1
|
Oyadomari WY, Santiago TC, Basso L, Oliveira V, Cruz FC, Nani JV, Hayashi MAF. Long-term treatment with haloperidol modulates angiotensin I-converting enzyme (ACE) activity in transgenic animal model with construct validity for schizophrenia studies. Brain Res 2025; 1859:149640. [PMID: 40228572 DOI: 10.1016/j.brainres.2025.149640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 03/27/2025] [Accepted: 04/09/2025] [Indexed: 04/16/2025]
Abstract
Elevated angiotensin I-converting enzyme (ACE) activity has been correlated with worse cognitive performance in patients with first-episode psychosis (FEP) and chronic schizophrenia (SZ). In this study, we investigated ACE activity in drug-naïve transgenic rats overexpressing the full-length non-mutated human Disrupted-in-Schizophrenia 1 (tgDISC1) compared to wild-type (WT) controls, while we also assessed the effects of long-term treatment with typical antipsychotic haloperidol. Our findings indicated that untreated tgDISC1 rats show elevated serum ACE activity compared to WT animals, which is consistent with clinical observations in drug-naïve FEP patients. In contrast, baseline ACE activity in the brain of tgDISC1 was generally lower than in WT rats, with the exception of no difference in ACE activity observed in brain regions associated with learning, memory, and reward, such as the hippocampus and nucleus accumbens. Consistent with clinical observations in FEP patients following treatment with antipsychotics, 30-days of daily haloperidol-treatment significantly increased serum ACE activity in blood serum of both tgDISC1 and WT rats. However, ACE responses in brain were markedly different, as haloperidol treatment reduced ACE activity in most brain regions of both rat strains. These results support the existence of a central renin-angiotensin system (RAS) distinct from the peripheral RAS, suggesting that the treatment with a dopamine blocker exerts brain-specific effects on ACE activity, which was essentially opposite to that observed in the periphery. This region-specific alterations observed in cognition-related brain areas (notably with a relative stronger effect size in hippocampus and nucleus accumbens of tgDISC1 compared to WT rats) also suggest a critical interplay among dopamine homeostasis, ACE activity, and cognitive deficits in SZ. Understanding this interplay could help identifying novel biomarkers and/or therapeutic strategies for improving cognitive outcomes in SZ patients.
Collapse
Affiliation(s)
- William Y Oyadomari
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Thays C Santiago
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Leonardo Basso
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Vitor Oliveira
- Department of Biophysics, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Fábio C Cruz
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - João V Nani
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil.
| | - Mirian A F Hayashi
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil.
| |
Collapse
|
2
|
Nucifora LG, Ishizuka K, El Demerdash N, Lee BJ, Imai MT, Ayala-Grosso C, Yenokyan G, Cascella NG, Lin S, Schretlen DJ, Harvey PD, Margolis RL, Ross CA, Sawa A, Nucifora FC. Protein aggregation identified in olfactory neuronal cells is associated with cognitive impairments in a subset of living schizophrenia patients. Mol Psychiatry 2025:10.1038/s41380-025-02956-8. [PMID: 40181190 DOI: 10.1038/s41380-025-02956-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 02/02/2025] [Accepted: 03/14/2025] [Indexed: 04/05/2025]
Abstract
Schizophrenia is a heterogeneous disorder, and likely results from multiple pathophysiological mechanisms. Protein aggregation, resulting from disruption of protein homeostasis (proteostasis), has been implicated in many diseases, including cancer, cardiac and pulmonary diseases, muscle diseases, and neurodegenerative disorders, but is a relatively new pathophysiological hypothesis for schizophrenia. Genetic findings implicate proteostasis in schizophrenia, and individual proteins associated with the disorder may undergo aggregation. While there is some evidence of associations between genetic variants and protein aggregation, the extent to which genetic variations influence protein aggregation remains unknown. We have previously reported increased protein insolubility and increased ubiquitination of the insoluble protein fraction, two markers of protein aggregation, in human postmortem brains from a subset of patients with schizophrenia. In the present study, we investigate whether protein aggregation is observed in an independent model system, olfactory neuronal cells derived from living patients with schizophrenia, and examine the relationship between aggregation and patient clinical and cognitive status. We demonstrate that, as in postmortem brain, olfactory neurons from a subset of patients with schizophrenia exhibit protein aggregation, identified by increased protein insolubility and ubiquitination of the insoluble protein fraction, and by ubiquitin positive protein aggregates. Patients with protein aggregation exhibit more severe cognitive deficits than those without aggregation, as revealed by between-group comparisons and correlational analyses. Understanding the mechanisms of the aggregation process, the factors that differentiate individuals who develop aggregates from those who do not, and the relationship between aggregation and cell function, has important implications for the pathophysiology of schizophrenia, and may provide insight into disease heterogeneity and novel therapeutic targets.
Collapse
Affiliation(s)
- Leslie G Nucifora
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Koko Ishizuka
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nagat El Demerdash
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Brian J Lee
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael T Imai
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Carlos Ayala-Grosso
- Unit of Cellular Therapy, Centre of Experimental Medicine, Instituto Venezolano de Investigaciones Cientificas, Caracas, Venezuela
- Unit of Advanced Therapies, Instituto Distrital de Ciencia Biotecnología e Innovación en Salud, Bogotá, Colombia
| | - Gayane Yenokyan
- Johns Hopkins Biostatistics Center, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Nicola G Cascella
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sandra Lin
- Department of Surgery, University of Wisconsin School of Medicine, Madison, WI, USA
| | - David J Schretlen
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Philip D Harvey
- Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Russell L Margolis
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Christopher A Ross
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Akira Sawa
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Mental Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Frederick C Nucifora
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
3
|
Baker CE, Marta AG, Zimmerman ND, Korade Z, Mathy NW, Wilton D, Simeone T, Kochvar A, Kramer KL, Stessman HAF, Shibata A. CPT2 Deficiency Modeled in Zebrafish: Abnormal Neural Development, Electrical Activity, Behavior, and Schizophrenia-Related Gene Expression. Biomolecules 2024; 14:914. [PMID: 39199302 PMCID: PMC11353230 DOI: 10.3390/biom14080914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/17/2024] [Accepted: 07/18/2024] [Indexed: 09/01/2024] Open
Abstract
Carnitine palmitoyltransferase 2 (CPT2) is an inner mitochondrial membrane protein of the carnitine shuttle and is involved in the beta-oxidation of long chain fatty acids. Beta-oxidation provides an alternative pathway of energy production during early development and starvation. CPT2 deficiency is a genetic disorder that we recently showed can be associated with schizophrenia. We hypothesize that CPT2 deficiency during early brain development causes transcriptional, structural, and functional abnormalities that may contribute to a CNS environment that is susceptible to the emergence of schizophrenia. To investigate the effect of CPT2 deficiency on early vertebrate development and brain function, CPT2 was knocked down in a zebrafish model system. CPT2 knockdown resulted in abnormal lipid utilization and deposition, reduction in body size, and abnormal brain development. Axonal projections, neurotransmitter synthesis, electrical hyperactivity, and swimming behavior were disrupted in CPT2 knockdown zebrafish. RT-qPCR analyses showed significant increases in the expression of schizophrenia-associated genes in CPT2 knockdown compared to control zebrafish. Taken together, these data demonstrate that zebrafish are a useful model for studying the importance of beta-oxidation for early vertebrate development and brain function. This study also presents novel findings linking CPT2 deficiency to the regulation of schizophrenia and neurodegenerative disease-associated genes.
Collapse
Affiliation(s)
- Carly E. Baker
- Department of Biomedical Sciences, Creighton University, Omaha, NE 68178, USA; (C.E.B.); (K.L.K.)
| | - Aaron G. Marta
- Department of Biology, Creighton University, Omaha, NE 68178, USA; (A.G.M.); (N.D.Z.); (N.W.M.); (D.W.); (A.K.)
| | - Nathan D. Zimmerman
- Department of Biology, Creighton University, Omaha, NE 68178, USA; (A.G.M.); (N.D.Z.); (N.W.M.); (D.W.); (A.K.)
| | - Zeljka Korade
- Department of Pediatrics, Department of Biochemistry & Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68178, USA;
| | - Nicholas W. Mathy
- Department of Biology, Creighton University, Omaha, NE 68178, USA; (A.G.M.); (N.D.Z.); (N.W.M.); (D.W.); (A.K.)
| | - Delaney Wilton
- Department of Biology, Creighton University, Omaha, NE 68178, USA; (A.G.M.); (N.D.Z.); (N.W.M.); (D.W.); (A.K.)
| | - Timothy Simeone
- Department of Pharmacology and Neuroscience, Creighton University, Omaha, NE 68178, USA; (T.S.); (H.A.F.S.)
| | - Andrew Kochvar
- Department of Biology, Creighton University, Omaha, NE 68178, USA; (A.G.M.); (N.D.Z.); (N.W.M.); (D.W.); (A.K.)
| | - Kenneth L. Kramer
- Department of Biomedical Sciences, Creighton University, Omaha, NE 68178, USA; (C.E.B.); (K.L.K.)
| | - Holly A. F. Stessman
- Department of Pharmacology and Neuroscience, Creighton University, Omaha, NE 68178, USA; (T.S.); (H.A.F.S.)
| | - Annemarie Shibata
- Department of Biology, Creighton University, Omaha, NE 68178, USA; (A.G.M.); (N.D.Z.); (N.W.M.); (D.W.); (A.K.)
| |
Collapse
|
4
|
Tao Y, Zhao R, Yang B, Han J, Li Y. Dissecting the shared genetic landscape of anxiety, depression, and schizophrenia. J Transl Med 2024; 22:373. [PMID: 38637810 PMCID: PMC11025255 DOI: 10.1186/s12967-024-05153-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 04/01/2024] [Indexed: 04/20/2024] Open
Abstract
BACKGROUND Numerous studies highlight the genetic underpinnings of mental disorders comorbidity, particularly in anxiety, depression, and schizophrenia. However, their shared genetic loci are not well understood. Our study employs Mendelian randomization (MR) and colocalization analyses, alongside multi-omics data, to uncover potential genetic targets for these conditions, thereby informing therapeutic and drug development strategies. METHODS We utilized the Consortium for Linkage Disequilibrium Score Regression (LDSC) and Mendelian Randomization (MR) analysis to investigate genetic correlations among anxiety, depression, and schizophrenia. Utilizing GTEx V8 eQTL and deCODE Genetics pQTL data, we performed a three-step summary-data-based Mendelian randomization (SMR) and protein-protein interaction analysis. This helped assess causal and comorbid loci for these disorders and determine if identified loci share coincidental variations with psychiatric diseases. Additionally, phenome-wide association studies, drug prediction, and molecular docking validated potential drug targets. RESULTS We found genetic correlations between anxiety, depression, and schizophrenia, and under a meta-analysis of MR from multiple databases, the causal relationships among these disorders are supported. Based on this, three-step SMR and colocalization analyses identified ITIH3 and CCS as being related to the risk of developing depression, while CTSS and DNPH1 are related to the onset of schizophrenia. BTN3A1, PSMB4, and TIMP4 were identified as comorbidity loci for both disorders. Molecules that could not be determined through colocalization analysis were also presented. Drug prediction and molecular docking showed that some drugs and proteins have good binding affinity and available structural data. CONCLUSIONS Our study indicates genetic correlations and shared risk loci between anxiety, depression, and schizophrenia. These findings offer insights into the underlying mechanisms of their comorbidities and aid in drug development.
Collapse
Affiliation(s)
- Yiming Tao
- Department of Intensive Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hankou, Wuhan, 430030, China
- Department of Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250101, Shandong, China
| | - Rui Zhao
- Department of Laboratory Medicine, The First Afliated Hospital of Chongqing Medical University, Chongqing, 400042, China
| | - Bin Yang
- Department of Intensive Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hankou, Wuhan, 430030, China
| | - Jie Han
- Department of Emergency, School of Medicine, Qingdao Municipal Hospital, Qingdao University, Qingdao, 266071, China.
| | - Yongsheng Li
- Department of Intensive Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hankou, Wuhan, 430030, China.
| |
Collapse
|
5
|
Pils M, Rutsch J, Eren F, Engberg G, Piehl F, Cervenka S, Sellgren C, Troßbach S, Willbold D, Erhardt S, Bannach O, Korth C. Disrupted-in-schizophrenia 1 protein aggregates in cerebrospinal fluid are elevated in patients with first-episode psychosis. Psychiatry Clin Neurosci 2023; 77:665-671. [PMID: 37668563 DOI: 10.1111/pcn.13594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 08/25/2023] [Accepted: 09/01/2023] [Indexed: 09/06/2023]
Abstract
AIM The disrupted-in-schizophrenia 1 (DISC1) protein is a key regulator at the intersection of major signaling pathways relevant for adaptive behavior. It is prone to posttranslational changes such as misassembly and aggregation but the significance of such transformations for human mental illness has remained unclear. We aimed to demonstrate the occurrence of DISC1 protein aggregates in patients with first-episode psychosis (FEP). METHOD Cerebrospinal fluid samples of patients with FEP (n = 50) and matched healthy controls (HCs; n = 47) were measured by the highly sensitive surface-based fluorescence intensity distribution analysis technology that enables single aggregate detection. RESULTS We demonstrate that DISC1 protein aggregates are increased in cerebrospinal fluid samples of patients with FEP versus HCs. The concentration was in the low femtomolar range. No correlations were found with specific symptom levels, but the difference was particularly significant in the subset of patients with the diagnoses schizophrenia, unspecified (DSM-IV 295.9) or schizoaffective disorder (DSM-IV 295.70) at 18-month follow-up. DISC1 protein aggregate levels did not significantly change within the 18-month observation interval and were on average higher for individuals carrying the major DISC1 rs821577 allele, before correction. CONCLUSION The occurrence of protein aggregates in vivo in patients with psychotic disorders has not been previously reported. It underscores the significance of posttranslational modifications of proteins both as pathogenetic mechanisms and as potential diagnostic markers in these disorders.
