recent clinical and preclinical studies, using genetic screening and transgenic rodents, respectively, suggest a major role of the CRF1 gene, and the central expression of CRF1 receptor protein in determining an individual's risk of developing MDD. This gene is widely expressed in brain tissue, and regulates an organism's immediate and long-term responses to social and environmental stressors, which are primary contributors to MDD. This review presents the current state of knowledge on CRF physiology, and how it may influence the occurrence of symptoms associated with MDD. Additionally, this review presents findings from multiple laboratories that were presented as part of a symposium on this topic at the annual 2014 meeting of the International Behavioral Neuroscience Society (IBNS). The ideas and data presented in this review demonstrate the great progress that has been made over the past few decades in our understanding of MDD, and provide a pathway forward toward developing novel treatments and detection methods for this disorder.
Collapse
Affiliation(s)
| | | | | | - J M Deussing
- Max Planck Institute of Psychiatry, Munich, Germany
| | - M Ising
- Max Planck Institute of Psychiatry, Munich, Germany
| | - S K Wood
- University of South Carolina School of Medicine, Columbia, SC, USA
| | - F Holsboer
- Max Planck Institute of Psychiatry, Munich, Germany; HMNC GmbH, Munich, Germany
| | - Cliff H Summers
- University of South Dakota, Vermillion, SD, USA; Sanford School of Medicine, Vermillion, SD, USA.
| |
Collapse
|
24
|
A composite peripheral blood gene expression measure as a potential diagnostic biomarker in bipolar disorder. Transl Psychiatry 2015; 5:e614. [PMID: 26241352 PMCID: PMC4564565 DOI: 10.1038/tp.2015.110] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 06/15/2015] [Accepted: 06/25/2015] [Indexed: 12/11/2022] Open
Abstract
Gene expression in peripheral blood has the potential to inform on pathophysiological mechanisms and has emerged as a viable avenue for the identification of biomarkers. Here, we aimed to identify gene expression candidate genes and to explore the potential for a composite gene expression measure as a diagnostic and state biomarker in bipolar disorder. First, messenger RNA levels of 19 candidate genes were assessed in peripheral blood mononuclear cells of 37 rapid cycling bipolar disorder patients in different affective states (depression, mania and euthymia) during a 6-12-month period and in 40 age- and gender-matched healthy control subjects. Second, a composite gene expression measure was constructed in the first half study sample and independently validated in the second half of the sample. We found downregulation of POLG and OGG1 expression in bipolar disorder patients compared with healthy control subjects. In patients with bipolar disorder, upregulation of NDUFV2 was observed in a depressed state compared with a euthymic state. The composite gene expression measure for discrimination between patients and healthy control subjects on the basis of 19 genes generated an area under the receiver-operating characteristic curve of 0.81 (P < 0.0001) in sample 1, which was replicated with a value of 0.73 (P < 0.0001) in sample 2, corresponding with a moderately accurate test. The present findings of altered POLG, OGG1 and NDUFV2 expression point to disturbances within mitochondrial function and DNA repair mechanisms in bipolar disorder. Further, a composite gene expression measure could hold promise as a potential diagnostic biomarker.
Collapse
|
25
|
Koyama Y, Hattori T, Nishida T, Hori O, Tohyama M. Alterations in dendrite and spine morphology of cortical pyramidal neurons in DISC1-binding zinc finger protein (DBZ) knockout mice. Front Neuroanat 2015; 9:52. [PMID: 25983680 PMCID: PMC4415407 DOI: 10.3389/fnana.2015.00052] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Accepted: 04/14/2015] [Indexed: 11/13/2022] Open
Abstract
Dendrite and dendritic spine formation are crucial for proper brain function. DISC1-binding zinc finger protein (DBZ) was first identified as a Disrupted-In-Schizophrenia1 (DISC1) binding partner. DBZ is highly expressed in the cerebral cortex of developing and adult rodents and is involved in neurite formation, cell positioning, and the development of interneurons and oligodendrocytes. The functional roles of DBZ in postnatal brain remain unknown; thus we investigated cortical pyramidal neuron morphology in DBZ knockout (KO) mice. Morphological analyses by Golgi staining alone in DBZ KO mice revealed decreased dendritic arborization, increased spine density. A morphological analysis of the spines revealed markedly increased numbers of thin spines. To investigate whole spine structure in detail, electron tomographic analysis using ultra-high voltage electron microscopy (UHVEM) combined with Golgi staining was performed. Tomograms and three-dimensional models of spines revealed that the spines of DBZ KO mice exhibited two types of characteristic morphology, filopodia-like spines and abnormal thin-necked spines having an extremely thin spine neck. Moreover, conventional electron microscopy revealed significantly decreased number of postsynaptic densities (PSDs) in spines of DBZ KO mice. In conclusion, DBZ deficiency impairs the morphogenesis of dendrites and spines in cortical pyramidal neurons.
Collapse
Affiliation(s)
- Yoshihisa Koyama
- Department of Anatomy and Neuroscience, Graduate School of Medicine, Osaka UniversitySuita, Osaka, Japan
| | - Tsuyoshi Hattori
- Department of Neuroanatomy, Kanazawa University Graduate School of Medical SciencesKanazawa, Ishikawa, Japan
- Department of Child Development and Molecular Brain Science, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui SuitaOsaka, Japan
| | - Tomoki Nishida
- Research Center for Ultra-High Voltage Electron Microscopy, Osaka UniversitySuita, Osaka, Japan
| | - Osamu Hori
- Department of Neuroanatomy, Kanazawa University Graduate School of Medical SciencesKanazawa, Ishikawa, Japan
| | - Masaya Tohyama
- Division of Molecular Brain Science, Research Institute of Traditional Asian Medicine, Kinki UniversityOsaka-sayama, Osaka, Japan
- Osaka Prefectural Hospital OrganizationOsaka, Japan
| |
Collapse
|
26
|
MIR137 variants identified in psychiatric patients affect synaptogenesis and neuronal transmission gene sets. Mol Psychiatry 2015; 20:472-81. [PMID: 24888363 DOI: 10.1038/mp.2014.53] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 04/24/2014] [Accepted: 04/28/2014] [Indexed: 02/07/2023]
Abstract
Sequence analysis of 13 microRNA (miRNA) genes expressed in the human brain and located in genomic regions associated with schizophrenia and/or bipolar disorder, in a northern Swedish patient/control population, resulted in the discovery of two functional variants in the MIR137 gene. On the basis of their location and the allele frequency differences between patients and controls, we explored the hypothesis that the discovered variants impact the expression of the mature miRNA and consequently influence global mRNA expression affecting normal brain functioning. Using neuronal-like SH-SY5Y cells, we demonstrated significantly reduced mature miR-137 levels in the cells expressing the variant miRNA gene. Subsequent transcriptome analysis showed that the reduction in miR-137 expression led to the deregulation of gene sets involved in synaptogenesis and neuronal transmission, all implicated in psychiatric disorders. Our functional findings add to the growing data, which implicate that miR-137 has an important role in the etiology of psychiatric disorders and emphasizes its involvement in nervous system development and proper synaptic function.
Collapse
|
27
|
Li Z, Deng W, Liu X, Zheng Z, Li M, Li Y, Han Y, Ma X, Wang Q, Liu X, Li T. Contingent negative variation in patients with deficit schizophrenia or bipolar I disorder with psychotic features: measurement and correlation with clinical characteristics. Nord J Psychiatry 2015; 69:196-203. [PMID: 25263850 DOI: 10.3109/08039488.2014.959562] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Schizophrenia is a highly heterogeneous disease. Event-related potentials have been regarded to establish intermediate phenotypes of schizophrenia. Our previous study found that patients with deficit schizophrenia (DS) are relatively homogeneous and show a significantly longer onset latency of contingent negative variation (CNV) expectancy wave. AIMS To further examine CNV in patients with first-episode and drug-naïve DS or bipolar I disorder (BP I) with psychotic features, and also investigate correlations between CNV and clinical characteristics in DS and BP I. METHOD We elicited a CNV using an alarm (S1)-imperative (S2) paradigm in 30 DS patients or 33 BP I with psychotic features as well as 40 healthy controls. RESULTS CNV amplitude was significantly smaller and reaction time significantly longer in the DS and BP I groups than in healthy controls. Post-imperative negative variation (PINV) interval was significantly shorter in the DS group than in healthy controls. The onset latency of CNV expectancy wave was significantly longer and PINV area significantly smaller in the DS group than in the other groups. In the DS group, CNV amplitude and PINV interval correlated negatively with the subscale of negative symptoms on the Positive and Negative Syndrome Scale (PANSS); CNV amplitude also correlated negatively with disease duration. In the BP I group, CNV amplitude and reaction time showed no correlation with clinical features. CONCLUSIONS CNV amplitude is a common trait marker for psychosis. The onset latency of CNV expectancy wave appears to be a specific trait marker and may be used to identify candidate genes for DS.
Collapse
Affiliation(s)
- Zhe Li
- Zhe Li, M.D., The Mental Health Center and the Psychiatric Laboratory, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University , Chengdu, Sichuan 610041 , China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Neale BM, Sklar P. Genetic analysis of schizophrenia and bipolar disorder reveals polygenicity but also suggests new directions for molecular interrogation. Curr Opin Neurobiol 2014; 30:131-8. [PMID: 25544106 DOI: 10.1016/j.conb.2014.12.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 11/25/2014] [Accepted: 12/01/2014] [Indexed: 12/12/2022]
Abstract
Over the last few years, genetics research has made significant strides in identifying many risk factors for schizophrenia and bipolar disorder. These risk factors include inherited common single nucleotide polymorphisms, copy number variants, and rare single nucleotide variants, as well as rare de novo variants. For all variants, the common theme has been that of polygenicity, meaning that many small genetic risk factors influence risk in the population and that no gene or variant on its own has been shown to be fully deterministic of schizophrenia or bipolar. When taken together, biological themes that have emerged including the importance of synaptic function and calcium signaling. This has implications for our understanding of the biological underpinnings of these diseases.
Collapse
Affiliation(s)
- Benjamin M Neale
- Analytical and Translational Genetics Unit and Psychiatric and Neurodevelopmental Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Stanley Center for Psychiatric Research and Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA.
| | - Pamela Sklar
- Division of Psychiatric Genomics, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
29
|
Gonzalez S, Camarillo C, Rodriguez M, Ramirez M, Zavala J, Armas R, Contreras SA, Contreras J, Dassori A, Almasy L, Flores D, Jerez A, Raventós H, Ontiveros A, Nicolini H, Escamilla M. A genome-wide linkage scan of bipolar disorder in Latino families identifies susceptibility loci at 8q24 and 14q32. Am J Med Genet B Neuropsychiatr Genet 2014; 165B:479-91. [PMID: 25044503 DOI: 10.1002/ajmg.b.32251] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 05/27/2014] [Indexed: 12/14/2022]
Abstract
A genome-wide nonparametric linkage screen was performed to localize Bipolar Disorder (BP) susceptibility loci in a sample of 3757 individuals of Latino ancestry. The sample included 963 individuals with BP phenotype (704 relative pairs) from 686 families recruited from the US, Mexico, Costa Rica, and Guatemala. Non-parametric analyses were performed over a 5 cM grid with an average genetic coverage of 0.67 cM. Multipoint analyses were conducted across the genome using non-parametric Kong & Cox LOD scores along with Sall statistics for all relative pairs. Suggestive and significant genome-wide thresholds were calculated based on 1000 simulations. Single-marker association tests in the presence of linkage were performed assuming a multiplicative model with a population prevalence of 2%. We identified two genome-wide significant susceptibly loci for BP at 8q24 and 14q32, and a third suggestive locus at 2q13-q14. Within these three linkage regions, the top associated single marker (rs1847694, P = 2.40 × 10(-5)) is located 195 Kb upstream of DPP10 in Chromosome 2. DPP10 is prominently expressed in brain neuronal populations, where it has been shown to bind and regulate Kv4-mediated A-type potassium channels. Taken together, these results provide additional evidence that 8q24, 14q32, and 2q13-q14 are susceptibly loci for BP and these regions may be involved in the pathogenesis of BP in the Latino population.
Collapse
Affiliation(s)
- Suzanne Gonzalez
- Center of Excellence for Neurosciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, Texas; Department of Psychiatry, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, Texas
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Fiorentino A, O'Brien NL, Locke DP, McQuillin A, Jarram A, Anjorin A, Kandaswamy R, Curtis D, Blizard RA, Gurling HMD. Analysis of ANK3 and CACNA1C variants identified in bipolar disorder whole genome sequence data. Bipolar Disord 2014; 16:583-91. [PMID: 24716743 PMCID: PMC4227602 DOI: 10.1111/bdi.12203] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Accepted: 12/27/2013] [Indexed: 12/30/2022]
Abstract
OBJECTIVES Genetic markers in the genes encoding ankyrin 3 (ANK3) and the α-calcium channel subunit (CACNA1C) are associated with bipolar disorder (BP). The associated variants in the CACNA1C gene are mainly within intron 3 of the gene. ANK3 BP-associated variants are in two distinct clusters at the ends of the gene, indicating disease allele heterogeneity. METHODS In order to screen both coding and non-coding regions to identify potential aetiological variants, we used whole-genome sequencing in 99 BP cases. Variants with markedly different allele frequencies in the BP samples and the 1,000 genomes project European data were genotyped in 1,510 BP cases and 1,095 controls. RESULTS We found that the CACNA1C intron 3 variant, rs79398153, potentially affecting an ENCyclopedia of DNA Elements (ENCODE)-defined region, showed an association with BP (p = 0.015). We also found the ANK3 BP-associated variant rs139972937, responsible for an asparagine to serine change (p = 0.042). However, a previous study had not found support for an association between rs139972937 and BP. The variants at ANK3 and CACNA1C previously known to be associated with BP were not in linkage disequilibrium with either of the two variants that we identified and these are therefore independent of the previous haplotypes implicated by genome-wide association. CONCLUSIONS Sequencing in additional BP samples is needed to find the molecular pathology that explains the previous association findings. If changes similar to those we have found can be shown to have an effect on the expression and function of ANK3 and CACNA1C, they might help to explain the so-called 'missing heritability' of BP.
Collapse
Affiliation(s)
- Alessia Fiorentino
- Molecular Psychiatry Laboratory, Division of Psychiatry, University College LondonLondon, UK
| | - Niamh Louise O'Brien
- Molecular Psychiatry Laboratory, Division of Psychiatry, University College LondonLondon, UK
| | | | - Andrew McQuillin
- Molecular Psychiatry Laboratory, Division of Psychiatry, University College LondonLondon, UK
| | - Alexandra Jarram
- Molecular Psychiatry Laboratory, Division of Psychiatry, University College LondonLondon, UK
| | - Adebayo Anjorin
- Molecular Psychiatry Laboratory, Division of Psychiatry, University College LondonLondon, UK
| | - Radhika Kandaswamy
- Molecular Psychiatry Laboratory, Division of Psychiatry, University College LondonLondon, UK
| | - David Curtis
- Department of Psychological Medicine, Queen Mary University of LondonLondon, UK
| | - Robert Alan Blizard
- Molecular Psychiatry Laboratory, Division of Psychiatry, University College LondonLondon, UK
| | | |
Collapse
|
31
|
Alteration of imprinted Dlk1-Dio3 miRNA cluster expression in the entorhinal cortex induced by maternal immune activation and adolescent cannabinoid exposure. Transl Psychiatry 2014; 4:e452. [PMID: 25268256 PMCID: PMC4203021 DOI: 10.1038/tp.2014.99] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Revised: 07/09/2014] [Accepted: 08/21/2014] [Indexed: 12/19/2022] Open
Abstract
A significant feature of the cortical neuropathology of schizophrenia is a disturbance in the biogenesis of short non-coding microRNA (miRNA) that regulate translation and stability of mRNA. While the biological origin of this phenomenon has not been defined, it is plausible that it relates to major environmental risk factors associated with the disorder such as exposure to maternal immune activation (MIA) and adolescent cannabis use. To explore this hypothesis, we administered the viral mimic poly I:C to pregnant rats and further exposed some of their maturing offsprings to daily injections of the synthetic cannabinoid HU210 for 14 days starting on postnatal day 35. Whole-genome miRNA expression analysis was then performed on the left and right hemispheres of the entorhinal cortex (EC), a region strongly associated with schizophrenia. Animals exposed to either treatment alone or in combination exhibited significant differences in the expression of miRNA in the left hemisphere, whereas the right hemisphere was less responsive. Hemisphere-associated differences in miRNA expression were greatest in the combined treatment and highly over-represented in a single imprinted locus on chromosome 6q32. This observation was significant as the syntenic 14q32 locus in humans encodes a large proportion of miRNAs differentially expressed in peripheral blood lymphocytes from patients with schizophrenia, suggesting that interaction of early and late environmental insults may affect miRNA expression, in a manner that is relevant to schizophrenia.