Collapse
Affiliation(s)
- Marlene Pils
- Institute of Biological Information Processing (Structural Biochemistry: IBI-7), Forschungszentrum Jülich, Jülich, Germany
- attyloid GmbH, Düsseldorf, Germany
| | - Julia Rutsch
- Department of Neuropathology, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Feride Eren
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Göran Engberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Fredrik Piehl
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Simon Cervenka
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, and Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
- Department of Medical Sciences, Psychiatry, Uppsala University, Uppsala, Sweden
| | - Carl Sellgren
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, and Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
| | - Svenja Troßbach
- Department of Neuropathology, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Dieter Willbold
- Institute of Biological Information Processing (Structural Biochemistry: IBI-7), Forschungszentrum Jülich, Jülich, Germany
- attyloid GmbH, Düsseldorf, Germany
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - Sophie Erhardt
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Oliver Bannach
- Institute of Biological Information Processing (Structural Biochemistry: IBI-7), Forschungszentrum Jülich, Jülich, Germany
- attyloid GmbH, Düsseldorf, Germany
| | - Carsten Korth
- Department of Neuropathology, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
6
|
Ochneva A, Zorkina Y, Abramova O, Pavlova O, Ushakova V, Morozova A, Zubkov E, Pavlov K, Gurina O, Chekhonin V. Protein Misfolding and Aggregation in the Brain: Common Pathogenetic Pathways in Neurodegenerative and Mental Disorders. Int J Mol Sci 2022; 23:14498. [PMID: 36430976 PMCID: PMC9695177 DOI: 10.3390/ijms232214498] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/07/2022] [Accepted: 11/19/2022] [Indexed: 11/23/2022] Open
Abstract
Mental disorders represent common brain diseases characterized by substantial impairments of social and cognitive functions. The neurobiological causes and mechanisms of psychopathologies still have not been definitively determined. Various forms of brain proteinopathies, which include a disruption of protein conformations and the formation of protein aggregates in brain tissues, may be a possible cause behind the development of psychiatric disorders. Proteinopathies are known to be the main cause of neurodegeneration, but much less attention is given to the role of protein impairments in psychiatric disorders' pathogenesis, such as depression and schizophrenia. For this reason, the aim of this review was to discuss the potential contribution of protein illnesses in the development of psychopathologies. The first part of the review describes the possible mechanisms of disruption to protein folding and aggregation in the cell: endoplasmic reticulum stress, dysfunction of chaperone proteins, altered mitochondrial function, and impaired autophagy processes. The second part of the review addresses the known proteins whose aggregation in brain tissue has been observed in psychiatric disorders (amyloid, tau protein, α-synuclein, DISC-1, disbindin-1, CRMP1, SNAP25, TRIOBP, NPAS3, GluA1, FABP, and ankyrin-G).
Collapse
Affiliation(s)
- Aleksandra Ochneva
- Department Basic and Applied Neurobiology, V.P. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia
- Healthcare Department, Mental-Health Clinic No. 1 Named after N.A. Alexeev of Moscow, 117152 Moscow, Russia
| | - Yana Zorkina
- Department Basic and Applied Neurobiology, V.P. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia
- Healthcare Department, Mental-Health Clinic No. 1 Named after N.A. Alexeev of Moscow, 117152 Moscow, Russia
| | - Olga Abramova
- Department Basic and Applied Neurobiology, V.P. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia
- Healthcare Department, Mental-Health Clinic No. 1 Named after N.A. Alexeev of Moscow, 117152 Moscow, Russia
| | - Olga Pavlova
- Department Basic and Applied Neurobiology, V.P. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia
| | - Valeriya Ushakova
- Department Basic and Applied Neurobiology, V.P. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia
- Healthcare Department, Mental-Health Clinic No. 1 Named after N.A. Alexeev of Moscow, 117152 Moscow, Russia
- Department of Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Anna Morozova
- Department Basic and Applied Neurobiology, V.P. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia
- Healthcare Department, Mental-Health Clinic No. 1 Named after N.A. Alexeev of Moscow, 117152 Moscow, Russia
| | - Eugene Zubkov
- Department Basic and Applied Neurobiology, V.P. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia
| | - Konstantin Pavlov
- Department Basic and Applied Neurobiology, V.P. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia
- Healthcare Department, Mental-Health Clinic No. 1 Named after N.A. Alexeev of Moscow, 117152 Moscow, Russia
| | - Olga Gurina
- Department Basic and Applied Neurobiology, V.P. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia
| | - Vladimir Chekhonin
- Department Basic and Applied Neurobiology, V.P. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia
- Department of Medical Nanobiotechnology, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
- National University of Science and Technology “MISiS”, Leninskiy Avenue 4, 119049 Moscow, Russia
| |
Collapse
|
7
|
Wang AL, Chao OY, Nikolaus S, Lamounier-Zepter V, Hollenberg CP, Lubec G, Trossbach SV, Korth C, Huston JP. Disrupted-in-schizophrenia 1 Protein Misassembly Impairs Cognitive Flexibility and Social Behaviors in a Transgenic Rat Model. Neuroscience 2022; 493:41-51. [PMID: 35461978 DOI: 10.1016/j.neuroscience.2022.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 04/12/2022] [Accepted: 04/13/2022] [Indexed: 11/19/2022]
Abstract
Alterations in cognitive functions, social behaviors and stress reactions are commonly diagnosed in chronic mental illnesses (CMI). Animal models expressing mutant genes associated to CMI represent either rare mutations or those contributing only minimally to genetic risk. Non-genetic causes of CMI can be modeled by disturbing downstream signaling pathways, for example through inducing protein misassembly or aggregation. The Disrupted-in-Schizophrenia 1 (DISC1) gene was identified to be disrupted and thereby haploinsufficient in a large pedigree where it associated to CMI. The DISC1 protein misassembles to an insoluble protein in a subset of CMI patients and this has been modeled in a rat (tgDISC1 rat) where the full-length, non mutant human transgene was overexpressed and cognitive impairments were observed. Here, we investigated the scope of effects of DISC1 protein misassembly by investigating spatial memory, social behavior and stress resilience. In water maze tasks, the tgDISC1 rats showed intact spatial learning and memory, but were deficient in flexible adaptation to spatial reversal learning compared to littermate controls. They also displayed less social interaction. Additionally, there was a trend towards increased corticosterone levels after restraint stress in the tgDISC1 rats. Our findings suggest that DISC1 protein misassembly leads to disturbances of cognitive flexibility and social behaviors, and might also be involved in stress sensitization. Since the observed behavioral features resemble symptoms of CMI, the tgDISC1 rat may be a valuable model for the investigation of cognitive, social and - possibly - also stress-related symptoms of major mental illnesses.
Collapse
Affiliation(s)
- An-Li Wang
- Center for Behavioral Neuroscience, Institute of Experimental Psychology, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany.
| | - Owen Y Chao
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN, USA.
| | - Susanne Nikolaus
- Department of Nuclear Medicine, University Hospital Düsseldorf, Heinrich-Heine University, Düsseldorf, Germany.
| | | | - Cornelis P Hollenberg
- Institute of Microbiology, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany.
| | - Gert Lubec
- Department of Neuroproteomics, Paracelsus Private Medical University, Salzburg, Austria.
| | - Svenja V Trossbach
- Department of Neuropathology, University Hospital Düsseldorf, Düsseldorf, Germany.
| | - Carsten Korth
- Department of Neuropathology, University Hospital Düsseldorf, Düsseldorf, Germany.
| | - Joseph P Huston
- Center for Behavioral Neuroscience, Institute of Experimental Psychology, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
8
|
Cukkemane A, Becker N, Zielinski M, Frieg B, Lakomek NA, Heise H, Schröder GF, Willbold D, Weiergräber OH. Conformational heterogeneity coupled with β-fibril formation of a scaffold protein involved in chronic mental illnesses. Transl Psychiatry 2021; 11:639. [PMID: 34921141 PMCID: PMC8683410 DOI: 10.1038/s41398-021-01765-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 11/23/2021] [Accepted: 12/07/2021] [Indexed: 12/17/2022] Open
Abstract
Chronic mental illnesses (CMIs) pose a significant challenge to global health due to their complex and poorly understood etiologies and hence, absence of causal therapies. Research of the past two decades has revealed dysfunction of the disrupted in schizophrenia 1 (DISC1) protein as a predisposing factor involved in several psychiatric disorders. DISC1 is a multifaceted protein that serves myriads of functions in mammalian cells, for instance, influencing neuronal development and synapse maintenance. It serves as a scaffold hub forming complexes with a variety (~300) of partners that constitute its interactome. Herein, using combinations of structural and biophysical tools, we demonstrate that the C-region of the DISC1 protein is highly polymorphic, with important consequences for its physiological role. Results from solid-state NMR spectroscopy and electron microscopy indicate that the protein not only forms symmetric oligomers but also gives rise to fibrils closely resembling those found in certain established amyloid proteinopathies. Furthermore, its aggregation as studied by isothermal titration calorimetry (ITC) is an exergonic process, involving a negative enthalpy change that drives the formation of oligomeric (presumably tetrameric) species as well as β-fibrils. We have been able to narrow down the β-core region participating in fibrillization to residues 716-761 of full-length human DISC1. This region is absent in the DISC1Δ22aa splice variant, resulting in reduced association with proteins from the dynein motor complex, viz., NDE-like 1 (NDEL1) and lissencephaly 1 (LIS1), which are crucial during mitosis. By employing surface plasmon resonance, we show that the oligomeric DISC1 C-region has an increased affinity and shows cooperativity in binding to LIS1 and NDEL1, in contrast to the noncooperative binding mode exhibited by the monomeric version. Based on the derived structural models, we propose that the association between the binding partners involves two neighboring subunits of DISC1 C-region oligomers. Altogether, our findings highlight the significance of the DISC1 C-region as a crucial factor governing the balance between its physiological role as a multifunctional scaffold protein and aggregation-related aberrations with potential significance for disease.
Collapse
Affiliation(s)
- Abhishek Cukkemane
- Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, Jülich, Germany. .,Institut für Physikalische Biologie, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| | - Nina Becker
- grid.8385.60000 0001 2297 375XInstitute of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, Jülich, Germany ,grid.411327.20000 0001 2176 9917Institut für Physikalische Biologie, Heinrich Heine University Düsseldorf, Düsseldorf, Germany ,grid.8385.60000 0001 2297 375XJülich Centre for Structural Biology (JuStruct), Forschungszentrum Jülich, Jülich, Germany
| | - Mara Zielinski
- grid.8385.60000 0001 2297 375XInstitute of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, Jülich, Germany
| | - Benedikt Frieg
- grid.8385.60000 0001 2297 375XInstitute of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, Jülich, Germany
| | - Nils-Alexander Lakomek
- grid.8385.60000 0001 2297 375XInstitute of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, Jülich, Germany ,grid.411327.20000 0001 2176 9917Institut für Physikalische Biologie, Heinrich Heine University Düsseldorf, Düsseldorf, Germany ,grid.8385.60000 0001 2297 375XJülich Centre for Structural Biology (JuStruct), Forschungszentrum Jülich, Jülich, Germany
| | - Henrike Heise
- grid.8385.60000 0001 2297 375XInstitute of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, Jülich, Germany ,grid.411327.20000 0001 2176 9917Institut für Physikalische Biologie, Heinrich Heine University Düsseldorf, Düsseldorf, Germany ,grid.8385.60000 0001 2297 375XJülich Centre for Structural Biology (JuStruct), Forschungszentrum Jülich, Jülich, Germany
| | - Gunnar F. Schröder
- grid.8385.60000 0001 2297 375XInstitute of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, Jülich, Germany ,grid.8385.60000 0001 2297 375XJülich Centre for Structural Biology (JuStruct), Forschungszentrum Jülich, Jülich, Germany ,grid.411327.20000 0001 2176 9917Physics Department, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Dieter Willbold
- Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, Jülich, Germany. .,Institut für Physikalische Biologie, Heinrich Heine University Düsseldorf, Düsseldorf, Germany. .,Jülich Centre for Structural Biology (JuStruct), Forschungszentrum Jülich, Jülich, Germany.