Collapse
|
32
|
Quednow BB, Brzózka MM, Rossner MJ. Transcription factor 4 (TCF4) and schizophrenia: integrating the animal and the human perspective. Cell Mol Life Sci 2014; 71:2815-35. [PMID: 24413739 PMCID: PMC11113759 DOI: 10.1007/s00018-013-1553-4] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Revised: 12/04/2013] [Accepted: 12/30/2013] [Indexed: 02/06/2023]
Abstract
Schizophrenia is a genetically complex disease considered to have a neurodevelopmental pathogenesis and defined by a broad spectrum of positive and negative symptoms as well as cognitive deficits. Recently, large genome-wide association studies have identified common alleles slightly increasing the risk for schizophrenia. Among the few schizophrenia-risk genes that have been consistently replicated is the basic Helix-Loop-Helix (bHLH) transcription factor 4 (TCF4). Haploinsufficiency of the TCF4 (formatting follows IUPAC nomenclature: TCF4 protein/protein function, Tcf4 rodent gene cDNA mRNA, TCF4 human gene cDNA mRNA) gene causes the Pitt-Hopkins syndrome-a neurodevelopmental disease characterized by severe mental retardation. Accordingly, Tcf4 null-mutant mice display developmental brain defects. TCF4-associated risk alleles are located in putative coding and non-coding regions of the gene. Hence, subtle changes at the level of gene expression might be relevant for the etiopathology of schizophrenia. Behavioural phenotypes obtained with a mouse model of slightly increased gene dosage and electrophysiological investigations with human risk-allele carriers revealed an overlapping spectrum of schizophrenia-relevant endophenotypes. Most prominently, early information processing and higher cognitive functions appear to be associated with TCF4 risk genotypes. Moreover, a recent human study unravelled gene × environment interactions between TCF4 risk alleles and smoking behaviour that were specifically associated with disrupted early information processing. Taken together, TCF4 is considered as an integrator ('hub') of several bHLH networks controlling critical steps of various developmental, and, possibly, plasticity-related transcriptional programs in the CNS and changes of TCF4 expression also appear to affect brain networks important for information processing. Consequently, these findings support the neurodevelopmental hypothesis of schizophrenia and provide a basis for identifying the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Boris B. Quednow
- Department of Psychiatry, Psychotherapy and Psychosomatics, Experimental and Clinical Pharmacopsychology, Psychiatric Hospital, University of Zurich, Lenggstrasse 31, 8032 Zurich, Switzerland
- Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Magdalena M. Brzózka
- Department of Psychiatry, Molecular and Behavioral Neurobiology, Ludwig-Maximillians-University, Nussbaumstr. 7, 80336 Munich, Germany
| | - Moritz J. Rossner
- Department of Psychiatry, Molecular and Behavioral Neurobiology, Ludwig-Maximillians-University, Nussbaumstr. 7, 80336 Munich, Germany
- Research Group Gene Expression, Max-Planck-Institute of Experimental Medicine, Hermann-Rein-Str. 3, Goettingen, 37075 Germany
| |
Collapse
|
33
|
Escott-Price V, Bellenguez C, Wang LS, Choi SH, Harold D, Jones L, Holmans P, Gerrish A, Vedernikov A, Richards A, DeStefano AL, Lambert JC, Ibrahim-Verbaas CA, Naj AC, Sims R, Jun G, Bis JC, Beecham GW, Grenier-Boley B, Russo G, Thornton-Wells TA, Denning N, Smith AV, Chouraki V, Thomas C, Ikram MA, Zelenika D, Vardarajan BN, Kamatani Y, Lin CF, Schmidt H, Kunkle B, Dunstan ML, Vronskaya M, Johnson AD, Ruiz A, Bihoreau MT, Reitz C, Pasquier F, Hollingworth P, Hanon O, Fitzpatrick AL, Buxbaum JD, Campion D, Crane PK, Baldwin C, Becker T, Gudnason V, Cruchaga C, Craig D, Amin N, Berr C, Lopez OL, De Jager PL, Deramecourt V, Johnston JA, Evans D, Lovestone S, Letenneur L, Hernández I, Rubinsztein DC, Eiriksdottir G, Sleegers K, Goate AM, Fiévet N, Huentelman MJ, Gill M, Brown K, Kamboh MI, Keller L, Barberger-Gateau P, McGuinness B, Larson EB, Myers AJ, Dufouil C, Todd S, Wallon D, Love S, Rogaeva E, Gallacher J, George-Hyslop PS, Clarimon J, Lleo A, Bayer A, Tsuang DW, Yu L, Tsolaki M, Bossù P, Spalletta G, Proitsi P, Collinge J, Sorbi S, Garcia FS, Fox NC, Hardy J, Naranjo MCD, Bosco P, Clarke R, Brayne C, Galimberti D, Scarpini E, Bonuccelli U, Mancuso M, Siciliano G, Moebus S, Mecocci P, Zompo MD, Maier W, Hampel H, Pilotto A, Frank-García A, Panza F, Solfrizzi V, Caffarra P, Nacmias B, Perry W, Mayhaus M, Lannfelt L, Hakonarson H, Pichler S, Carrasquillo MM, Ingelsson M, Beekly D, Alvarez V, Zou F, Valladares O, Younkin SG, Coto E, Hamilton-Nelson KL, Gu W, Razquin C, Pastor P, Mateo I, Owen MJ, Faber KM, Jonsson PV, Combarros O, O'Donovan MC, Cantwell LB, Soininen H, Blacker D, Mead S, Mosley TH, Bennett DA, Harris TB, Fratiglioni L, Holmes C, de Bruijn RFAG, Passmore P, Montine TJ, Bettens K, Rotter JI, Brice A, Morgan K, Foroud TM, Kukull WA, Hannequin D, Powell JF, Nalls MA, Ritchie K, Lunetta KL, Kauwe JSK, Boerwinkle E, Riemenschneider M, Boada M, Hiltunen M, Martin ER, Schmidt R, Rujescu D, Dartigues JF, Mayeux R, Tzourio C, Hofman A, Nöthen MM, Graff C, Psaty BM, Haines JL, Lathrop M, Pericak-Vance MA, Launer LJ, Van Broeckhoven C, Farrer LA, van Duijn CM, Ramirez A, Seshadri S, Schellenberg GD, Amouyel P, Williams J. Gene-wide analysis detects two new susceptibility genes for Alzheimer's disease. PLoS One 2014; 9:e94661. [PMID: 24922517 PMCID: PMC4055488 DOI: 10.1371/journal.pone.0094661] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 03/17/2014] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Alzheimer's disease is a common debilitating dementia with known heritability, for which 20 late onset susceptibility loci have been identified, but more remain to be discovered. This study sought to identify new susceptibility genes, using an alternative gene-wide analytical approach which tests for patterns of association within genes, in the powerful genome-wide association dataset of the International Genomics of Alzheimer's Project Consortium, comprising over 7 m genotypes from 25,580 Alzheimer's cases and 48,466 controls. PRINCIPAL FINDINGS In addition to earlier reported genes, we detected genome-wide significant loci on chromosomes 8 (TP53INP1, p = 1.4×10-6) and 14 (IGHV1-67 p = 7.9×10-8) which indexed novel susceptibility loci. SIGNIFICANCE The additional genes identified in this study, have an array of functions previously implicated in Alzheimer's disease, including aspects of energy metabolism, protein degradation and the immune system and add further weight to these pathways as potential therapeutic targets in Alzheimer's disease.
Collapse
Affiliation(s)
- Valentina Escott-Price
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, United Kingdom
| | - Céline Bellenguez
- Inserm U744, Lille, France
- Université Lille 2, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Li-San Wang
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Seung-Hoan Choi
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America
| | - Denise Harold
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, United Kingdom
| | - Lesley Jones
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, United Kingdom
| | - Peter Holmans
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, United Kingdom
| | - Amy Gerrish
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, United Kingdom
| | - Alexey Vedernikov
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, United Kingdom
| | - Alexander Richards
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, United Kingdom
| | - Anita L. DeStefano
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America
| | - Jean-Charles Lambert
- Inserm U744, Lille, France
- Université Lille 2, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Carla A. Ibrahim-Verbaas
- Department of Epidemiology and Neurology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Adam C. Naj
- Department of Biostatistics and Epidemiology and Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Rebecca Sims
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, United Kingdom
| | - Gyungah Jun
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Ophthalmology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Joshua C. Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Gary W. Beecham
- The John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida, United States of America
- Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami, Miami, Florida, United States of America
| | - Benjamin Grenier-Boley
- Inserm U744, Lille, France
- Université Lille 2, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Giancarlo Russo
- Functional Genomics Center Zurich, ETH/University of Zurich, Zurich, Switzerland
| | - Tricia A. Thornton-Wells
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Nicola Denning
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, United Kingdom
| | - Albert V. Smith
- University of Iceland, Faculty of Medicine, Reykjavik, Iceland
- Icelandic Heart Association, Kopavogur, Iceland
| | - Vincent Chouraki
- Inserm U744, Lille, France
- Université Lille 2, Lille, France
- Institut Pasteur de Lille, Lille, France
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Charlene Thomas
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, United Kingdom
| | - M. Arfan Ikram
- Departments of Epidemiology, Neurology and Radiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
- Netherlands Consortium for Healthy Aging, Leiden, The Netherlands
| | - Diana Zelenika
- Centre National de Genotypage, Institut Genomique, Commissariat à l'énergie Atomique, Evry, France
| | - Badri N. Vardarajan
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, Massachusetts, United States of America
- Taub Institute on Alzheimer's Disease and the Aging Brain, Department of Neurology, Columbia University New York, New York, United States of America
- Gertrude H. Sergievsky Center, Department of Neurology, Columbia University, New York, New York, United States of America
| | | | - Chiao-Feng Lin
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Helena Schmidt
- Institute for Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Brian Kunkle
- The John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida, United States of America
| | - Melanie L. Dunstan
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, United Kingdom
| | - Maria Vronskaya
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, United Kingdom
| | | | - Andrew D. Johnson
- NHLBI Cardiovascular Epidemiology and Human Genomics Branch, The Framingham Heart Study, Framingham, Massachusetts, United States of America
| | - Agustin Ruiz
- Memory Clinic of Fundació ACE. Institut Català de Neurociències Aplicades, Barcelona, Spain
| | - Marie-Thérèse Bihoreau
- Centre National de Genotypage, Institut Genomique, Commissariat à l'énergie Atomique, Evry, France
| | - Christiane Reitz
- Taub Institute on Alzheimer's Disease and the Aging Brain, Department of Neurology, Columbia University New York, New York, United States of America
- Gertrude H. Sergievsky Center, Department of Neurology, Columbia University, New York, New York, United States of America
| | - Florence Pasquier
- Université Lille 2, Lille, France
- CNR-MAJ, Centre Hospitalier Régional Universitaire de Lille, Lille, France
| | - Paul Hollingworth
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, United Kingdom
| | - Olivier Hanon
- University Paris Descartes, Sorbonne Paris V, Broca Hospital, Geriatrics department, Paris, France
| | - Annette L. Fitzpatrick
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington, United States of America
- Departments of Epidemiology and Global Health, University of Washington, Seattle, Washington, United States of America
| | - Joseph D. Buxbaum
- Department of Neuroscience, Mount Sinai School of Medicine, New York, New York, United States of America
- Department of Psychiatry, Mount Sinai School of Medicine, New York, New York, United States of America
- Departments of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Dominique Campion
- CNR-MAJ, Inserm U1079, Rouen University Hospital, 76031 France, Rouen, France
| | - Paul K. Crane
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Clinton Baldwin
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Tim Becker
- German Center for Neurodegenerative Diseases (DZNE), Bonn, and Institute for Medical Biometry, Informatics and Epidemiology, University of Bonn, Bonn, Germany
| | - Vilmundur Gudnason
- University of Iceland, Faculty of Medicine, Reykjavik, Iceland
- Icelandic Heart Association, Kopavogur, Iceland
| | - Carlos Cruchaga
- Department of Psychiatry and Hope Center Program on Protein Aggregation and Neurodegeneration, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - David Craig
- Ageing Group, Centre for Public Health, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, United Kingdom
| | - Najaf Amin
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Claudine Berr
- INSERM U1061, Faculty of Medicine, Hôpital La Colombière, Montpellier, France
| | - Oscar L. Lopez
- Departments of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Philip L. De Jager
- Program in Translational NeuroPsychiatric Genomics, Institute for the Neurosciences, Department of Neurology & Psychiatry, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- Program in Medical and Population Genetics, Broad Institute, Boston, Massachusetts, United States of America
| | - Vincent Deramecourt
- Université Lille 2, Lille, France
- CNR-MAJ, Centre Hospitalier Régional Universitaire de Lille, Lille, France
| | - Janet A. Johnston
- Ageing Group, Centre for Public Health, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, United Kingdom
| | - Denis Evans
- Rush Institute for Healthy Aging, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, United States of America
| | - Simon Lovestone
- King's College London, Institute of Psychiatry, Department of Neuroscience, De Crespigny Park, Denmark Hill, London, United Kingom
| | - Luc Letenneur
- Inserm U897, Victor Segalen University, F-33076, Bordeaux, France
| | - Isabel Hernández
- Memory Clinic of Fundació ACE. Institut Català de Neurociències Aplicades, Barcelona, Spain
| | - David C. Rubinsztein
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | | | - Kristel Sleegers
- Neurodegenerative Brain Diseases Group, Department of Molecular Genetics, VIB, Antwerp, Belgium
- Laboratory of Neurogenetics, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Alison M. Goate
- Department of Psychiatry and Hope Center Program on Protein Aggregation and Neurodegeneration, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Nathalie Fiévet
- Inserm U744, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Matthew J. Huentelman
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, Arizona, United States of America
| | - Michael Gill
- Discipline of Psychiatry, Trinity College, Dublin, Ireland
| | - Kristelle Brown
- Institute of Genetics, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom
| | - M. Ilyas Kamboh
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Alzheimer's Disease Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Lina Keller
- Aging Reasearch Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | - Pascale Barberger-Gateau
- King's College London, Institute of Psychiatry, Department of Neuroscience, De Crespigny Park, Denmark Hill, London, United Kingom
| | - Bernadette McGuinness
- Ageing Group, Centre for Public Health, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, United Kingdom
| | - Eric B. Larson
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
- Group Health Research Institute, Group Health, Seattle, Washington, United States of America
| | - Amanda J. Myers
- Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| | - Carole Dufouil
- Inserm U897, Victor Segalen University, F-33076, Bordeaux, France
| | - Stephen Todd
- Ageing Group, Centre for Public Health, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, United Kingdom
| | - David Wallon
- CNR-MAJ, Inserm U1079, Rouen University Hospital, 76031 France, Rouen, France
| | - Seth Love
- University of Bristol Institute of Clinical Neurosciences, School of Clinical Sciences, Frenchay Hospital, Bristol, United Kingdom
| | - Ekaterina Rogaeva
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, Ontario, Canada
| | - John Gallacher
- Institute of Primary Care and Public Health, Cardiff University, Neuadd Meirionnydd, University Hospital of Wales, Heath Park, Cardiff, United Kingdom
| | - Peter St George-Hyslop
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, Ontario, Canada
- Cambridge Institute for Medical Research and Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Jordi Clarimon
- Neurology Department. IIB Sant Pau. Sant Pau Hospital. Universitat Autònoma de Barcelona, Barcelona, Spain
- Center for Networker Biomedical Research in Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Alberto Lleo
- Neurology Department. IIB Sant Pau. Sant Pau Hospital. Universitat Autònoma de Barcelona, Barcelona, Spain
- Center for Networker Biomedical Research in Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Anthony Bayer
- Institute of Primary Care and Public Health, Cardiff University, Neuadd Meirionnydd, University Hospital of Wales, Heath Park, Cardiff, United Kingdom
| | - Debby W. Tsuang
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, Washington, United States of America
| | - Lei Yu
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, United States of America
| | - Magda Tsolaki
- 3rd Department of Neurology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Paola Bossù
- Clinical and Behavioral Neurology, Fondazione Santa Lucia, Roma, Italy
| | | | - Petra Proitsi
- King's College London, Institute of Psychiatry, Department of Neuroscience, De Crespigny Park, Denmark Hill, London, United Kingom
| | - John Collinge
- MRC Prion Unit, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - Sandro Sorbi
- NEUROFARBA Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
- Centro di Ricerca, Trasferimento e Alta Formazione DENOTHE, University of Florence, Florence, Italy
| | | | - Nick C. Fox
- Dementia Research Center, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - John Hardy
- Department of Molecular Neuroscience and Reta Lilla Weston Laboratories, Institute of Neurology, London, United Kingdom
| | | | - Paolo Bosco
- IRCCS Associazione Oasi Maria SS, Troina, Italy
| | - Robert Clarke
- Oxford Healthy Aging Project (OHAP), Clinical Trial Service Unit, University of Oxford, Oxford, United Kingdom
| | - Carol Brayne
- Cognitive Function and Ageing Study (CFAS), Institute of Public Health, University of Cambridge, Cambridge, United Kingdom
| | - Daniela Galimberti
- University of Milan, Fondazione Cà Granda, IRCCS Ospedale Policlinico, Milan, Italy
| | - Elio Scarpini
- University of Milan, Fondazione Cà Granda, IRCCS Ospedale Policlinico, Milan, Italy
| | | | | | | | - Susanne Moebus
- Urban Epidemiology, Institute for Medical Informatics, Biometry and Epidemiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Patrizia Mecocci
- Section of Gerontology and Geriatrics, Department of Clinical and Experimental Medicine, University of Perugia, Perugia, Italy
| | - Maria Del Zompo
- Section of Neuroscience and Clinical Pharmacology, Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Wolfgang Maier
- Department of Psychiatry and Psychotherapy, University of Bonn, Germany and German Center for Neurodegenerative Diseases (DZNE, Bonn), Bonn, Germany
| | - Harald Hampel
- Department of Psychiatry, University of Frankfurt am Main, Frankfurt am Main, Germany (H.H.)
- Department of Psychiatry, Ludwig-Maximilians University, Munich, Germany
| | - Alberto Pilotto
- Gerontology and Geriatrics Research Laboratory, I.R.C.C.S. Casa Sollievo della Sofferenza, San Giovanni Rotondo (FG), Italy
| | - Ana Frank-García
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM); Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Instituto de Investigación Sanitaria “Hospital la Paz” (IdIPaz), Madrid, Spain
| | - Francesco Panza
- Department of Geriatrics,Center for Aging Brain,University of Bari, Bari, Italy
| | - Vincenzo Solfrizzi
- Department of Geriatrics,Center for Aging Brain,University of Bari, Bari, Italy
| | - Paolo Caffarra
- Department of Neuroscience-University of Parma, Parma, Italy
- Center for Cognitive Disorders AUSL, Parma, Italy
| | - Benedetta Nacmias
- NEUROFARBA Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
- Centro di Ricerca, Trasferimento e Alta Formazione DENOTHE, University of Florence, Florence, Italy
| | - William Perry
- The John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida, United States of America
- Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami, Miami, Florida, United States of America
| | - Manuel Mayhaus
- Department Of Psychiatry, University Hospital, Saarland, Germany
| | - Lars Lannfelt
- Department of Public Health/Geriatrics, Uppsala University, Uppsala, Sweden
| | - Hakon Hakonarson
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - Sabrina Pichler
- Department Of Psychiatry, University Hospital, Saarland, Germany
| | | | - Martin Ingelsson
- Department of Public Health/Geriatrics, Uppsala University, Uppsala, Sweden
| | - Duane Beekly
- National Alzheimer's Coordinating Center, University of Washington, Seattle, Washington, United States of America
| | | | - Fanggeng Zou
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, United States of America
| | - Otto Valladares
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Steven G. Younkin
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, United States of America
| | | | - Kara L. Hamilton-Nelson
- The John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida, United States of America
| | - Wei Gu
- Department of Psychiatry, University Hospital, Saarland, Germany
| | - Cristina Razquin
- Neurogenetics Laboratory, Division of Neurosciences, Center for Applied Medical Research, University of Navarra School of Medicine, Pamplona, Spain
| | - Pau Pastor
- Neurogenetics Laboratory, Division of Neurosciences, Center for Applied Medical Research, University of Navarra School of Medicine, Pamplona, Spain
- CIBERNED, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | - Ignacio Mateo
- Neurology Service and CIBERNED, "Marqués de Valdecilla" University Hospital (University of Cantabria and IFIMAV), Santander, Spain
| | - Michael J. Owen
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, United Kingdom
| | - Kelley M. Faber
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis, Indiana, United States of America
| | - Palmi V. Jonsson
- University of Iceland, Faculty of Medicine, Reykjavik, Iceland
- Landspitali University Hospital, Reykjavik, Iceland
| | - Onofre Combarros
- Neurology Service and CIBERNED, "Marqués de Valdecilla" University Hospital (University of Cantabria and IFIMAV), Santander, Spain
| | - Michael C. O'Donovan
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, United Kingdom
| | - Laura B. Cantwell
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Hilkka Soininen
- Institute of Clinical Medicine - Neurology, University of Eastern Finland, Kuopio, Finland
- Department of Neurology, Kuopio University Hospital, Kuopio, Finland
| | - Deborah Blacker
- Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts, United States of America
- Department of Psychiatry, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, United States of America
| | - Simon Mead
- MRC Prion Unit, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - Thomas H. Mosley
- Department of Medicine (Geriatrics), University of Mississippi Medical Center, Jackson, Mississippi, United States of America
| | - David A. Bennett
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, United States of America
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois, United States of America
| | - Tamara B. Harris
- Laboratory of Epidemiology, Demography, and Biometry, National Institute of Health, Bethesda, Maryland, United States of America
| | - Laura Fratiglioni
- Aging Research Center, Department Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