| | - Oliver H. Weiergräber
- grid.8385.60000 0001 2297 375XInstitute of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, Jülich, Germany ,grid.8385.60000 0001 2297 375XJülich Centre for Structural Biology (JuStruct), Forschungszentrum Jülich, Jülich, Germany
| |
Collapse
|
9
|
Antipsychotic clozapine binding to alpha-2-macroglobulin protects interacting partners against oxidation and preserves the anti-proteinase activity of the protein. Int J Biol Macromol 2021; 183:502-512. [PMID: 33930446 DOI: 10.1016/j.ijbiomac.2021.04.155] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/23/2021] [Accepted: 04/24/2021] [Indexed: 12/20/2022]
Abstract
In this study, the interaction between clozapine, an atypical antipsychotic drug, and alpha-2-macroglobulin (α2M), a multipurpose anti-proteinase, was investigated under simulated (patho) physiological conditions using multiple spectroscopic techniques and molecular modeling. It was found that α2M binds clozapine with a moderate affinity (the binding constant of 0.9 × 105 M-1 at 37 °C). The preferable binding site for both clozapine's atropisomers was revealed to be a large pocket at the interface of C and D monomer subunits of the protein. Hydrogen bonds and the hydrophobic effect were proposed as dominant forces in complex formation. The binding of clozapine did not induce significant conformational change of the protein, as confirmed by virtually unaltered α2M secondary structure and anti-proteinase activity. However, both clozapine and α2M shielded each other from the deleterious influence of strong oxidants: sodium hypochlorite and 2,2'-azobis-2-methyl-propanimidamide dihydrochloride (AAPH). Moreover, clozapine in a concentration range that is usually targeted in the plasma during patients' treatment effectively protected the anti-proteinase activity of α2M under AAPH-induced free radical overproduction. Our results suggest that the cooperation between α2M and clozapine may be a path by which these two molecules synergistically protect neural tissue against injury caused by disturbed proteostasis or oxidative stress.
Collapse
|
10
|
Cauda F, Mancuso L, Nani A, Ficco L, Premi E, Manuello J, Liloia D, Gelmini G, Duca S, Costa T. Hubs of long-distance co-alteration characterize brain pathology. Hum Brain Mapp 2020; 41:3878-3899. [PMID: 32562581 PMCID: PMC7469792 DOI: 10.1002/hbm.25093] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 05/06/2020] [Accepted: 05/26/2020] [Indexed: 12/14/2022] Open
Abstract
It is becoming clearer that the impact of brain diseases is more convincingly represented in terms of co-alterations rather than in terms of localization of alterations. In this context, areas characterized by a long mean distance of co-alteration may be considered as hubs with a crucial role in the pathology. We calculated meta-analytic transdiagnostic networks of co-alteration for the gray matter decreases and increases, and we evaluated the mean Euclidean, fiber-length, and topological distance of its nodes. We also examined the proportion of co-alterations between canonical networks, and the transdiagnostic variance of the Euclidean distance. Furthermore, disease-specific analyses were conducted on schizophrenia and Alzheimer's disease. The anterodorsal prefrontal cortices appeared to be a transdiagnostic hub of long-distance co-alterations. Also, the disease-specific analyses showed that long-distance co-alterations are more able than classic meta-analyses to identify areas involved in pathology and symptomatology. Moreover, the distance maps were correlated with the normative connectivity. Our findings substantiate the network degeneration hypothesis in brain pathology. At the same time, they suggest that the concept of co-alteration might be a useful tool for clinical neuroscience.
Collapse
Affiliation(s)
- Franco Cauda
- GCS‐fMRI, Koelliker Hospital and Department of PsychologyUniversity of TurinTurinItaly
- FOCUS Lab, Department of PsychologyUniversity of TurinTurinItaly
| | - Lorenzo Mancuso
- GCS‐fMRI, Koelliker Hospital and Department of PsychologyUniversity of TurinTurinItaly
- FOCUS Lab, Department of PsychologyUniversity of TurinTurinItaly
| | - Andrea Nani
- GCS‐fMRI, Koelliker Hospital and Department of PsychologyUniversity of TurinTurinItaly
- FOCUS Lab, Department of PsychologyUniversity of TurinTurinItaly
| | - Linda Ficco
- GCS‐fMRI, Koelliker Hospital and Department of PsychologyUniversity of TurinTurinItaly
- FOCUS Lab, Department of PsychologyUniversity of TurinTurinItaly
| | - Enrico Premi
- Stroke Unit, Azienda Socio‐Sanitaria Territoriale Spedali CiviliSpedali Civili HospitalBresciaItaly
- Centre for Neurodegenerative Disorders, Neurology Unit, Department of Clinical and Experimental SciencesUniversity of BresciaBresciaItaly
| | - Jordi Manuello
- GCS‐fMRI, Koelliker Hospital and Department of PsychologyUniversity of TurinTurinItaly
- FOCUS Lab, Department of PsychologyUniversity of TurinTurinItaly
| | - Donato Liloia
- GCS‐fMRI, Koelliker Hospital and Department of PsychologyUniversity of TurinTurinItaly
- FOCUS Lab, Department of PsychologyUniversity of TurinTurinItaly
| | - Gabriele Gelmini
- FOCUS Lab, Department of PsychologyUniversity of TurinTurinItaly
| | - Sergio Duca
- GCS‐fMRI, Koelliker Hospital and Department of PsychologyUniversity of TurinTurinItaly
| | - Tommaso Costa
- GCS‐fMRI, Koelliker Hospital and Department of PsychologyUniversity of TurinTurinItaly
- FOCUS Lab, Department of PsychologyUniversity of TurinTurinItaly
| |
Collapse
|
11
|
Zafarullah M, Tang HT, Durbin-Johnson B, Fourie E, Hessl D, Rivera SM, Tassone F. FMR1 locus isoforms: potential biomarker candidates in fragile X-associated tremor/ataxia syndrome (FXTAS). Sci Rep 2020; 10:11099. [PMID: 32632326 PMCID: PMC7338407 DOI: 10.1038/s41598-020-67946-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Accepted: 05/18/2020] [Indexed: 12/12/2022] Open
Abstract
Fragile X associated tremor/ataxia syndrome (FXTAS) is a late adult-onset neurodegenerative disorder that affects movement and cognition in male and female carriers of a premutation allele of 55-200 CGG repeats in the Fragile X mental retardation (FMR1) gene. It is currently unknown if and when an individual carrier of a premutation allele will develop FXTAS, as clinical assessment fails to identify carriers at risk before significant neurological symptoms are evident. The primary objective of this study was to investigate the alternative splicing landscape at the FMR1 locus in conjunction with brain measures in male individuals with a premutation allele enrolled in a very first longitudinal study, compared to age-matched healthy male controls, with the purpose of identifying biomarkers for early diagnosis, disease prediction and, a progression of FXTAS. Our findings indicate that increased expression of FMR1 mRNA isoforms, including Iso4/4b, Iso10/10b, as well as of the ASFMR1 mRNAs Iso131bp, are present in premutation carriers as compared to non-carrier healthy controls. More specifically, we observed a higher expression of Iso4/4b and Iso10/10b, which encode for truncated proteins, only in those premutation carriers who developed symptoms of FXTAS over time as compared to non-carrier healthy controls, suggesting a potential role in the development of the disorder. In addition, we found a significant association of these molecular changes with various measurements of brain morphology, including the middle cerebellar peduncle (MCP), superior cerebellar peduncle (SCP), pons, and midbrain, indicating their potential contribution to the pathogenesis of FXTAS. Interestingly, the high expression levels of Iso4/4b observed both at visit 1 and visit 2 and found to be associated with a decrease in mean MCP width only in those individuals who developed FXTAS over time, suggests their role as potential biomarkers for early diagnosis of FXTAS.
Collapse
Affiliation(s)
- Marwa Zafarullah
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, 95817 CA, USA
| | - Hiu-Tung Tang
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, 95817 CA, USA
| | - Blythe Durbin-Johnson
- Division of Biostatistics, School of Medicine, University of California Davis, Davis, CA, USA
| | - Emily Fourie
- Center for Mind and Brain, University of California Davis, Davis, CA, USA
- Department of Psychology, University of California, Davis, Davis, CA, USA
| | - David Hessl
- MIND Institute, University of California Davis Medical Center, Sacramento, 95817 CA, USA
- Department of Psychiatry and Behavioral Sciences, University of California Davis Medical Center, Sacramento, 95817 CA, USA
| | - Susan M Rivera
- Center for Mind and Brain, University of California Davis, Davis, CA, USA
- Department of Psychology, University of California, Davis, Davis, CA, USA
- MIND Institute, University of California Davis Medical Center, Sacramento, 95817 CA, USA
| | - Flora Tassone
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, 95817 CA, USA.
- MIND Institute, University of California Davis Medical Center, Sacramento, 95817 CA, USA.
| |
Collapse
|
12
|
Tomoda T, Yang K, Sawa A. Neuronal Autophagy in Synaptic Functions and Psychiatric Disorders. Biol Psychiatry 2020; 87:787-796. [PMID: 31542152 PMCID: PMC6986983 DOI: 10.1016/j.biopsych.2019.07.018] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 07/02/2019] [Accepted: 07/19/2019] [Indexed: 12/22/2022]
Abstract
Homeostatic maintenance of physiological functions is fundamental to organismal well-being. Disruption or imbalance in homeostasis results in functional disturbances at molecular, cellular, and tissue levels, leading to manifestation as physical and mental illnesses. Homeostatic imbalance is caused by a range of pathophysiological mechanisms, including disrupted reduction-oxidation reactions, inflammatory responses, metabolic disturbances, or failure in quality control of cellular proteins and organelles. However, the roles for the protein/organelle quality control in the regulation of behaviors, in particular of cognitive processes, had not been well documented, until recent reports finally supported this concept. The frontline studies in neuroscience have revealed that synaptic components (e.g., synaptic proteins, organelles, neurotransmitters and their receptors) are selectively degraded by autophagy, a cellular recycling machinery implicated in surveillance and quality control of proteins and organelles responsible for the maintenance of cellular homeostasis. Apart from the canonical role of autophagy in supporting cell viability, synaptic autophagy appears to regulate synapse remodeling and plasticity. Consistently, emerging evidence suggests novel roles of autophagy in memory encoding, information processing, or cognitive functions. In this review, we overview recent progress in understanding the roles of neuronal autophagy in homeostatic maintenance of synaptic functions, with particular focus on how disruptions in these processes may contribute to the pathophysiology of psychiatric disorders.
Collapse
Affiliation(s)
- Toshifumi Tomoda
- Centre for Addiction and Mental Health, University of Toronto, Toronto, Ontario, Canada.
| | - Kun Yang
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA
| | - Akira Sawa
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland.
| |
Collapse
|
13
|
Effects of Cannabis Use on the Protein and Lipid Profile of Olfactory Neuroepithelium Cells from Schizophrenia Patients Studied by Synchrotron-Based FTIR Spectroscopy. Biomolecules 2020; 10:biom10020329. [PMID: 32092878 PMCID: PMC7072126 DOI: 10.3390/biom10020329] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 02/12/2020] [Accepted: 02/15/2020] [Indexed: 12/14/2022] Open
Abstract
Schizophrenia (SCZ) is a neurodevelopmental disorder with a high genetic component, but the presence of environmental stressors can be important for its onset and progression. Cannabis use can be a major risk factor for developing SCZ. However, despite the available data on the neurobiological underpinnings of SCZ, there is an important lack of studies in human neuronal tissue and living cells addressing the effects of cannabis in SCZ patients. In this study, we analysed the most relevant bio-macromolecular constituents in olfactory neuroepithelium (ON) cells of healthy controls non-cannabis users, healthy cannabis users, SCZ patients non-cannabis users, and SCZ patients cannabis users using Synchrotron Radiation-Fourier Transform Infrared (SR-FTIR) spectrometry and microscopy. Our results revealed that SCZ patients non-cannabis users, and healthy cannabis users exhibit similar alterations in the macromolecular profile of ON cells, including disruption in lipid composition, increased lipid membrane renewal rate and lipid peroxidation, altered proteins containing more β-sheet structures, and showed an increase in DNA and histone methylation. Notably, these alterations were not observed in SCZ patients who use cannabis regularly. These data suggest a differential effect of cannabis in healthy controls and in SCZ patients in terms of the macromolecular constituents of ON cells.