- Department Geriatric Medicine, Genetics Unit, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Clive Holmes
- Division of Clinical Neurosciences, School of Medicine, University of Southampton, Southampton, United Kingdom
| | - Renee F. A. G. de Bruijn
- Departments of Neurology and Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Peter Passmore
- Ageing Group, Centre for Public Health, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, United Kingdom
| | - Thomas J. Montine
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
| | - Karolien Bettens
- Neurodegenerative Brain Diseases Group, Department of Molecular Genetics, VIB, Antwerp, Belgium
- Laboratory of Neurogenetics, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Jerome I. Rotter
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Alexis Brice
- INSERM UMR_S975-CNRS UMR 7225, Université Pierre et Marie Curie, Centre de recherche de l'Institut du Cerveau et de la Moëlle épinière-CRICM, Hôpital de la Salpêtrière, Paris France
- AP-HP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Kevin Morgan
- Institute of Genetics, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom
| | - Tatiana M. Foroud
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis, Indiana, United States of America
| | - Walter A. Kukull
- Department of Epidemiology, University of Washington, Seattle, Washington, United States of America
| | - Didier Hannequin
- CNR-MAJ, Inserm U1079, Rouen University Hospital, 76031 France, Rouen, France
| | - John F. Powell
- King's College London, Institute of Psychiatry, Department of Neuroscience, De Crespigny Park, Denmark Hill, London, United Kingom
| | - Michael A. Nalls
- Laboratory of Neurogenetics, Intramural Research Program, National Institute on Aging, Bethesda, Maryland, United States of America
| | - Karen Ritchie
- INSERM U1061, Faculty of Medicine, Hôpital La Colombière, Montpellier, France
- Imperial College, London, United Kingdom
| | - Kathryn L. Lunetta
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America
| | - John S. K. Kauwe
- Department of Biology, Brigham Young University, Provo, Utah, United States of America
| | - Eric Boerwinkle
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
- Human Genetics Center and Div. of Epidemiology, University of Texas Health Sciences Center at Houston, Houston, Texas, United States of America
| | | | - Mercè Boada
- Memory Clinic of Fundació ACE. Institut Català de Neurociències Aplicades, Barcelona, Spain
- Hospital Universitari Vall d'Hebron - Institut de Recerca, Universitat Autònoma de Barcelona. (VHIR-UAB), Barcelona, Spain
| | - Mikko Hiltunen
- Institute of Clinical Medicine - Neurology, University of Eastern Finland, Kuopio, Finland
- Department of Neurology, Kuopio University Hospital, Kuopio, Finland
| | - Eden R. Martin
- The John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida, United States of America
- Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami, Miami, Florida, United States of America
| | | | - Dan Rujescu
- Department of Psychiatry, Ludwig-Maximilians University, Munich, Germany
| | - Jean-François Dartigues
- Inserm U897, Victor Segalen University, F-33076, Bordeaux, France
- Centre de Mémoire de Ressources et de Recherche de Bordeaux, CHU de Bordeaux, Bordeaux, France
| | - Richard Mayeux
- Taub Institute on Alzheimer's Disease and the Aging Brain, Department of Neurology, Columbia University New York, New York, United States of America
- Gertrude H. Sergievsky Center, Department of Neurology, Columbia University, New York, New York, United States of America
| | | | - Albert Hofman
- Departments of Epidemiology, Neurology and Radiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
- Netherlands Consortium for Healthy Aging, Leiden, The Netherlands
| | - Markus M. Nöthen
- Institute of Human Genetics, Department of Genomics, Life and Brain Center, University of Bonn, and German Center for Neurodegenerative Diseases (DZNE, Bonn), Bonn, Germany
| | - Caroline Graff
- Department Geriatric Medicine, Genetics Unit, Karolinska University Hospital Huddinge, Stockholm, Sweden
- Karolinska Institutet, Department of Neurobiology, Care Sciences and Society, KIADRC, Stockholm, Sweden
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington, United States of America
- Group Health Research Institute, Group Health Cooperative, Seattle, Washington, United States of America
| | - Jonathan L. Haines
- Vanderbilt Center for Human Genetics Research, Vanderbilt University, Nashville, Tennessee, United States of America
- Department of Epidemiology & Biostatistics, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Mark Lathrop
- Department of Ophthalmology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Fondation Jean Dausset- CEPH, Paris, France
- McGill University and Génome Québec Innovation Centre, Montreal, Canada
| | - Margaret A. Pericak-Vance
- The John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida, United States of America
- Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami, Miami, Florida, United States of America
| | - Lenore J. Launer
- Laboratory of Epidemiology, Demography, and Biometry, National Institute of Health, Bethesda, Maryland, United States of America
| | - Christine Van Broeckhoven
- Neurodegenerative Brain Diseases Group, Department of Molecular Genetics, VIB, Antwerp, Belgium
- Laboratory of Neurogenetics, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Lindsay A. Farrer
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Ophthalmology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Epidemiology, Boston University School of Public Health, Boston, Massachusetts, United States of America
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Cornelia M. van Duijn
- Netherlands Consortium for Healthy Aging, Leiden, The Netherlands
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
- Center for Medical Systems Biology, Leiden, The Netherlands
| | - Alfredo Ramirez
- Department of Psychiatry and Psychotherapy and Institute of Human Genetics, University of Bonn, Bonn, Germany
| | - Sudha Seshadri
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- The Framingham Heart Study, Framingham, Massachusetts, United States of America
| | - Gerard D. Schellenberg
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Philippe Amouyel
- Inserm U744, Lille, France
- Université Lille 2, Lille, France
- Institut Pasteur de Lille, Lille, France
- CNR-MAJ, Centre Hospitalier Régional Universitaire de Lille, Lille, France
- Centre Hospitalier Régional Universitaire de Lille, Lille, France
| | - Julie Williams
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
34
|
Abstract
The past year has been extremely successful with regard to the genetics of dystonia, with the identification of four new dystonia genes (CIZ1, ANO3, GNAL, and TUBB4A). This progress was primarily achieved because of the application of a new technology, next-generation DNA sequencing, which allows rapid and comprehensive assessment of a patient's genome. In addition, a combination of next-generation and traditional Sanger sequencing has expanded the phenotypic spectrum associated with some of the dystonia plus (ATP1A3) and paroxysmal (PRRT2) loci. This article reviews the newly identified genes and phenotypes and discusses the future applications of next-generation sequencing to dystonia research.
Collapse
Affiliation(s)
- Tania Fuchs
- Department of Genetics and Genomic Sciences, Ichan School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1498, New York, NY, 10029, USA,
| | | |
Collapse
|
35
|
Gragnoli C. Hypothesis of the neuroendocrine cortisol pathway gene role in the comorbidity of depression, type 2 diabetes, and metabolic syndrome. APPLICATION OF CLINICAL GENETICS 2014; 7:43-53. [PMID: 24817815 PMCID: PMC4012344 DOI: 10.2147/tacg.s39993] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Depression, type 2 diabetes (T2D), and metabolic syndrome (MetS) are often comorbid. Depression per se increases the risk for T2D by 60%. This risk is not accounted for by the use of antidepressant therapy. Stress causes hyperactivation of the hypothalamic–pituitary–adrenal (HPA) axis, by triggering the hypothalamic corticotropin-releasing hormone (CRH) secretion, which stimulates the anterior pituitary to release the adrenocorticotropin hormone (ACTH), which causes the adrenal secretion of cortisol. Depression is associated with an increased level of cortisol, and CRH and ACTH at inappropriately “normal” levels, that is too high compared to their expected lower levels due to cortisol negative feedback. T2D and MetS are also associated with hypercortisolism. High levels of cortisol can impair mood as well as cause hyperglycemia and insulin resistance and other traits typical of T2D and MetS. We hypothesize that HPA axis hyperactivation may be due to variants in the genes of the CRH receptors (CRHR1, CRHR2), corticotropin receptors (or melanocortin receptors, MC1R-MC5R), glucocorticoid receptor (NR3C1), mineralocorticoid receptor (NR3C2), and of the FK506 binding protein 51 (FKBP5), and that these variants may be partially responsible for the clinical association of depression, T2D and MetS. In this review, we will focus on the correlation of stress, HPA axis hyperactivation, and the possible genetic role of the CRHR1, CRHR2, MCR1–5, NR3C1, and NR3C2 receptors and FKBP5 in the susceptibility to the comorbidity of depression, T2D, and MetS. New studies are needed to confirm the hypothesized role of these genes in the clinical association of depression, T2D, and MetS.
Collapse
Affiliation(s)
- Claudia Gragnoli
- Center for Biotechnology and Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA, USA ; Molecular Biology Laboratory, Bios Biotech Multi-Diagnostic Health Center, Rome, Italy
| |
Collapse
|
36
|
Chen HM, DeLong CJ, Bame M, Rajapakse I, Herron TJ, McInnis MG, O'Shea KS. Transcripts involved in calcium signaling and telencephalic neuronal fate are altered in induced pluripotent stem cells from bipolar disorder patients. Transl Psychiatry 2014; 4:e375. [PMID: 25116795 PMCID: PMC3966040 DOI: 10.1038/tp.2014.12] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Accepted: 01/09/2014] [Indexed: 12/12/2022] Open
Abstract
Bipolar disorder (BP) is a chronic psychiatric condition characterized by dynamic, pathological mood fluctuations from mania to depression. To date, a major challenge in studying human neuropsychiatric conditions such as BP has been limited access to viable central nervous system tissue to examine disease progression. Patient-derived induced pluripotent stem cells (iPSCs) now offer an opportunity to analyze the full compliment of neural tissues and the prospect of identifying novel disease mechanisms. We have examined changes in gene expression as iPSC derived from well-characterized patients differentiate into neurons; there was little difference in the transcriptome of iPSC, but BP neurons were significantly different than controls in their transcriptional profile. Expression of transcripts for membrane bound receptors and ion channels was significantly increased in BP-derived neurons compared with controls, and we found that lithium pretreatment of BP neurons significantly altered their calcium transient and wave amplitude. The expression of transcription factors involved in the specification of telencephalic neuronal identity was also altered. Control neurons expressed transcripts that confer dorsal telencephalic fate, whereas BP neurons expressed genes involved in the differentiation of ventral (medial ganglionic eminence) regions. Cells were responsive to dorsal/ventral patterning cues, as addition of the Hedgehog (ventral) pathway activator purmorphamine or a dorsalizing agent (lithium) stimulated expression of NKX2-1 (ventral identity) or EMX2 (dorsal) in both groups. Cell-based models should have a significant impact on our understanding of the genesis and therefore treatment of BP; the iPSC cell lines themselves provide an important resource for comparison with other neurodevelopmental disorders.
Collapse
Affiliation(s)
- H M Chen
- Department of Psychiatry, University of Michigan Medical School, Ann Arbor, MI, USA
| | - C J DeLong
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - M Bame
- Department of Psychiatry, University of Michigan Medical School, Ann Arbor, MI, USA
| | - I Rajapakse
- Center for Computational Medicine & Bioinformatics, Department of Mathematics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - T J Herron
- Department of Cardiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - M G McInnis
- Department of Psychiatry, University of Michigan Medical School, Ann Arbor, MI, USA
| | - K S O'Shea
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA,Department of Cell and Developmental Biology, University of Michigan Medical School, 3051 BSRB, 109 Zina Pitcher Pl, Ann Arbor, MI 48109, USA. E-mail:
| |
Collapse
|
37
|
Jan WC, Yang SY, Chuang LC, Lu RB, Lu MK, Sun HS, Kuo PH. Exploring the associations between genetic variants in genes encoding for subunits of calcium channel and subtypes of bipolar disorder. J Affect Disord 2014; 157:80-6. [PMID: 24581832 DOI: 10.1016/j.jad.2013.12.044] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 12/31/2013] [Accepted: 12/31/2013] [Indexed: 01/11/2023]
Abstract
BACKGROUND Associations of two voltage-gated calcium channel (Cav) genes, CACNA1C and CACNB2, were identified for bipolar disorder (BP) in different ethnic groups in recent genome-wide association studies. The current study aimed to evaluate the associations of several Cav genes and subtypes of BP in genetically more homogeneous Taiwanese samples. Additionally, we tested interaction effects among genes that encode for α1, β and γ-subunits of calcium channel. METHODS 8 Cav genes were selected based on evidence in prior association studies and significant linkage regions for BP. 280 BP patients and 200 controls were recruited. Multifactor dimensionality reduction was performed for interaction testing in these discovery samples. Replication was conducted for two markers using additional 495 Taiwanese cases and 1341 controls. RESULTS Weak associations for CACNA1C (rs10848635), CACNA1E (rs10848635), CACNB2 (rs11013860), and CACNG2 (rs2284018) genes were observed. Joint analysis of four markers revealed higher accumulative risk with increasing numbers of risk genotypes an individual endorsed for BP-I (Ptrend=0.006) and BP-II (Ptrend=0.017) disorders. Combined analysis with independent replication samples further supported the association of rs11013860 in CACNB2 with BP subtype I (P=1×10(-6)). Suggestive interactions were found between genes encoded for different subunits of calcium channel (α1, β, and γ). LIMITATIONS Moderate sample size and incomplete markers coverage for the chosen Cav genes. CONCLUSIONS Our results support the involvement of different calcium channel genes in bipolar illness, in particular the beta-subunit in the Asian population. Further investigation of functional property of these genes can contribute on understanding the etiological mechanisms of bipolar illness.
Collapse
Affiliation(s)
- Wen-Chi Jan
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taiwan
| | - Shi-Yi Yang
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taiwan
| | - Li-Chung Chuang
- Department of Nursing, Cardinal Tien College of Healthcare & Management, I-Lan, Taiwan
| | - Ru-Band Lu
- Department of Psychiatry, National Cheng Kung University and Hospital, Taiwan
| | - Ming-Kun Lu
- Department of Health, Jia Nan Mental Hospital, Taiwan
| | - H Sunny Sun
- Institute of Molecular Medicine, National Cheng Kung University, Taiwan
| | - Po-Hsiu Kuo
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taiwan; Research Center for Genes, Environment and Human Health, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
38
|
Revisiting DARPP-32 in postmortem human brain: changes in schizophrenia and bipolar disorder and genetic associations with t-DARPP-32 expression. Mol Psychiatry 2014; 19:192-9. [PMID: 23295814 DOI: 10.1038/mp.2012.174] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Revised: 10/04/2012] [Accepted: 10/15/2012] [Indexed: 12/23/2022]
Abstract
Dopamine- and cAMP-regulated phosphoprotein of molecular weight 32 kDa (DARPP-32 or PPP1R1B) has been of interest in schizophrenia owing to its critical function in integrating dopaminergic and glutaminergic signaling. In a previous study, we identified single-nucleotide polymorphisms (SNPs) and a frequent haplotype associated with cognitive and imaging phenotypes that have been linked with schizophrenia, as well as with expression of prefrontal cortical DARPP-32 messenger RNA (mRNA) in a relatively small sample of postmortem brains. In this study, we examined the association of expression of two major DARPP-32 transcripts, full-length (FL-DARPP-32) and truncated (t-DARPP-32), with genetic variants of DARPP-32 in three brain regions receiving dopaminergic input and implicated in schizophrenia (the dorsolateral prefrontal cortex (DLPFC), hippocampus and caudate) in a much larger set of postmortem samples from patients with schizophrenia, bipolar disorder, major depression and normal controls (>700 subjects). We found that the expression of t-DARPP-32 was increased in the DLPFC of patients with schizophrenia and bipolar disorder, and was strongly associated with genotypes at SNPs (rs879606, rs90974 and rs3764352), as well as the previously identified 7-SNP haplotype related to cognitive functioning. The genetic variants that predicted worse cognitive performance were associated with higher t-DARPP-32 expression. Our results suggest that variation in PPP1R1B affects the abundance of the splice variant t-DARPP-32 mRNA and may reflect potential molecular mechanisms implicated in schizophrenia and affective disorders.
Collapse
|
39
|
Daviss WB, O'Donnell L, Soileau BT, Heard P, Carter E, Pliszka SR, Gelfond JAL, Hale DE, Cody JD. Mood disorders in individuals with distal 18q deletions. Am J Med Genet B Neuropsychiatr Genet 2013; 162B:879-88. [PMID: 24006251 DOI: 10.1002/ajmg.b.32197] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Accepted: 08/02/2013] [Indexed: 12/27/2022]
Abstract
We examined 36 participants at least 4 years old with hemizygous distal deletions of the long arm of Chromosome 18 (18q-) for histories of mood disorders and to characterize these disorders clinically. Since each participant had a different region of 18q hemizygosity, our goal was also to identify their common region of hemizygosity associated with mood disorders; thereby identifying candidate causal genes in that region. Lifetime mood and other psychiatric disorders were determined by semi-structured interviews of patients and parents, supplemented by reviews of medical and psychiatric records, and norm-referenced psychological assessment instruments, for psychiatric symptoms, cognitive problems, and adaptive functioning. Sixteen participants were identified with lifetime mood disorders (ages 12-42 years, 71% female, 14 having had unipolar depression and 2 with bipolar disorders). From the group of 20 who did not meet criteria for a mood disorder; a comparison group of 6 participants were identified who were matched for age range and deletion size. Mood-disordered patients had high rates of anxiety (75%) and externalizing behavior disorders (44%), and significant mean differences from comparison patients (P < 0.05), including higher overall and verbal IQs and lower autistic symptoms. A critical region was defined in the mood-disordered group that included a hypothetical gene, C18orf62, and two known genes, ZADH2 and TSHZ1. We conclude that patients having terminal deletions of this critical region of the long arm of Chromosome 18 are highly likely to have mood disorders, which are often comorbid with anxiety and to a lesser extent with externalizing disorders.
Collapse
Affiliation(s)
- William B Daviss
- Department of Psychiatry, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Cruceanu C, Ambalavanan A, Spiegelman D, Gauthier J, Lafrenière RG, Dion PA, Alda M, Turecki G, Rouleau GA. Family-based exome-sequencing approach identifies rare susceptibility variants for lithium-responsive bipolar disorder. Genome 2013; 56:634-40. [DOI: 10.1139/gen-2013-0081] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Bipolar disorder (BD) is a psychiatric condition characterized by the occurrence of at least two episodes of clinically disturbed mood including mania and depression. A vast literature describing BD studies suggests that a strong genetic contribution likely underlies this condition; heritability is estimated to be as high as 80%. Many studies have identified BD susceptibility loci, but because of the genetic and phenotypic heterogeneity observed across individuals, very few loci were subsequently replicated. Research in BD genetics to date has consisted of classical linkage or genome-wide association studies, which have identified candidate genes hypothesized to present common susceptibility variants. Although the observation of such common variants is informative, they can only explain a small fraction of the predicted BD heritability, suggesting a considerable contribution would come from rare and highly penetrant variants. We are seeking to identify such rare variants, and to increase the likelihood of being successful, we aimed to reduce the phenotypic heterogeneity factor by focusing on a well-defined subphenotype of BD: excellent response to lithium monotherapy. Our group has previously shown positive response to lithium therapy clusters in families and has a consistent clinical presentation with minimal comorbidity. To identify such rare variants, we are using a targeted exome capture and high-throughput DNA sequencing approach, and analyzing the entire coding sequences of BD affected individuals from multigenerational families. We are prioritizing rare variants with a frequency of less than 1% in the population that segregate with affected status within each family, as well as being potentially highly penetrant (e.g., protein truncating, missense, or frameshift) or functionally relevant (e.g., 3′UTR, 5′UTR, or splicing). By focusing on rare variants in a familial cohort, we hope to explain a significant portion of the missing heritability in BD, as well as to narrow our current insight on the key biochemical pathways implicated in this complex disorder.