Collapse
|
14
|
Toyoshima M, Jiang X, Ogawa T, Ohnishi T, Yoshihara S, Balan S, Yoshikawa T, Hirokawa N. Enhanced carbonyl stress induces irreversible multimerization of CRMP2 in schizophrenia pathogenesis. Life Sci Alliance 2019; 2:2/5/e201900478. [PMID: 31591136 PMCID: PMC6781483 DOI: 10.26508/lsa.201900478] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 09/17/2019] [Accepted: 09/19/2019] [Indexed: 12/31/2022] Open
Abstract
Enhanced carbonyl stress results in neurodevelopmental deficits by affecting microtubule function through the formation of irreversible dysfunctional multimer of carbonylated CRMP2. Enhanced carbonyl stress underlies a subset of schizophrenia, but its causal effects remain elusive. Here, we elucidated the molecular mechanism underlying the effects of carbonyl stress in iPS cells in which the gene encoding zinc metalloenzyme glyoxalase I (GLO1), a crucial enzyme for the clearance of carbonyl stress, was disrupted. The iPS cells exhibited significant cellular and developmental deficits, and hyper-carbonylation of collapsing response mediator protein 2 (CRMP2). Structural and biochemical analyses revealed an array of multiple carbonylation sites in the functional motifs of CRMP2, particularly D-hook (for dimerization) and T-site (for tetramerization), which are critical for the activity of the CRMP2 tetramer. Interestingly, carbonylated CRMP2 was stacked in the multimer conformation by irreversible cross-linking, resulting in loss of its unique function to bundle microtubules. Thus, the present study revealed that the enhanced carbonyl stress stemmed from the genetic aberrations results in neurodevelopmental deficits through the formation of irreversible dysfunctional multimer of carbonylated CRMP2.
Collapse
Affiliation(s)
- Manabu Toyoshima
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Wako, Japan
| | - Xuguang Jiang
- Department of Cell Biology and Anatomy, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Tadayuki Ogawa
- Department of Cell Biology and Anatomy, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Tetsuo Ohnishi
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Wako, Japan
| | - Shogo Yoshihara
- Department of Cell Biology and Anatomy, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Shabeesh Balan
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Wako, Japan
| | - Takeo Yoshikawa
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Wako, Japan
| | - Nobutaka Hirokawa
- Department of Cell Biology and Anatomy, Graduate School of Medicine, University of Tokyo, Tokyo, Japan .,Center of Excellence in Genome Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
15
|
Cauda F, Nani A, Manuello J, Premi E, Palermo S, Tatu K, Duca S, Fox PT, Costa T. Brain structural alterations are distributed following functional, anatomic and genetic connectivity. Brain 2019; 141:3211-3232. [PMID: 30346490 DOI: 10.1093/brain/awy252] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 08/22/2018] [Indexed: 12/18/2022] Open
Abstract
The pathological brain is characterized by distributed morphological or structural alterations in the grey matter, which tend to follow identifiable network-like patterns. We analysed the patterns formed by these alterations (increased and decreased grey matter values detected with the voxel-based morphometry technique) conducting an extensive transdiagnostic search of voxel-based morphometry studies in a large variety of brain disorders. We devised an innovative method to construct the networks formed by the structurally co-altered brain areas, which can be considered as pathological structural co-alteration patterns, and to compare these patterns with three associated types of connectivity profiles (functional, anatomical, and genetic). Our study provides transdiagnostical evidence that structural co-alterations are influenced by connectivity constraints rather than being randomly distributed. Analyses show that although all the three types of connectivity taken together can account for and predict with good statistical accuracy, the shape and temporal development of the co-alteration patterns, functional connectivity offers the better account of the structural co-alteration, followed by anatomic and genetic connectivity. These results shed new light on the possible mechanisms at the root of neuropathological processes and open exciting prospects in the quest for a better understanding of brain disorders.
Collapse
Affiliation(s)
- Franco Cauda
- GCS-fMRI, Koelliker Hospital and Department of Psychology, University of Turin, Turin, Italy.,FOCUS Lab, Department of Psychology, University of Turin, Turin, Italy
| | - Andrea Nani
- GCS-fMRI, Koelliker Hospital and Department of Psychology, University of Turin, Turin, Italy.,FOCUS Lab, Department of Psychology, University of Turin, Turin, Italy
| | - Jordi Manuello
- GCS-fMRI, Koelliker Hospital and Department of Psychology, University of Turin, Turin, Italy.,FOCUS Lab, Department of Psychology, University of Turin, Turin, Italy
| | - Enrico Premi
- Stroke Unit, Azienda Socio Sanitaria Territoriale Spedali Civili, Spedali Civili Hospital, Brescia, Italy.,Centre for Neurodegenerative Disorders, Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Sara Palermo
- Department of Neuroscience, University of Turin, Turin, Italy
| | - Karina Tatu
- GCS-fMRI, Koelliker Hospital and Department of Psychology, University of Turin, Turin, Italy.,FOCUS Lab, Department of Psychology, University of Turin, Turin, Italy
| | - Sergio Duca
- GCS-fMRI, Koelliker Hospital and Department of Psychology, University of Turin, Turin, Italy
| | - Peter T Fox
- Research Imaging Institute, University of Texas Health Science Center at San Antonio, Texas, USA.,South Texas Veterans Health Care System, San Antonio, Texas, USA
| | - Tommaso Costa
- GCS-fMRI, Koelliker Hospital and Department of Psychology, University of Turin, Turin, Italy.,FOCUS Lab, Department of Psychology, University of Turin, Turin, Italy
| |
Collapse
|
16
|
Kakuda K, Niwa A, Honda R, Yamaguchi KI, Tomita H, Nojebuzzaman M, Hara A, Goto Y, Osawa M, Kuwata K. A DISC1 point mutation promotes oligomerization and impairs information processing in a mouse model of schizophrenia. J Biochem 2019; 165:369-378. [PMID: 30561706 DOI: 10.1093/jb/mvy116] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 12/14/2018] [Indexed: 12/15/2022] Open
Abstract
Disrupted-in-schizophrenia 1 (DISC1) is strongly associated with schizophrenia, but it remains elusive how the modification of the intermolecular interaction of DISC1 affects the information processing in brain. We show that a DISC1 point mutation alters intermolecular cohesiveness promoting the phase separation, and disrupts sensorimotor gating monitored by the prepulse inhibition in a mouse model of schizophrenia. Although the conformation of DISC1 partial peptide with the schizophrenia-related mutation L607F in human or the corresponding L604F in mouse was essentially indistinguishable from the wild type (WT) as long as monitored by fluorescence, circular dichroism, ultracentrifugation, dynamic light scattering and nuclear magnetic resonance, the atomic force microscopy was able to detect their morphological distinctions. The WT peptides were round and well dispersed, while mutants were inhomogeneous and disrupted to form dimer to trimer that aligned along one direction without apparent aggregate formation. Homozygous L604F mutant mice created by CRISPR exhibited the significant decrease in DISC1 level in the immunohistopathology at the hippocampal region compared to the WTs. The ratio of prepulse inhibition of the homozygous mutant mice was significantly impaired compared to WTs. Altered DISC1 distribution or function caused by aberrant intermolecular interactions may contribute to information processing characteristics in schizophrenia.
Collapse
Affiliation(s)
- Kyosuke Kakuda
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, 1-1 Yanagido, Gifu
| | - Ayumi Niwa
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu
| | - Ryo Honda
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, 1-1 Yanagido, Gifu
| | - Kei-Ichi Yamaguchi
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka
| | - Hiroyuki Tomita
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu
| | - Md Nojebuzzaman
- Division of Regeneration Technology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Akira Hara
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu
| | - Yuji Goto
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka
| | - Masatake Osawa
- Division of Regeneration Technology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Kazuo Kuwata
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, 1-1 Yanagido, Gifu
| |
Collapse
|
17
|
Wang Q, Wang C, Ji B, Zhou J, Yang C, Chen J. Hapln2 in Neurological Diseases and Its Potential as Therapeutic Target. Front Aging Neurosci 2019; 11:60. [PMID: 30949044 PMCID: PMC6437066 DOI: 10.3389/fnagi.2019.00060] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Accepted: 03/01/2019] [Indexed: 01/18/2023] Open
Abstract
Hyaluronan and proteoglycan link protein 2 (Hapln2) is important for the binding of chondroitin sulfate proteoglycans to hyaluronan. Hapln2 deficiency leads to the abnormal expression of extracellular matrix (ECM) proteins and dysfunctional neuronal conductivity, demonstrating the vital role of Hapln2 in these processes. Studies have revealed that Hapln2 promotes the aggregation of α-synuclein, thereby contributing to neurodegeneration in Parkinson’s disease (PD), and it was recently suggested to be in intracellular neurofibrillary tangles (NFTs). Additionally, the expression levels of Hapln2 showed lower in the anterior temporal lobes of individuals with schizophrenia than those of healthy subjects. Together, these studies implicate the involvement of Hapln2 in the pathological processes of neurological diseases. A better understanding of the function of Hapln2 in the central nervous system (CNS) will provide new insights into the molecular mechanisms of these diseases and help to establish promising therapeutic strategies. Herein, we review the recent progress in defining the role of Hapln2 in brain physiology and pathology.
Collapse
Affiliation(s)
- Qinqin Wang
- Neurobiology Key Laboratory, Jining Medical University, Jining, China
| | - Chunmei Wang
- Neurobiology Key Laboratory, Jining Medical University, Jining, China
| | - Bingyuan Ji
- Neurobiology Key Laboratory, Jining Medical University, Jining, China
| | - Jiawei Zhou
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Chunqing Yang
- Neurobiology Key Laboratory, Jining Medical University, Jining, China
| | - Jing Chen
- Neurobiology Key Laboratory, Jining Medical University, Jining, China.,Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, United Kingdom
| |
Collapse
|
18
|
Chung CG, Lee H, Lee SB. Mechanisms of protein toxicity in neurodegenerative diseases. Cell Mol Life Sci 2018; 75:3159-3180. [PMID: 29947927 PMCID: PMC6063327 DOI: 10.1007/s00018-018-2854-4] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 06/06/2018] [Accepted: 06/07/2018] [Indexed: 12/12/2022]
Abstract
Protein toxicity can be defined as all the pathological changes that ensue from accumulation, mis-localization, and/or multimerization of disease-specific proteins. Most neurodegenerative diseases manifest protein toxicity as one of their key pathogenic mechanisms, the details of which remain unclear. By systematically deconstructing the nature of toxic proteins, we aim to elucidate and illuminate some of the key mechanisms of protein toxicity from which therapeutic insights may be drawn. In this review, we focus specifically on protein toxicity from the point of view of various cellular compartments such as the nucleus and the mitochondria. We also discuss the cell-to-cell propagation of toxic disease proteins that complicates the mechanistic understanding of the disease progression as well as the spatiotemporal point at which to therapeutically intervene. Finally, we discuss selective neuronal vulnerability, which still remains largely enigmatic.
Collapse
Affiliation(s)
- Chang Geon Chung
- Department of Brain and Cognitive Sciences, DGIST, Daegu, 42988, Republic of Korea
| | - Hyosang Lee
- Department of Brain and Cognitive Sciences, DGIST, Daegu, 42988, Republic of Korea.
| | - Sung Bae Lee
- Department of Brain and Cognitive Sciences, DGIST, Daegu, 42988, Republic of Korea.
| |
Collapse
|
19
|
Zhu S, Abounit S, Korth C, Zurzolo C. Transfer of disrupted-in-schizophrenia 1 aggregates between neuronal-like cells occurs in tunnelling nanotubes and is promoted by dopamine. Open Biol 2018; 7:rsob.160328. [PMID: 28275106 PMCID: PMC5376705 DOI: 10.1098/rsob.160328] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 02/10/2017] [Indexed: 12/22/2022] Open
Abstract
The disrupted-in-schizophrenia 1 (DISC1) gene was identified as a genetic risk factor for chronic mental illnesses (CMI) such as schizophrenia, bipolar disorder and severe recurrent depression. Insoluble aggregated DISC1 variants were found in the cingular cortex of sporadic, i.e. non-genetic, CMI patients. This suggests protein pathology as a novel, additional pathogenic mechanism, further corroborated in a recent transgenic rat model presenting DISC1 aggregates. Since the potential role of aggregation of DISC1 in sporadic CMI is unknown, we investigated whether DISC1 undergoes aggregation in cell culture and could spread between neuronal cells in a prion-like manner, as shown for amyloid proteins in neurodegenerative diseases. Co-culture experiments between donor cells forming DISC1 aggregates and acceptor cells showed that 4.5% of acceptor cells contained donor-derived DISC1 aggregates, thus indicating an efficient transfer in vitro. DISC1 aggregates were found inside tunnelling nanotubes (TNTs) and transfer was enhanced by increasing TNT formation and notably by dopamine treatment, which also induces DISC1 aggregation. These data indicate that DISC1 aggregates can propagate between cells similarly to prions, thus providing some molecular basis for the role of protein pathology in CMI.