Collapse
Affiliation(s)
- Cristiana Cruceanu
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- McGill Group for Suicide Studies, McGill University, Montréal, QC, Canada
| | - Amirthagowri Ambalavanan
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- Center of Excellence in Neuroscience of the Université de Montréal-CENUM, Centre de Recherche du Centre Hospitalier de l’Université de Montréal-CRCHUM, University of Montreal, Montréal, QC, Canada
| | - Dan Spiegelman
- Center of Excellence in Neuroscience of the Université de Montréal-CENUM, Centre de Recherche du Centre Hospitalier de l’Université de Montréal-CRCHUM, University of Montreal, Montréal, QC, Canada
| | - Julie Gauthier
- Center of Excellence in Neuroscience of the Université de Montréal-CENUM, Centre de Recherche du Centre Hospitalier de l’Université de Montréal-CRCHUM, University of Montreal, Montréal, QC, Canada
| | - Ronald G. Lafrenière
- Center of Excellence in Neuroscience of the Université de Montréal-CENUM, Centre de Recherche du Centre Hospitalier de l’Université de Montréal-CRCHUM, University of Montreal, Montréal, QC, Canada
| | - Patrick A. Dion
- Center of Excellence in Neuroscience of the Université de Montréal-CENUM, Centre de Recherche du Centre Hospitalier de l’Université de Montréal-CRCHUM, University of Montreal, Montréal, QC, Canada
| | - Martin Alda
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
| | - Gustavo Turecki
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- McGill Group for Suicide Studies, McGill University, Montréal, QC, Canada
| | - Guy A. Rouleau
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- Center of Excellence in Neuroscience of the Université de Montréal-CENUM, Centre de Recherche du Centre Hospitalier de l’Université de Montréal-CRCHUM, University of Montreal, Montréal, QC, Canada
- Montreal Neurological Institute and Hospital, McGill University, Montréal, QC, Canada
| |
Collapse
|
41
|
Greenwood TA, Badner JA, Byerley W, Keck PE, McElroy SL, Remick RA, Sadovnick AD, Akiskal HS, Kelsoe JR. Heritability and genome-wide SNP linkage analysis of temperament in bipolar disorder. J Affect Disord 2013; 150:1031-40. [PMID: 23759419 PMCID: PMC3759543 DOI: 10.1016/j.jad.2013.05.035] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 05/13/2013] [Accepted: 05/15/2013] [Indexed: 01/30/2023]
Abstract
BACKGROUND The many attempts to identify genes for bipolar disorder (BD) have met with limited success, which has generally been attributed to genetic heterogeneity and small gene effects. However, it is also possible that the categorical phenotypes used in genetic studies of BD are not the most informative or biologically relevant. We have explored aspects of temperament as quantitative phenotypes for BD through the use of the Temperament Evaluation of Memphis, Pisa, Paris, and San Diego Auto-questionnaire (TEMPS-A), which is designed to assess lifelong, milder aspects of bipolar symptomatology and defines five temperaments: hyperthymic, dysthymic, cyclothymic, irritable, and anxious. METHODS We compared temperament scores between diagnostic groups and assessed heritability in a sample of 101 families collected for genetic studies of BD. A genome-wide SNP linkage study was then performed in the subset of 51 families for which genetic data was available. RESULTS Significant group differences were observed between BD subjects, their first-degree relatives, and independent controls, and all five temperaments were found to be significantly heritable, with heritabilities ranging from 21% for the hyperthymic to 52% for the irritable temperaments. Suggestive evidence for linkage was observed for the hyperthymic (chromosomes 1q44, 2p16, 6q16, and 14q23), dysthymic (chromosomes 3p21 and 13q34), and irritable (chromosome 6q24) temperaments. LIMITATIONS The relatively small size of our linkage sample likely limited our ability to reach genome-wide significance in this study. CONCLUSIONS While not genome-wide significant, these results suggest that aspects of temperament may prove useful in the identification of genes underlying BD susceptibility.
Collapse
Affiliation(s)
- Tiffany A Greenwood
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Koyama Y, Hattori T, Shimizu S, Taniguchi M, Yamada K, Takamura H, Kumamoto N, Matsuzaki S, Ito A, Katayama T, Tohyama M. DBZ (DISC1-binding zinc finger protein)-deficient mice display abnormalities in basket cells in the somatosensory cortices. J Chem Neuroanat 2013; 53:1-10. [PMID: 23912123 DOI: 10.1016/j.jchemneu.2013.07.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Revised: 07/22/2013] [Accepted: 07/24/2013] [Indexed: 10/26/2022]
Abstract
Disrupted-in-schizophrenia 1 (DISC1)-binding zinc finger protein (DBZ) is a DISC1-interacting molecule and the interaction between DBZ and DISC1 is involved in neurite outgrowth in vitro. DBZ is highly expressed in brain, especially in the cortex. However, the physiological roles of DBZ in vivo have not been clarified. Here, we show that development of basket cells, a morphologically defined class of parvalbumin (PV)-containing interneurons, is disturbed in DBZ knockout (KO) mice. DBZ mRNA was highly expressed in the ventral area of the subventricular zone of the medial ganglionic eminence, where PV-containing cortical interneurons were generated, at embryonic 14.5 days (E14.5). Although the expression level for PV and the number of PV-containing interneurons were not altered in the cortices of DBZ KO mice, basket cells were less branched and had shorter processes in the somatosensory cortices of DBZ KO mice compared with those in the cortices of WT mice. Furthermore, in the somatosensory cortices of DBZ KO mice, the level of mRNAs for the gamma-aminobutyric acid-synthesizing enzymes GAD67 was decreased. These findings show that DBZ is involved in the morphogenesis of basket cells.
Collapse
Affiliation(s)
- Yoshihisa Koyama
- Department of Anatomy and Neuroscience, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
Dystonia is a common movement disorder seen by neurologists in clinic. Genetic forms of the disease are important to recognize clinically and also provide valuable information about possible pathogenic mechanisms within the wider disorder. In the past few years, with the advent of new sequencing technologies, there has been a step change in the pace of discovery in the field of dystonia genetics. In just over a year, four new genes have been shown to cause primary dystonia (CIZ1, ANO3, TUBB4A and GNAL), PRRT2 has been identified as the cause of paroxysmal kinesigenic dystonia and other genes, such as SLC30A10 and ATP1A3, have been linked to more complicated forms of dystonia or new phenotypes. In this review, we provide an overview of the current state of knowledge regarding genetic forms of dystonia—related to both new and well-known genes alike—and incorporating genetic, clinical and molecular information. We discuss the mechanistic insights provided by the study of the genetic causes of dystonia and provide a helpful clinical algorithm to aid clinicians in correctly predicting the genetic basis of various forms of dystonia.
Collapse
Affiliation(s)
- Gavin Charlesworth
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | | | | |
Collapse
|
44
|
Bernstein HG, Dobrowolny H, Schott BH, Gorny X, Becker V, Steiner J, Seidenbecher CI, Bogerts B. Increased density of AKAP5-expressing neurons in the anterior cingulate cortex of subjects with bipolar disorder. J Psychiatr Res 2013; 47:699-705. [PMID: 23462372 DOI: 10.1016/j.jpsychires.2012.12.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Revised: 12/12/2012] [Accepted: 12/17/2012] [Indexed: 12/17/2022]
Abstract
Brain anatomical abnormalities as well as cognitive and emotional processing deficits have been reported for the prefrontal cortex in bipolar disorder, which are in part attributable to cellular and laminar abnormalities in postsynaptic protein expression. A kinase anchoring protein (AKAP) 5/79 plays a key role in postsynaptic signalling of excitatory synapses. We aimed to reveal if the cellular expression of AKAP5/79 protein is altered in the anterior cingulate cortex and the dorsolateral prefrontal cortex in bipolar disorder. Ten subjects with bipolar disorder and ten control cases were investigated by use of immunohistochemical and morphometric techniques. Compared with controls in subjects with bipolar disorder, the numerical density of AKAP5-expressing neurons was significantly increased in the left (p = 0.002) and right (p = 0.008) anterior cingulate cortex. Layer-specific counting revealed that left side layers II (p = 0.000), III (p = 0.001) and V (p = 0.005) as well as right side layers III (p = 0.007), IV (p = 0.007) and V (p = 0.004) had significantly increased AKAP5-positive cell densities in bipolar disorder. In contrast, no statistically significant differences were found for the dorsolateral prefrontal cortex. However, we observed a more intense intraneuronal immunostaining in both prefrontal areas in bipolar disorder patients. Elevated cell numbers and increased intracellular expression of AKAP, together with the altered expression patterns of most intracellular interaction partners of this protein in bipolar disorder as known from the literature, might point to disease-related abnormalities of the AKAP-associated signalosome in prefrontal cortex neurons.
Collapse
|
45
|
Li JM, Lu CL, Cheng MC, Luu SU, Hsu SH, Chen CH. Genetic analysis of the DLGAP1 gene as a candidate gene for schizophrenia. Psychiatry Res 2013; 205:13-7. [PMID: 22940546 DOI: 10.1016/j.psychres.2012.08.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Revised: 06/06/2012] [Accepted: 08/15/2012] [Indexed: 12/21/2022]
Abstract
Schizophrenia is a severe chronic mental disorder with high genetic components in its etiology. Several studies indicated that synaptic dysfunction is involved in the pathophysiology of schizophrenia. Postsynaptic synapse-associated protein 90/postsynaptic density 95-associated proteins (SAPAPs) constitute a part of the N-methyl-d-aspartate receptor-associated postsynaptic density proteins, and are involved in synapse formation. We hypothesized that genetic variants of the SAPAPs might be associated with schizophrenia. Thus, we systemically sequenced all the exons of the discs, large (Drosophila) homolog-associated protein 1 (DLGAP1) gene that encodes SAPAP1 in a sample of 121 schizophrenic patients and 120 controls from Taiwan. We totally identified six genetic variants, including five known SNPs (rs145691437, rs3786431, rs201567254, rs3745051 and rs11662259) and one rare missense mutation (c.1922A>G) in this sample. SNP- and haplotype-based analyses showed no association of these SNPs with schizophrenia. The c.1922A>G mutation that changes the amino acid lysine to arginine at codon 641 was found in one out of 121 patients, but not in 275 control subjects, suggesting it might be a patient-specific mutation. Nevertheless, bioinformatic analysis showed this mutation does not affect the function of the DLGAP1 gene and appears to be a benign variant. Hence, its relationship with the pathogenesis remains to be investigated.
Collapse
Affiliation(s)
- Jun-Ming Li
- Department of Psychiatry, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan
| | | | | | | | | | | |
Collapse
|
46
|
Nissen S, Liang S, Shehktman T, Kelsoe JR, Greenwood TA, Nievergelt CM, McKinney R, Shilling PD, Smith EN, Schork NJ, Bloss CS, Nurnberger JI, Edenberg HJ, Foroud T, Koller DL, Gershon ES, Liu C, Badner JA, Scheftner WA, Lawson WB, Nwulia EA, Hipolito M, Coryell W, Rice J, Byerley W, McMahon FJ, Berrettini WH, Potash JB, Zandi PP, Mahon PB, McInnis MG, Zöllner S, Zhang P, Craig DW, Szelinger S, Barrett TB, Schulze TG. Evidence for association of bipolar disorder to haplotypes in the 22q12.3 region near the genes stargazin, IFT27 and parvalbumin. Am J Med Genet B Neuropsychiatr Genet 2012; 159B:941-50. [PMID: 23038240 PMCID: PMC3665332 DOI: 10.1002/ajmg.b.32099] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Accepted: 08/22/2012] [Indexed: 12/13/2022]
Abstract
We have previously reported genome-wide significant linkage of bipolar disorder to a region on 22q12.3 near the marker D22S278. Towards identifying the susceptibility gene, we have conducted a fine-mapping association study of the region in two independent family samples, an independent case-control sample and a genome-wide association dataset. Two hundred SNPs were first examined in a 5 Mb region surrounding the D22S278 marker in a sample of 169 families and analyzed using PLINK. The peak of association was a haplotype near the genes stargazin (CACNG2), intraflagellar transport protein homolog 27 (IFT27) and parvalbumin (PVALB; P = 4.69 × 10(-4)). This peak overlapped a significant haplotype in a family based association study of a second independent sample of 294 families (P = 1.42 × 10(-5)). Analysis of the combined family sample yielded statistically significant evidence of association to a rare three SNP haplotype in the gene IFT27 (P = 8.89 × 10(-6)). Twelve SNPs comprising these haplotypes were genotyped in an independent sample of 574 bipolar I cases and 550 controls. Statistically significant association was found for a haplotype window that overlapped the region from the first two family samples (P = 3.43 × 10(-4)). However, analyses of the two family samples using the program LAMP, found no evidence for association in this region, but did yield significant evidence for association to a haplotype 3' of CACNG2 (P = 1.76 × 10(-6)). Furthermore, no evidence for association was found in a large genome-wide association dataset. The replication of association to overlapping haplotypes in three independent datasets suggests the presence of a bipolar disorder susceptibility gene in this region.
Collapse
Affiliation(s)
- Stephanie Nissen
- Department of Psychiatry, University of California San Diego, La Jolla 92093, California
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Anglin RES, Mazurek MF, Tarnopolsky MA, Rosebush PI. The mitochondrial genome and psychiatric illness. Am J Med Genet B Neuropsychiatr Genet 2012; 159B:749-59. [PMID: 22887963 DOI: 10.1002/ajmg.b.32086] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Accepted: 07/09/2012] [Indexed: 02/01/2023]
Abstract
Psychiatric disorders are a leading cause of morbidity and mortality, yet their underlying pathophysiology remains unclear. Searches for a genetic cause of bipolar disorder, schizophrenia, and major depressive disorder have yielded inconclusive results. There is increasing interest in the possibility that defects in the mitochondrial genome may play an important role in psychiatric illness. We undertook a review of the literature investigating mitochondria and adult psychiatric disorders. MEDLINE, PsycINFO, and EMBASE were searched from their inception through September 2011, and the reference lists of identified articles were reviewed for additional studies. While multiple lines of evidence, including clinical, genetic, ultrastructural, and biochemical studies, support the involvement of mitochondria in the pathophysiology of psychiatric illness, many studies have methodological limitations and their findings have not been replicated. Clinical studies suggest that psychiatric features can be prominent, and the presenting features of mitochondrial disorders. There is limited but inconsistent evidence for the involvement of mitochondrial DNA haplogroups and mitochondria-related nuclear gene polymorphisms, and for mitochondrial ultrastructural and biochemical abnormalities in psychiatric illness. The current literature suggests that mitochondrial dysfunction and mitochondrial genetic variations may play an important role in psychiatric disorders, but additional methodologically rigorous and adequately powered studies are needed before definitive conclusions can be drawn.
Collapse
Affiliation(s)
- Rebecca E S Anglin
- Department of Psychiatry & Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada.
| | | | | | | |
Collapse
|
48
|
Imprinted DLK1-DIO3 region of 14q32 defines a schizophrenia-associated miRNA signature in peripheral blood mononuclear cells. Mol Psychiatry 2012; 17:827-40. [PMID: 21727898 PMCID: PMC3404364 DOI: 10.1038/mp.2011.78] [Citation(s) in RCA: 178] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
MicroRNAs (miRNAs) regulate gene expression at the post-transcriptional level and are important for coordinating nervous system development and neuronal function in the mature brain. We have recently identified schizophrenia-associated alteration of cortical miRNA biogenesis and expression in post-mortem brain tissue with implications for the dysregulation of schizophrenia candidate genes. Although these changes were observed in the central nervous system, it is plausible that schizophrenia-associated miRNA expression signatures may also be detected in non-neural tissue. To explore this possibility, we investigated the miRNA expression profile of peripheral blood mononuclear cells (PBMCs) from 112 patients with schizophrenia and 76 non-psychiatric controls. miRNA expression analysis of total RNA conducted using commercial miRNA arrays revealed that 33 miRNAs were significantly downregulated after correction for multiple testing with a false discovery rate (FDR) of 0%, which increased to 83 when we considered miRNA with an FDR<5%. Seven miRNAs altered in microarray analysis of schizophrenia were also confirmed to be downregulated by quantitative real-time reverse transcription-polymerase chain reaction. A large subgroup consisting of 17 downregulated miRNAs is transcribed from a single imprinted locus at the maternally expressed DLK1-DIO3 region on chromosome 14q32. This pattern of differentially expressed miRNA in PBMCs may be indicative of significant underlying genetic or epigenetic alteration associated with schizophrenia.
Collapse
|
49
|
Saul MC, Gessay GM, Gammie SC. A new mouse model for mania shares genetic correlates with human bipolar disorder. PLoS One 2012; 7:e38128. [PMID: 22675514 PMCID: PMC3366954 DOI: 10.1371/journal.pone.0038128] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Accepted: 05/04/2012] [Indexed: 11/18/2022] Open
Abstract
Bipolar disorder (BPD) is a debilitating heritable psychiatric disorder. Contemporary rodent models for the manic pole of BPD have primarily utilized either single locus transgenics or treatment with psychostimulants. Our lab recently characterized a mouse strain termed Madison (MSN) that naturally displays a manic phenotype, exhibiting elevated locomotor activity, increased sexual behavior, and higher forced swimming relative to control strains. Lithium chloride and olanzapine treatments attenuate this phenotype. In this study, we replicated our locomotor activity experiment, showing that MSN mice display generationally-stable mania relative to their outbred ancestral strain, hsd:ICR (ICR). We then performed a gene expression microarray experiment to compare hippocampus of MSN and ICR mice. We found dysregulation of multiple transcripts whose human orthologs are associated with BPD and other psychiatric disorders including schizophrenia and ADHD, including: Epor, Smarca4, Cmklr1, Cat, Tac1, Npsr1, Fhit, and P2rx7. RT-qPCR confirmed dysregulation for all of seven transcripts tested. Using a novel genome enrichment algorithm, we found enrichment in genome regions homologous to human loci implicated in BPD in replicated linkage studies including homologs of human cytobands 1p36, 3p14, 3q29, 6p21–22, 12q24, 16q24, and 17q25. Using a functional network analysis, we found dysregulation of a gene system related to chromatin packaging, a result convergent with recent human findings on BPD. Our findings suggest that MSN mice represent a polygenic model for the manic pole of BPD showing much of the genetic systems complexity of the corresponding human disorder. Further, the high degree of convergence between our findings and the human literature on BPD brings up novel questions about evolution by analogy in mammalian genomes.
Collapse
Affiliation(s)
- Michael C Saul
- Department of Zoology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America.
| | | | | |
Collapse
|
50
|
Fanous AH, Middleton FA, Gentile K, Amdur RL, Maher BS, Zhao Z, Sun J, Medeiros H, Carvalho C, Ferreira SR, Macedo A, Knowles JA, Azevedo MH, Pato MT, Pato CN. Genetic overlap of schizophrenia and bipolar disorder in a high-density linkage survey in the Portuguese Island population. Am J Med Genet B Neuropsychiatr Genet 2012; 159B:383-91. [PMID: 22461138 DOI: 10.1002/ajmg.b.32041] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2011] [Accepted: 02/16/2012] [Indexed: 11/06/2022]
Abstract
Recent family and genome-wide association studies strongly suggest shared genetic risk factors for schizophrenia (SZ) and bipolar disorder (BP). However, linkage studies have not been used to test for statistically significant genome-wide overlap between them. Forty-seven Portuguese families with sibpairs concordant for SZ, BP, or psychosis (PSY, which includes either SZ or psychotic BP) were genotyped for over 57,000 markers using the Affymetrix 50K Xba SNP array. NPL and Kong and Cox LOD scores were calculated in Merlin for all three phenotypes. Empirical significance was determined using 1,000 gene-dropping simulations. Significance of genome-wide genetic overlap between SZ and BP was determined by the number of simulated BP scans having the same number of loci jointly linked with the real SZ scan, and vice versa. For all three phenotypes, a number of regions previously linked in this sample remained so. For BP, chromosome 1p36 achieved significance (11.54-15.71 MB, LOD = 3.51), whereas it was not even suggestively linked at lower marker densities, as did chromosome 11q14.1 (89.32-90.15 MB, NPL = 4.15). Four chromosomes had loci at which both SZ and BP had NPL ≥ 1.98, which was more than would be expected by chance (empirical P = 0.01 using simulated SZ scans; 0.07 using simulated BP scans), although they did not necessarily meet criteria for suggestive linkage individually. These results suggest that high-density marker maps may provide greater power and precision in linkage studies than lower density maps. They also further support the hypothesis that SZ and BP share at least some risk alleles.
Collapse
Affiliation(s)
- Ayman H Fanous
- Mental Health Service Line, Washington VA Medical Center, Washington, DC, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|