Collapse
Affiliation(s)
- Seng Zhu
- Institut Pasteur, Membrane Traffic and Pathogenesis Unit, 25-28 rue du Docteur Roux, 75724 Paris, France
| | - Saïda Abounit
- Institut Pasteur, Membrane Traffic and Pathogenesis Unit, 25-28 rue du Docteur Roux, 75724 Paris, France
| | - Carsten Korth
- Department of Neuropathology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Chiara Zurzolo
- Institut Pasteur, Membrane Traffic and Pathogenesis Unit, 25-28 rue du Docteur Roux, 75724 Paris, France
| |
Collapse
|
20
|
Intra-nasal dopamine alleviates cognitive deficits in tgDISC1 rats which overexpress the human DISC1 gene. Neurobiol Learn Mem 2017; 146:12-20. [PMID: 29107702 DOI: 10.1016/j.nlm.2017.10.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 10/26/2017] [Accepted: 10/27/2017] [Indexed: 01/15/2023]
Abstract
The Disrupted-in-Schizophrenia 1 (DISC1) gene has been associated with mental illnesses such as major depression and schizophrenia. The transgenic DISC1 (tgDISC1) rat, which overexpresses the human DISC1 gene, is known to exhibit deficient dopamine (DA) homeostasis. To ascertain whether the DISC1 gene also impacts cognitive functions, 14-15 months old male tgDISC1 rats and wild-type controls were subjected to the novel object preference (NOP) test and the object-based attention test (OBAT) in order to assess short-term memory (1 h), long-term memory (24 h), and attention. RESULTS The tgDISC1 group exhibited intact short-term memory, but deficient long-term-memory in the NOP test and deficient attention-related behavior in the OBAT. In a different group of tgDISC1 rats, 3 mg/kg intranasally applied dopamine (IN-DA) or its vehicle was applied prior to the NOP or the OBAT test. IN-DA reversed cognitive deficits in both the NOP and OBAT tests. In a further cohort of tgDISC1 rats, post-mortem levels of DA, noradrenaline, serotonin and acetylcholine were determined in a variety of brain regions. The tgDISC1 group had less DA in the neostriatum, hippocampus and amygdala, less acetylcholine in neostriatum, nucleus accumbens, hippocampus, and amygdala, more serotonin in the nucleus accumbens, and less serotonin and noradrenaline in the amygdala. CONCLUSIONS Our findings show that DISC1 overexpression and misassembly is associated with deficits in long-term memory and attention-related behavior. Since behavioral impairments in tgDISC1 rats were reversed by IN-DA, DA deficiency may be a major cause for the behavioral deficits expressed in this model.
Collapse
|
21
|
Dittrich L, Petese A, Jackson WS. The natural Disc1-deletion present in several inbred mouse strains does not affect sleep. Sci Rep 2017; 7:5665. [PMID: 28720848 PMCID: PMC5515846 DOI: 10.1038/s41598-017-06015-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 06/06/2017] [Indexed: 02/03/2023] Open
Abstract
The gene Disrupted in Schizophrenia-1 (DISC1) is linked to a range of psychiatric disorders. Two recent transgenic studies suggest DISC1 is also involved in homeostatic sleep regulation. Several strains of inbred mice commonly used for genome manipulation experiments, including several Swiss and likely all 129 substrains, carry a natural deletion mutation of Disc1. This constitutes a potential confound for studying sleep in genetically modified mice. Since disturbed sleep can also influence psychiatric and neurodegenerative disease models, this putative confound might affect a wide range of studies in several fields. Therefore, we asked to what extent the natural Disc1 deletion affects sleep. To this end, we first compared sleep and electroencephalogram (EEG) phenotypes of 129S4 mice carrying the Disc1 deletion and C57BL/6N mice carrying the full-length version. We then bred Disc1 from C57BL/6N into the 129S4 background, resulting in S4-Disc1 mice. The differences between 129S4 and C57BL/6N were not detected in the 129S4 to S4-Disc1 comparison. We conclude that the mutation has no effect on the measured sleep and EEG characteristics. Thus, it is unlikely the widespread Disc1 deletion has led to spurious results in previous sleep studies or that it alters sleep in mouse models of psychiatric or neurodegenerative diseases.
Collapse
Affiliation(s)
- Lars Dittrich
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Sigmund-Freud-Str, 27 53127, Bonn, Germany
| | - Alessandro Petese
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Sigmund-Freud-Str, 27 53127, Bonn, Germany
| | - Walker S Jackson
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Sigmund-Freud-Str, 27 53127, Bonn, Germany.
| |
Collapse
|
22
|
Bradshaw NJ, Yerabham ASK, Marreiros R, Zhang T, Nagel-Steger L, Korth C. An unpredicted aggregation-critical region of the actin-polymerizing protein TRIOBP-1/Tara, determined by elucidation of its domain structure. J Biol Chem 2017; 292:9583-9598. [PMID: 28438837 DOI: 10.1074/jbc.m116.767939] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 04/20/2017] [Indexed: 12/22/2022] Open
Abstract
Aggregation of specific proteins in the brains of patients with chronic mental illness as a result of disruptions in proteostasis is an emerging theme in the study of schizophrenia in particular. Proteins including DISC1 (disrupted in schizophrenia 1) and dysbindin-1B are found in insoluble forms within brain homogenates from such patients. We recently identified TRIOBP-1 (Trio-binding protein 1, also known as Tara) to be another such protein through an epitope discovery and proteomics approach by comparing post-mortem brain material from schizophrenia patients and control individuals. We hypothesized that this was likely to occur as a result of a specific subcellular process and that it, therefore, should be possible to identify a region of the TRIOBP-1 protein that is essential for its aggregation to occur. Here, we probe the domain organization of TRIOBP-1, finding it to possess two distinct coiled-coil domains: the central and C-terminal domains. The central domain inhibits the depolymerization of F-actin and is also responsible for oligomerization of TRIOBP-1. Along with an N-terminal pleckstrin homology domain, the central domain affects neurite outgrowth. In neuroblastoma cells it was found that the aggregation propensity of TRIOBP-1 arises from its central domain, with a short "linker" region narrowed to within amino acids 324-348, between its first two coiled coils, as essential for the formation of TRIOBP-1 aggregates. TRIOBP-1 aggregation, therefore, appears to occur through one or more specific cellular mechanisms, which therefore have the potential to be of physiological relevance for the biological process underlying the development of chronic mental illness.
Collapse
Affiliation(s)
| | | | | | - Tao Zhang
- the Institute of Physical Biology, Heinrich Heine University, 40225 Düsseldorf, Germany and.,the Institute of Complex Systems, Structural Biochemistry (ICS-6), Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Luitgard Nagel-Steger
- the Institute of Physical Biology, Heinrich Heine University, 40225 Düsseldorf, Germany and.,the Institute of Complex Systems, Structural Biochemistry (ICS-6), Forschungszentrum Jülich, 52425 Jülich, Germany
| | | |
Collapse
|
23
|
Defective synaptic connectivity and axonal neuropathology in a human iPSC-based model of familial Parkinson's disease. Proc Natl Acad Sci U S A 2017; 114:E3679-E3688. [PMID: 28416701 DOI: 10.1073/pnas.1617259114] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
α-Synuclein (αSyn) is the major gene linked to sporadic Parkinson's disease (PD), whereas the G209A (p.A53T) αSyn mutation causes a familial form of PD characterized by early onset and a generally severe phenotype, including nonmotor manifestations. Here we generated de novo induced pluripotent stem cells (iPSCs) from patients harboring the p.A53T mutation and developed a robust model that captures PD pathogenic processes under basal conditions. iPSC-derived mutant neurons displayed novel disease-relevant phenotypes, including protein aggregation, compromised neuritic outgrowth, and contorted or fragmented axons with swollen varicosities containing αSyn and Tau. The identified neuropathological features closely resembled those in brains of p.A53T patients. Small molecules targeting αSyn reverted the degenerative phenotype under both basal and induced stress conditions, indicating a treatment strategy for PD and other synucleinopathies. Furthermore, mutant neurons showed disrupted synaptic connectivity and widespread transcriptional alterations in genes involved in synaptic signaling, a number of which have been previously linked to mental disorders, raising intriguing implications for potentially converging disease mechanisms.
Collapse
|
24
|
Yerabham ASK, Mas PJ, Decker C, Soares DC, Weiergräber OH, Nagel-Steger L, Willbold D, Hart DJ, Bradshaw NJ, Korth C. A structural organization for the Disrupted in Schizophrenia 1 protein, identified by high-throughput screening, reveals distinctly folded regions, which are bisected by mental illness-related mutations. J Biol Chem 2017; 292:6468-6477. [PMID: 28249940 DOI: 10.1074/jbc.m116.773903] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 02/22/2017] [Indexed: 11/06/2022] Open
Abstract
Disrupted in Schizophrenia 1 (DISC1) is a scaffolding protein of significant importance for neurodevelopment and a prominent candidate protein in the pathology of major mental illness. DISC1 modulates a number of critical neuronal signaling pathways through protein-protein interactions; however, the mechanism by which this occurs and how DISC1 causes mental illness is unclear, partly because knowledge of the structure of DISC1 is lacking. A lack of homology with known proteins has hindered attempts to define its domain composition. Here, we employed the high-throughput Expression of Soluble Proteins by Random Incremental Truncation (ESPRIT) technique to identify discretely folded regions of human DISC1 via solubility assessment of tens of thousands of fragments of recombinant DISC1. We identified four novel structured regions, named D, I, S, and C, at amino acids 257-383, 539-655, 635-738, and 691-836, respectively. One region (D) is located in a DISC1 section previously predicted to be unstructured. All regions encompass coiled-coil or α-helical structures, and three are involved in DISC1 oligomerization. Crucially, three of these domains would be lost or disrupted by a chromosomal translocation event after amino acid 597, which has been strongly linked to major mental illness. Furthermore, we observed that a known illness-related frameshift mutation after amino acid 807 causes the C region to form aberrantly multimeric and aggregated complexes with an unstable secondary structure. This newly revealed domain architecture of DISC1, therefore, provides a powerful framework for understanding the critical role of this protein in a variety of devastating mental illnesses.
Collapse
Affiliation(s)
| | - Philippe J Mas
- the Integrated Structural Biology Grenoble (ISBG) CNRS, CEA, Université Grenoble Alpes, EMBL, 38042 Grenoble, France
| | - Christina Decker
- the Institute of Physical Biology, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Dinesh C Soares
- the MRC Human Genetics Unit, Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, United Kingdom
| | - Oliver H Weiergräber
- the Institute of Complex Systems (ICS-6: Structural Biochemistry), Forschungszentrum Jülich, 52425 Jülich, Germany, and
| | - Luitgard Nagel-Steger
- the Institute of Physical Biology, Heinrich Heine University, 40225 Düsseldorf, Germany.,the Institute of Complex Systems (ICS-6: Structural Biochemistry), Forschungszentrum Jülich, 52425 Jülich, Germany, and
| | - Dieter Willbold
- the Institute of Physical Biology, Heinrich Heine University, 40225 Düsseldorf, Germany.,the Institute of Complex Systems (ICS-6: Structural Biochemistry), Forschungszentrum Jülich, 52425 Jülich, Germany, and
| | - Darren J Hart
- the Institute of Complex Systems (ICS-6: Structural Biochemistry), Forschungszentrum Jülich, 52425 Jülich, Germany, and
| | | | | |
Collapse
|
25
|
Dahoun T, Trossbach SV, Brandon NJ, Korth C, Howes OD. The impact of Disrupted-in-Schizophrenia 1 (DISC1) on the dopaminergic system: a systematic review. Transl Psychiatry 2017; 7:e1015. [PMID: 28140405 PMCID: PMC5299392 DOI: 10.1038/tp.2016.282] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 11/16/2016] [Accepted: 11/27/2016] [Indexed: 12/21/2022] Open
Abstract
Disrupted-in-Schizophrenia 1 (DISC1) is a gene known as a risk factor for mental illnesses possibly associated with dopamine impairments. DISC1 is a scaffold protein interacting with proteins involved in the dopamine system. Here we summarise the impact of DISC1 disruption on the dopamine system in animal models, considering its effects on presynaptic dopaminergic function (tyrosine hydroxylase levels, dopamine transporter levels, dopamine levels at baseline and after amphetamine administration) and postsynaptic dopaminergic function (dopamine D1 and D2 receptor levels, dopamine receptor-binding potential and locomotor activity after amphetamine administration). Our findings show that many but not all DISC1 models display (1) increased locomotion after amphetamine administration, (2) increased dopamine levels after amphetamine administration in the nucleus accumbens, and (3) inconsistent basal dopamine levels, dopamine receptor levels and binding potentials. There is also limited evidence for decreased tyrosine hydroxylase levels in the frontal cortex and increased dopamine transporter levels in the striatum but not nucleus accumbens, but these conclusions warrant further replication. The main dopaminergic findings are seen across different DISC1 models, providing convergent evidence that DISC1 has a role in regulating dopaminergic function. These results implicate dopaminergic dysregulation as a mechanism underlying the increased rate of schizophrenia seen in DISC1 variant carriers, and provide insights into how DISC1, and potentially DISC1-interacting proteins such as AKT and GSK-3, could be used as novel therapeutic targets for schizophrenia.
Collapse
Affiliation(s)
- T Dahoun
- Psychiatric Imaging Group MRC London Institute of Medical Sciences, Hammersmith Hospital, London, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College, Hammersmith Hospital, London, UK
- Department of the Institute of Clinical Sciences, Psychiatric Imaging Group, MRC London Institute of Medical Sciences (LMS), Imperial College-Hammersmith Hospital Campus, London, UK
| | - S V Trossbach
- Department of Neuropathology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - N J Brandon
- AstraZeneca Neuroscience, Innovative Medicines and Early Development Biotech Unit, R&D Boston, Waltham, MA, USA
| | - C Korth
- Department of Neuropathology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - O D Howes
- Psychiatric Imaging Group MRC London Institute of Medical Sciences, Hammersmith Hospital, London, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College, Hammersmith Hospital, London, UK
- Department of the Institute of Clinical Sciences, Psychiatric Imaging Group, MRC London Institute of Medical Sciences (LMS), Imperial College-Hammersmith Hospital Campus, London, UK
- Department of Psychosis Studies, Institute of Psychiatry, Neurology and Neuroscience (IoPPN), King's College London, London, UK
| |
Collapse
|
26
|
Cracco L, Notari S, Cali I, Sy MS, Chen SG, Cohen ML, Ghetti B, Appleby BS, Zou WQ, Caughey B, Safar JG, Gambetti P. Novel strain properties distinguishing sporadic prion diseases sharing prion protein genotype and prion type. Sci Rep 2017; 7:38280. [PMID: 28091514 PMCID: PMC5238384 DOI: 10.1038/srep38280] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 11/04/2016] [Indexed: 11/23/2022] Open
Abstract
In most human sporadic prion diseases the phenotype is consistently associated with specific pairings of the genotype at codon 129 of the prion protein gene and conformational properties of the scrapie PrP (PrPSc) grossly identified types 1 and 2. This association suggests that the 129 genotype favours the selection of a distinct strain that in turn determines the phenotype. However, this mechanism cannot play a role in the phenotype determination of sporadic fatal insomnia (sFI) and a subtype of sporadic Creutzfeldt-Jakob disease (sCJD) identified as sCJDMM2, which share 129 MM genotype and PrPSc type 2 but are associated with quite distinct phenotypes. Our detailed comparative study of the PrPSc conformers has revealed major differences between the two diseases, which preferentially involve the PrPSc component that is sensitive to digestion with proteases (senPrPSc) and to a lesser extent the resistant component (resPrPSc). We conclude that these variations are consistent with two distinct strains in sFI and sCJDMM2, and that the rarer sFI is the result of a variant strain selection pathway that might be favoured by a different brain site of initial PrPSc formation in the two diseases.
Collapse
Affiliation(s)
- Laura Cracco
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Silvio Notari
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Ignazio Cali
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Man-Sun Sy
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Shu G Chen
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Mark L Cohen
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, United States of America.,National Prion Disease Pathology Surveillance Center, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Bernardino Ghetti
- Department of Pathology and Laboratory Medicine, Division of Neuropathology, Indiana University, Indianapolis, Indiana, United States of America
| | - Brian S Appleby
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, United States of America.,National Prion Disease Pathology Surveillance Center, Case Western Reserve University, Cleveland, Ohio, United States of America.,Department of Neurology, Case Western Reserve University, Cleveland, Ohio, United States of America.,Department of Psychiatry, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Wen-Quan Zou
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, United States of America.,National Prion Disease Pathology Surveillance Center, Case Western Reserve University, Cleveland, Ohio, United States of America.,Department of Neurology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Byron Caughey
- Laboratory of Persistent Viral Diseases, NIH/NIAID Rocky Mountain Laboratories, Hamilton, Montana, United States of America
| | - Jiri G Safar
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, United States of America.,National Prion Disease Pathology Surveillance Center, Case Western Reserve University, Cleveland, Ohio, United States of America.,Department of Neurology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Pierluigi Gambetti
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, United States of America
| |
Collapse
|
27
|
Simultaneous effects on parvalbumin-positive interneuron and dopaminergic system development in a transgenic rat model for sporadic schizophrenia. Sci Rep 2016; 6:34946. [PMID: 27721451 PMCID: PMC5056355 DOI: 10.1038/srep34946] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 09/20/2016] [Indexed: 11/08/2022] Open
Abstract
To date, unequivocal neuroanatomical features have been demonstrated neither for sporadic nor for familial schizophrenia. Here, we investigated the neuroanatomical changes in a transgenic rat model for a subset of sporadic chronic mental illness (CMI), which modestly overexpresses human full-length, non-mutant Disrupted-in-Schizophrenia 1 (DISC1), and for which aberrant dopamine homeostasis consistent with some schizophrenia phenotypes has previously been reported. Neuroanatomical analysis revealed a reduced density of dopaminergic neurons in the substantia nigra and reduced dopaminergic fibres in the striatum. Parvalbumin-positive interneuron occurrence in the somatosensory cortex was shifted from layers II/III to V/VI, and the number of calbindin-positive interneurons was slightly decreased. Reduced corpus callosum thickness confirmed trend-level observations from in vivo MRI and voxel-wise tensor based morphometry. These neuroanatomical changes help explain functional phenotypes of this animal model, some of which resemble changes observed in human schizophrenia post mortem brain tissues. Our findings also demonstrate how a single molecular factor, DISC1 overexpression or misassembly, can account for a variety of seemingly unrelated morphological phenotypes and thus provides a possible unifying explanation for similar findings observed in sporadic schizophrenia patients. Our anatomical investigation of a defined model for sporadic mental illness enables a clearer definition of neuroanatomical changes associated with subsets of human sporadic schizophrenia.
Collapse
|
28
|
Tovo-Rodrigues L, Recamonde-Mendoza M, Paixão-Côrtes VR, Bruxel EM, Schuch JB, Friedrich DC, Rohde LA, Hutz MH. The role of protein intrinsic disorder in major psychiatric disorders. Am J Med Genet B Neuropsychiatr Genet 2016; 171:848-60. [PMID: 27184105 DOI: 10.1002/ajmg.b.32455] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 04/22/2016] [Indexed: 01/26/2023]
Abstract
Although new candidate genes for Autism Spectrum Disorder (ASD), Schizophrenia (SCZ), Attention-Deficit/Hyperactivity Disorder (ADHD), and Bipolar Disorder (BD) emerged from genome-wide association studies (GWAS), their underlying molecular mechanisms remain poorly understood. Evidences of the involvement of intrinsically disordered proteins in diseases have grown in the last decade. These proteins lack tridimensional structure under physiological conditions and are involved in important cellular functions such as signaling, recognition and regulation. The aim of the present study was to identify the role and abundance of intrinsically disordered proteins in a set of psychiatric diseases and to test whether diseases are different regarding protein intrinsic disorder. Our hypothesis is that differences across psychiatric illnesses phenotypes and symptoms may arise from differences in intrinsic protein disorder content and properties of each group. A bioinformatics prediction of intrinsic disorder was performed in proteins retrieved based on top findings from GWAS, Copy Number Variation and candidate gene investigations for each disease. This approach revealed that about 80% of studied proteins presented long stretches of disorder. This amount was significantly higher than that observed in general eukaryotic proteins, and those involved in cardiovascular diseases. These results suggest that proteins with intrinsic disorder are a common feature of neurodevelopment and synaptic transmission processes which are potentially involved in the etiology of psychiatric diseases. Moreover, we identified differences between ADHD and ASD when the binary prediction of structure and putative binding sites were compared. These differences may be related to variation in symptom complexity between both diseases. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Luciana Tovo-Rodrigues
- Department of Genetics, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil.,Postgraduate Program in Epidemiology, Federal University of Pelotas, Pelotas, RS, Brazil
| | - Mariana Recamonde-Mendoza
- Experimental and Molecular Cardiovascular Laboratory, Hospital de Clinicas de Porto Alegre, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | | - Estela M Bruxel
- Department of Genetics, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Jaqueline B Schuch
- Department of Genetics, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Deise C Friedrich
- Department of Genetics, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Luis A Rohde
- Child and Adolescent Psychiatric Division, Hospital de Clinicas de Porto Alegre, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil.,National Institute of Developmental Psychiatry for Children and Adolescents (INCT-CNPq), Brazil
| | - Mara H Hutz
- Department of Genetics, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
29
|
Furukubo-Tokunaga K, Kurita K, Honjo K, Pandey H, Ando T, Takayama K, Arai Y, Mochizuki H, Ando M, Kamiya A, Sawa A. DISC1 causes associative memory and neurodevelopmental defects in fruit flies. Mol Psychiatry 2016; 21:1232-43. [PMID: 26976042 PMCID: PMC4993648 DOI: 10.1038/mp.2016.15] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 01/16/2016] [Accepted: 01/20/2016] [Indexed: 01/18/2023]
Abstract
Originally found in a Scottish family with diverse mental disorders, the DISC1 protein has been characterized as an intracellular scaffold protein that associates with diverse binding partners in neural development. To explore its functions in a genetically tractable system, we expressed the human DISC1 in fruit flies (Drosophila melanogaster). As in mammalian neurons, DISC1 is localized to diverse subcellular domains of developing fly neurons including the nuclei, axons and dendrites. Overexpression of DISC1 impairs associative memory. Experiments with deletion/mutation constructs have revealed the importance of amino-terminal domain (46-290) for memory suppression whereas carboxyl domain (598-854) and the amino-terminal residues (1-45) including the nuclear localization signal (NLS1) are dispensable. DISC1 overexpression also causes suppression of axonal and dendritic branching of mushroom body neurons, which mediate a variety of cognitive functions in the fly brain. Analyses with deletion/mutation constructs reveal that protein domains 598-854 and 349-402 are both required for the suppression of axonal branching, while amino-terminal domains including NLS1 are dispensable. In contrast, NLS1 was required for the suppression of dendritic branching, suggesting a mechanism involving gene expression. Moreover, domain 403-596 is also required for the suppression of dendritic branching. We also show that overexpression of DISC1 suppresses glutamatergic synaptogenesis in developing neuromuscular junctions. Deletion/mutation experiments have revealed the importance of protein domains 403-596 and 349-402 for synaptic suppression, while amino-terminal domains including NLS1 are dispensable. Finally, we show that DISC1 functionally interacts with the fly homolog of Dysbindin (DTNBP1) via direct protein-protein interaction in developing synapses.
Collapse
Affiliation(s)
| | - Kazuki Kurita
- Life and Environmental Sciences, University of Tsukuba, Tsukuba 305-8572, Japan
| | - Ken Honjo
- Life and Environmental Sciences, University of Tsukuba, Tsukuba 305-8572, Japan
| | - Himani Pandey
- Life and Environmental Sciences, University of Tsukuba, Tsukuba 305-8572, Japan
| | - Tetsuya Ando
- Life and Environmental Sciences, University of Tsukuba, Tsukuba 305-8572, Japan
| | - Kojiro Takayama
- Life and Environmental Sciences, University of Tsukuba, Tsukuba 305-8572, Japan
| | - Yuko Arai
- Life and Environmental Sciences, University of Tsukuba, Tsukuba 305-8572, Japan
| | - Hiroaki Mochizuki
- Life and Environmental Sciences, University of Tsukuba, Tsukuba 305-8572, Japan
| | - Mai Ando
- Life and Environmental Sciences, University of Tsukuba, Tsukuba 305-8572, Japan
| | - Atsushi Kamiya
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore MD, USA
| | - Akira Sawa
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore MD, USA
| |
Collapse
|
30
|
Nucifora LG, Wu YC, Lee BJ, Sha L, Margolis RL, Ross CA, Sawa A, Nucifora FC. A Mutation in NPAS3 That Segregates with Schizophrenia in a Small Family Leads to Protein Aggregation. MOLECULAR NEUROPSYCHIATRY 2016; 2:133-144. [PMID: 27867938 DOI: 10.1159/000447358] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 05/31/2016] [Indexed: 12/21/2022]
Abstract
Schizophrenia and other major mental illnesses result from a complex interplay of genetic and environmental factors. We previously identified a mutation in NPAS3 that results in a valine to isoleucine (V304I) amino acid substitution segregating with schizophrenia in a small family. The amino acid change occurs in a potentially critical region for protein function. Furthermore, the same amino acid substitution in proteins related to familial Alzheimer's disease and transthyretin amyloidosis has been associated with protein aggregation. In this study, we demonstrate that NPAS3 is prone to aggregation, and that the V304I mutation in NPAS3 increases this propensity in both bacterial and mammalian expression systems. We also show that NPAS3-V304I reduces soluble endogenous NPAS3, and increases insoluble endogenous NPAS3 and leads to alteration of transcriptional activity. These results suggest that protein aggregation, potentially leading to cell dysfunction via a loss of protein function through sequestration, may contribute to the pathogenesis of schizophrenia and other forms of mental illness. Further exploration of the mechanisms leading to abnormal protein quality control could lead to new therapeutic targets.
Collapse
Affiliation(s)
- Leslie G Nucifora
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, Md., USA
| | - YeeWen Candace Wu
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, Md., USA
| | - Brian J Lee
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, Md., USA
| | - Li Sha
- Department of Biotechnology, Dalian Medical University, Dalian, China
| | - Russell L Margolis
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, Md., USA
| | - Christopher A Ross
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, Md., USA
| | - Akira Sawa
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, Md., USA
| | - Frederick C Nucifora
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, Md., USA
| |
Collapse
|
31
|
Increased dysbindin-1B isoform expression in schizophrenia and its propensity in aggresome formation. Cell Discov 2015; 1:15032. [PMID: 27462430 PMCID: PMC4860834 DOI: 10.1038/celldisc.2015.32] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 09/13/2015] [Indexed: 02/06/2023] Open
Abstract
Genetic variations in the human dysbindin-1 gene (DTNBP1) have been associated with schizophrenia. As a result of alternative splicing, the human DTNBP1 gene generates at least three distinct protein isoforms, dysbindin-1A, -1B and -1C. Significant effort has focused on dysbindin-1A, an important player in multiple steps of neurodevelopment. However, the other isoforms, dysbindin-1B and dysbindin-1C have not been well characterized. Nor have been associated with human diseases. Here we report an increase in expression of DTNBP1b mRNA in patients with paranoid schizophrenia as compared with healthy controls. A single-nucleotide polymorphism located in intron 9, rs117610176, has been identified and associated with paranoid schizophrenia, and its C allele leads to an increase of DTNBP1b mRNA splicing. Our data show that different dysbindin splicing isoforms exhibit distinct subcellular distribution, suggesting their distinct functional activities. Dysbindin-1B forms aggresomes at the perinuclear region, whereas dysbindin-1A and -1C proteins exhibit diffused patterns in the cytoplasm. Dysbindin-1A interacts with dysbindin-1B, getting recruited to the aggresome structure when co-expressed with dysbindin-1B. Moreover, cortical neurons over-expressing dysbindin-1B show reduction in neurite outgrowth, suggesting that dysbindin-1B may interfere with dysbindin-1A function in a dominant-negative manner. Taken together, our study uncovers a previously unknown association of DTNBP1b expression with schizophrenia in addition to its distinct biochemical and functional properties.
Collapse
|
32
|
Turck CW, Filiou MD. What Have Mass Spectrometry-Based Proteomics and Metabolomics (Not) Taught Us about Psychiatric Disorders? MOLECULAR NEUROPSYCHIATRY 2015; 1:69-75. [PMID: 27602358 PMCID: PMC4996030 DOI: 10.1159/000381902] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 03/25/2015] [Indexed: 12/16/2022]
Abstract
Understanding the molecular causes and finding appropriate therapies for psychiatric disorders are challenging tasks for research; -omics technologies are used to elucidate the molecular mechanisms underlying brain dysfunction in a hypothesis-free manner. In this review, we will focus on mass spectrometry-based proteomics and metabolomics and address how these approaches have contributed to our understanding of psychiatric disorders. Specifically, we will discuss what we have learned from mass spectrometry-based proteomics and metabolomics studies in rodent models and human cohorts, outline current limitations and discuss the potential of these methods for future applications in psychiatry.
Collapse
|
33
|
Millan MJ, Goodwin GM, Meyer-Lindenberg A, Ögren SO, Ögren SO. 60 years of advances in neuropsychopharmacology for improving brain health, renewed hope for progress. Eur Neuropsychopharmacol 2015; 25:591-8. [PMID: 25799919 DOI: 10.1016/j.euroneuro.2015.01.015] [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: 01/09/2015] [Accepted: 01/28/2015] [Indexed: 02/01/2023]
Abstract
Pharmacotherapy is effective in helping many patients suffering from psychiatric and neurological disorders, and both psychotherapeutic and stimulation-based techniques likewise have important roles to play in their treatment. However, therapeutic progress has recently been slow. Future success for improving the control and prevention of brain disorders will depend upon deeper insights into their causes and pathophysiological substrates. It will also necessitate new and more rigorous methods for identifying, validating, developing and clinically deploying new treatments. A field of Research and Development (R and D) that remains critical to this endeavour is Neuropsychopharmacology which transformed the lives of patients by introducing pharmacological treatments for psychiatric disorder some 60 years ago. For about half of this time, the European College of Neuropsychopharmacology (ECNP) has fostered efforts to enhance our understanding of the brain, and to improve the management of psychiatric disorders. Further, together with partners in academia and industry, and in discussions with regulators and patients, the ECNP is implicated in new initiatives to achieve this goal. This is then an opportune moment to survey the field, to analyse what we have learned from the achievements and failures of the past, and to identify major challenges for the future. It is also important to highlight strategies that are being put in place in the quest for more effective treatment of brain disorders: from experimental research and drug discovery to clinical development and collaborative ventures for reinforcing "R and D". The present article sets the scene, then introduces and interlinks the eight articles that comprise this Special Volume of European Neuropsychopharmacology. A broad-based suite of themes is covered embracing: the past, present and future of "R and D" for psychiatric disorders; complementary contributions of genetics and epigenetics; efforts to improve the treatment of depression, neurodevelopmental and neurodegenerative disorders; and advances in the analysis and neuroimaging of cellular and cerebral circuits.
Collapse
Affiliation(s)
- Mark J Millan
- Pole for Innovation in Neurosciences, IDR Servier, 125 chemin de ronde, 78290 Croissy sur Seine, France.
| | - Guy M Goodwin
- University Department of Psychiatry, Oxford University, Warneford Hospital, Oxford OX3 7JX, England
| | - Andreas Meyer-Lindenberg
- Central Institute of Mental Health, University of Heidelberg/Medical Faculty Mannheim, J5, D-68159 Mannheim, Germany
| | - Sven Ove Ögren
- Department of Neuroscience, Karolinska Institutet, Retzius väg 8, S-17177 Stockholm, Sweden
| | | |
Collapse
|
34
|
Propagation of dysbindin-1B aggregates: Exosome-mediated transmission of neurotoxic deposits. Neuroscience 2015; 291:301-16. [DOI: 10.1016/j.neuroscience.2015.02.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 02/09/2015] [Accepted: 02/09/2015] [Indexed: 11/21/2022]
|
35
|
Mas S, Gassó P, Parellada E, Bernardo M, Lafuente A. Network analysis of gene expression in peripheral blood identifies mTOR and NF-κB pathways involved in antipsychotic-induced extrapyramidal symptoms. THE PHARMACOGENOMICS JOURNAL 2015; 15:452-60. [PMID: 25623440 DOI: 10.1038/tpj.2014.84] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 09/22/2014] [Accepted: 11/05/2014] [Indexed: 02/06/2023]
Abstract
To identify the candidate genes for pharmacogenetic studies of antipsychotic (AP)-induced extrapyramidal symptoms (EPS), we propose a systems biology analytical approach, based on protein-protein interaction network construction and functional annotation analysis, of changes in gene expression (Human Genome U219 Array Plate) induced by treatment with risperidone or paliperidone in peripheral blood. 12 AP-naïve patients with first-episode psychosis participated in the present study. Our analysis revealed that, in response to AP treatment, constructed networks were enriched for different biological processes in patients without EPS (ubiquitination, protein folding and adenosine triphosphate (ATP) metabolism) compared with those presenting EPS (insulin receptor signaling, lipid modification, regulation of autophagy and immune response). Moreover, the observed differences also involved specific pathways, such as anaphase promoting complex /cdc20, prefoldin/CCT/triC and ATP synthesis in no-EPS patients, and mammalian target of rapamycin and NF-κB kinases in patients with EPS. Our results showing different patterns of gene expression in EPS patients, offer new and valuable markers for pharmacogenetic studies.
Collapse
Affiliation(s)
- S Mas
- Department Pathological Anatomy, Pharmacology and Microbiology, University of Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Barcelona, Spain
| | - P Gassó
- Department Pathological Anatomy, Pharmacology and Microbiology, University of Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - E Parellada
- Department Pathological Anatomy, Pharmacology and Microbiology, University of Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Barcelona, Spain.,Clinic Schizophrenia program, Psychiatry service, Hospital Clínic de Barcelona, Barcelona, Spain
| | - M Bernardo
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Barcelona, Spain.,Clinic Schizophrenia program, Psychiatry service, Hospital Clínic de Barcelona, Barcelona, Spain.,Department Psychiatry and Clinical Psychobiology, University of Barcelona, Barcelona, Spain
| | - A Lafuente
- Department Pathological Anatomy, Pharmacology and Microbiology, University of Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Barcelona, Spain
| |
Collapse
|
36
|
Ruocco LA, Treno C, Gironi Carnevale UA, Arra C, Boatto G, Pagano C, Tino A, Nieddu M, Michel M, Prikulis I, Carboni E, de Souza Silva MA, Huston JP, Sadile AG, Korth C. Immunization with DISC1 protein in an animal model of ADHD influences behavior and excitatory amino acids in prefrontal cortex and striatum. Amino Acids 2015; 47:637-50. [DOI: 10.1007/s00726-014-1897-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 12/09/2014] [Indexed: 12/11/2022]
|
37
|
Randall AD, Kurihara M, Brandon NJ, Brown JT. Disrupted in schizophrenia 1 and synaptic function in the mammalian central nervous system. Eur J Neurosci 2014; 39:1068-73. [PMID: 24712987 PMCID: PMC4232872 DOI: 10.1111/ejn.12500] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 01/02/2014] [Accepted: 01/07/2014] [Indexed: 11/28/2022]
Abstract
The disrupted in schizophrenia 1 (DISC1) gene is found at the breakpoint of an inherited chromosomal translocation, and segregates with major mental illnesses. Its potential role in central nervous system (CNS) malfunction has triggered intensive investigation of the biological roles played by DISC1, with the hope that this may shed new light on the pathobiology of psychiatric disease. Such work has ranged from investigations of animal behavior to detailed molecular-level analysis of the assemblies that DISC1 forms with other proteins. Here, we discuss the evidence for a role of DISC1 in synaptic function in the mammalian CNS.
Collapse
Affiliation(s)
- Andrew D Randall
- School of Physiology and Pharmacology, School of Medical Sciences, University of Bristol, Bristol, UK; Institute of Biomedical and Clinical Sciences, University of Exeter, The Hatherley Building, Prince of Wales Road, Exeter, EX4 4PS, UK
| | | | | | | |
Collapse
|
38
|
Marreiros R, Müller-Schiffmann A, Bader V, Selvarajah S, Dey D, Lingappa VR, Korth C. Viral capsid assembly as a model for protein aggregation diseases: Active processes catalyzed by cellular assembly machines comprising novel drug targets. Virus Res 2014; 207:155-64. [PMID: 25451064 DOI: 10.1016/j.virusres.2014.10.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 09/09/2014] [Accepted: 10/01/2014] [Indexed: 11/18/2022]
Abstract
Viruses can be conceptualized as self-replicating multiprotein assemblies, containing coding nucleic acids. Viruses have evolved to exploit host cellular components including enzymes to ensure their replicative life cycle. New findings indicate that also viral capsid proteins recruit host factors to accelerate their assembly. These assembly machines are RNA-containing multiprotein complexes whose composition is governed by allosteric sites. In the event of viral infection, the assembly machines are recruited to support the virus over the host and are modified to achieve that goal. Stress granules and processing bodies may represent collections of such assembly machines, readily visible by microscopy but biochemically labile and difficult to isolate by fractionation. We hypothesize that the assembly of protein multimers such as encountered in neurodegenerative or other protein conformational diseases, is also catalyzed by assembly machines. In the case of viral infection, the assembly machines have been modified by the virus to meet the virus' need for rapid capsid assembly rather than host homeostasis. In the case of the neurodegenerative diseases, it is the monomers and/or low n oligomers of the so-called aggregated proteins that are substrates of assembly machines. Examples for substrates are amyloid β peptide (Aβ) and tau in Alzheimer's disease, α-synuclein in Parkinson's disease, prions in the prion diseases, Disrupted-in-schizophrenia 1 (DISC1) in subsets of chronic mental illnesses, and others. A likely continuum between virus capsid assembly and cell-to-cell transmissibility of aggregated proteins is remarkable. Protein aggregation diseases may represent dysfunction and dysregulation of these assembly machines analogous to the aberrations induced by viral infection in which cellular homeostasis is pathologically reprogrammed. In this view, as for viral infection, reset of assembly machines to normal homeostasis should be the goal of protein aggregation therapeutics. A key basis for the commonality between viral and neurodegenerative disease aggregation is a broader definition of assembly as more than just simple aggregation, particularly suited for the crowded cytoplasm. The assembly machines are collections of proteins that catalytically accelerate an assembly reaction that would occur spontaneously but too slowly to be relevant in vivo. Being an enzyme complex with a functional allosteric site, appropriated for a non-physiological purpose (e.g. viral infection or conformational disease), these assembly machines present a superior pharmacological target because inhibition of their active site will amplify an effect on their substrate reaction. Here, we present this hypothesis based on recent proof-of-principle studies against Aβ assembly relevant in Alzheimer's disease.
Collapse
Affiliation(s)
- Rita Marreiros
- Department Neuropathology, Heinrich Heine University Düsseldorf Medical School, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Andreas Müller-Schiffmann
- Department Neuropathology, Heinrich Heine University Düsseldorf Medical School, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Verian Bader
- Department Neuropathology, Heinrich Heine University Düsseldorf Medical School, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | | | | | | | - Carsten Korth
- Department Neuropathology, Heinrich Heine University Düsseldorf Medical School, Moorenstrasse 5, 40225 Düsseldorf, Germany.
| |
Collapse
|
39
|
Bradshaw NJ, Bader V, Prikulis I, Lueking A, Müllner S, Korth C. Aggregation of the protein TRIOBP-1 and its potential relevance to schizophrenia. PLoS One 2014; 9:e111196. [PMID: 25333879 PMCID: PMC4205090 DOI: 10.1371/journal.pone.0111196] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 09/29/2014] [Indexed: 11/29/2022] Open
Abstract
We have previously proposed that specific proteins may form insoluble aggregates as a response to an illness-specific proteostatic dysbalance in a subset of brains from individuals with mental illness, as is the case for other chronic brain conditions. So far, established risk factors DISC1 and dysbindin were seen to specifically aggregate in a subset of such patients, as was a novel schizophrenia-related protein, CRMP1, identified through a condition-specific epitope discovery approach. In this process, antibodies are raised against the pooled insoluble protein fractions (aggregomes) of post mortem brain samples from schizophrenia patients, followed by epitope identification and confirmation using additional techniques. Pursuing this epitope discovery paradigm further, we reveal TRIO binding protein (TRIOBP) to be a major substrate of a monoclonal antibody with a high specificity to brain aggregomes from patients with chronic mental illness. TRIOBP is a gene previously associated with deafness which encodes for several distinct protein species, each involved in actin cytoskeletal dynamics. The 3′ splice variant TRIOBP-1 is found to be the antibody substrate and has a high aggregation propensity when over-expressed in neuroblastoma cells, while the major 5′ splice variant, TRIOBP-4, does not. Endogenous TRIOBP-1 can also spontaneously aggregate, doing so to a greater extent in cell cultures which are post-mitotic, consistent with aggregated TRIOBP-1 being able to accumulate in the differentiated neurons of the brain. Finally, upon expression in Neuroscreen-1 cells, aggregated TRIOBP-1 affects cell morphology, indicating that TRIOBP-1 aggregates may directly affect cell development, as opposed to simply being a by-product of other processes involved in major mental illness. While further experiments in clinical samples are required to clarify their relevance to chronic mental illness in the general population, TRIOBP-1 aggregates are thus implicated for the first time as a biological element of the neuropathology of a subset of chronic mental illness.
Collapse
Affiliation(s)
- Nicholas J. Bradshaw
- Department of Neuropathology, Heinrich Heine University, Düsseldorf, Germany
- * E-mail: (NJB); (CK)
| | - Verian Bader
- Department of Neuropathology, Heinrich Heine University, Düsseldorf, Germany
| | - Ingrid Prikulis
- Department of Neuropathology, Heinrich Heine University, Düsseldorf, Germany
| | | | | | - Carsten Korth
- Department of Neuropathology, Heinrich Heine University, Düsseldorf, Germany
- * E-mail: (NJB); (CK)
| |
Collapse
|
40
|
Trossbach SV, Fehsel K, Henning U, Winterer G, Luckhaus C, Schäble S, Silva MADS, Korth C. Peripheral DISC1 protein levels as a trait marker for schizophrenia and modulating effects of nicotine. Behav Brain Res 2014; 275:176-82. [PMID: 25218871 DOI: 10.1016/j.bbr.2014.08.064] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 08/31/2014] [Indexed: 11/25/2022]
Abstract
The Disrupted-in-Schizophrenia 1 (DISC1) protein plays a key role in behavioral control and vulnerability for mental illnesses, including schizophrenia. In this study we asked whether peripheral DISC1 protein levels in lymphocytes of patients diagnosed with schizophrenia can serve as a trait marker for the disease. Since a prominent comorbidity of schizophrenia patients is nicotine abuse or addiction, we also examined modulation of lymphocyte DISC1 protein levels in smokers, as well as the relationship between nicotine and DISC1 solubility status. We show decreased DISC1 levels in patients diagnosed with schizophrenia independent of smoking, indicating its potential use as a trait marker of this disease. In addition, lymphocytic DISC1 protein levels were decreased in smoking, mentally healthy individuals but not to the degree of overriding the trait level. Since DISC1 protein has been reported to exist in different solubility states in the brain, we also investigated DISC1 protein solubility in brains of rats treated with nicotine. Sub-chronic treatment with progressively increasing doses of nicotine from 0.25mg/kg to 1mg/kg for 15 days led to a decrease of insoluble DISC1 in the medial prefrontal cortex. Our results demonstrate that DISC1 protein levels in human lymphocytes are correlated with the diagnosis of schizophrenia independent of smoking and thus present a potential biomarker. Reduced DISC1 protein levels in lymphocytes of healthy individuals exposed to nicotine suggest that peripheral DISC1 could have potential for monitoring the effects of psychoactive substances.
Collapse
Affiliation(s)
- Svenja V Trossbach
- Department Neuropathology, Medical Faculty, Heinrich Heine University Düsseldorf, Germany
| | - Karin Fehsel
- Department of Psychiatry and Psychotherapy, Medical Faculty, Heinrich Heine University Düsseldorf, Germany
| | - Uwe Henning
- Department of Psychiatry and Psychotherapy, Medical Faculty, Heinrich Heine University Düsseldorf, Germany
| | - Georg Winterer
- Department of Psychiatry and Psychotherapy, Medical Faculty, Heinrich Heine University Düsseldorf, Germany
| | - Christian Luckhaus
- Department of Psychiatry and Psychotherapy, Medical Faculty, Heinrich Heine University Düsseldorf, Germany
| | - Sandra Schäble
- Center for Behavioral Neuroscience, Heinrich Heine University Düsseldorf, Germany
| | | | - Carsten Korth
- Department Neuropathology, Medical Faculty, Heinrich Heine University Düsseldorf, Germany.
| |
Collapse
|
41
|
Polajnar M, Zerovnik E. Impaired autophagy: a link between neurodegenerative and neuropsychiatric diseases. J Cell Mol Med 2014; 18:1705-11. [PMID: 25139375 PMCID: PMC4196646 DOI: 10.1111/jcmm.12349] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2014] [Accepted: 05/16/2014] [Indexed: 12/12/2022] Open
Abstract
Protein misfolding, and subsequent aggregation have been proven as the leading cause of most known dementias. Many of these, in addition to neurodegeneration, show profound changes in behaviour and thinking, thus, psychiatric symptoms. On the basis of the observation that progressive myoclonic epilepsies and neurodegenerative diseases share some common features of neurodegeneration, we proposed autophagy as a possible common impairment in these diseases. Here, we argue along similar lines for some neuropsychiatric conditions, among them depression and schizophrenia. We propose that existing and new therapies for these seemingly different diseases could be augmented with drugs used for neurodegenerative or neuropsychiatric diseases, respectively, among them some which modulate or augment autophagy.
Collapse
Affiliation(s)
- Mira Polajnar
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, Ljubljana, Slovenia
| | | |
Collapse
|
42
|
Yerabham AS, Weiergräber OH, Bradshaw NJ, Korth C. Revisiting Disrupted-in-Schizophrenia 1 as a scaffold protein. Biol Chem 2013; 394:1425-37. [DOI: 10.1515/hsz-2013-0178] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 07/03/2013] [Indexed: 01/28/2023]
Abstract
Abstract
Disrupted-in-Schizophrenia 1 (DISC1) is a widely-accepted genetic risk factor for schizophrenia and many other major mental illnesses. Traditionally DISC1 has been referred to as a ‘scaffold protein’ because of its ability to bind to a wide array of other proteins, including those of importance for neurodevelopment. Here, we review the characteristic properties shared between established scaffold proteins and DISC1. We find DISC1 to have many, but not all, of the characteristics of a scaffold protein, as it affects a considerable number of different, but related, signaling pathways, in most cases through inhibition of key enzymes. Using threading algorithms, the C-terminal portion of DISC1 could be mapped to extended helical structures, yet it may not closely resemble any of the known tertiary folds. While not completely fitting the classification of a classical scaffold protein, DISC1 does appear to be a tightly regulated and multi-faceted inhibitor of a wide range of enzymes from interrelated signaling cascades (Diverse Inhibitor of Signaling Cascades), which together contribute to neurodevelopment and synaptic homeostasis. Consequently, disruption of this complex regulation would be expected to lead to the range of major mental illnesses in which the DISC1 gene has been implicated.
Collapse
|
43
|
MESH Headings
- Causality
- Chromosome Breakpoints
- Chromosomes, Human, Pair 1/genetics
- Chromosomes, Human, Pair 1/ultrastructure
- Chromosomes, Human, Pair 11/genetics
- Chromosomes, Human, Pair 11/ultrastructure
- Conduct Disorder/genetics
- Genetic Predisposition to Disease
- Genome-Wide Association Study
- Humans
- Lod Score
- Mental Disorders/genetics
- Mood Disorders/genetics
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/physiology
- Pedigree
- Phenotype
- RNA, Long Noncoding/genetics
- Risk Factors
- Schizophrenia/genetics
- Terminology as Topic
- Translocation, Genetic
Collapse
Affiliation(s)
- P F Sullivan
- Departments of Genetics and Psychiatry, Center for Psychiatric Genomics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
44
|
Thomson PA, Malavasi ELV, Grünewald E, Soares DC, Borkowska M, Millar JK. DISC1 genetics, biology and psychiatric illness. FRONTIERS IN BIOLOGY 2013; 8:1-31. [PMID: 23550053 PMCID: PMC3580875 DOI: 10.1007/s11515-012-1254-7] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Psychiatric disorders are highly heritable, and in many individuals likely arise from the combined effects of genes and the environment. A substantial body of evidence points towards DISC1 being one of the genes that influence risk of schizophrenia, bipolar disorder and depression, and functional studies of DISC1 consequently have the potential to reveal much about the pathways that lead to major mental illness. Here, we review the evidence that DISC1 influences disease risk through effects upon multiple critical pathways in the developing and adult brain.
Collapse
Affiliation(s)
- Pippa A Thomson
- The Centre for Molecular Medicine at the Medical Research Council Institute of Genetics and Molecular Medicine, The University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK
| | | | | | | | | | | |
Collapse
|
45
|
Millan MJ. An epigenetic framework for neurodevelopmental disorders: from pathogenesis to potential therapy. Neuropharmacology 2012; 68:2-82. [PMID: 23246909 DOI: 10.1016/j.neuropharm.2012.11.015] [Citation(s) in RCA: 151] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2012] [Revised: 11/11/2012] [Accepted: 11/22/2012] [Indexed: 12/12/2022]
Abstract
Neurodevelopmental disorders (NDDs) are characterized by aberrant and delayed early-life development of the brain, leading to deficits in language, cognition, motor behaviour and other functional domains, often accompanied by somatic symptoms. Environmental factors like perinatal infection, malnutrition and trauma can increase the risk of the heterogeneous, multifactorial and polygenic disorders, autism and schizophrenia. Conversely, discrete genetic anomalies are involved in Down, Rett and Fragile X syndromes, tuberous sclerosis and neurofibromatosis, the less familiar Phelan-McDermid, Sotos, Kleefstra, Coffin-Lowry and "ATRX" syndromes, and the disorders of imprinting, Angelman and Prader-Willi syndromes. NDDs have been termed "synaptopathies" in reference to structural and functional disturbance of synaptic plasticity, several involve abnormal Ras-Kinase signalling ("rasopathies"), and many are characterized by disrupted cerebral connectivity and an imbalance between excitatory and inhibitory transmission. However, at a different level of integration, NDDs are accompanied by aberrant "epigenetic" regulation of processes critical for normal and orderly development of the brain. Epigenetics refers to potentially-heritable (by mitosis and/or meiosis) mechanisms controlling gene expression without changes in DNA sequence. In certain NDDs, prototypical epigenetic processes of DNA methylation and covalent histone marking are impacted. Conversely, others involve anomalies in chromatin-modelling, mRNA splicing/editing, mRNA translation, ribosome biogenesis and/or the regulatory actions of small nucleolar RNAs and micro-RNAs. Since epigenetic mechanisms are modifiable, this raises the hope of novel therapy, though questions remain concerning efficacy and safety. The above issues are critically surveyed in this review, which advocates a broad-based epigenetic framework for understanding and ultimately treating a diverse assemblage of NDDs ("epigenopathies") lying at the interface of genetic, developmental and environmental processes. This article is part of the Special Issue entitled 'Neurodevelopmental Disorders'.
Collapse
Affiliation(s)
- Mark J Millan
- Unit for Research and Discovery in Neuroscience, IDR Servier, 125 chemin de ronde, 78290 Croissy sur Seine, Paris, France.
| |
Collapse
|