1
|
Sarriés-Serrano U, Miquel-Rio L, Santana N, Paz V, Sancho-Alonso M, Callado LF, Meana JJ, Bortolozzi A. Impaired unfolded protein response, BDNF and synuclein markers in postmortem dorsolateral prefrontal cortex and caudate nucleus of patients with depression and Parkinson's disease. Prog Neuropsychopharmacol Biol Psychiatry 2025; 138:111299. [PMID: 40015617 DOI: 10.1016/j.pnpbp.2025.111299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 02/17/2025] [Accepted: 02/23/2025] [Indexed: 03/01/2025]
Abstract
Major depressive disorder (MDD) is characterized by significant impairment in social, emotional, and cognitive functioning. Its precise pathophysiology remains poorly understood. Alterations in protein homeostasis and some misfolded proteins have been identified within the brains of patients diagnosed with neuropsychiatric disorders. In contrast to neurodegenerative processes such as Parkinson's disease (PD), where the accumulation of aggregated α-synuclein (α-Syn) protein is a primary cause of significant neuronal loss, altered proteostasis in MDD may result in loss-of-function effects by modifying synaptic neuroplasticity. Moreover, aberrant activation of endoplasmic reticulum (ER) pathways may intensify the pathological alterations due to altered proteostasis. In this study, dorsolateral prefrontal cortex (dlPFC) and caudate nucleus from MDD patients and non-psychiatric controls were used. Postmortem samples of same brain areas from PD patients (Braak 2-3 and 5-6) and controls were also included. Protein levels of ER and unfolded protein response (UPR), synucleins (α-, β- and γ-Syn), and brain-derived neurotrophic factor (BDNF) were measured by Western-Blot. Phospho-eIF2α/eIF2α ratio was increased in the dlPFC and caudate nucleus of MDD and PD patients compared to their respective controls. Brain area-dependent changes in BiP and GRP94 levels were also found. We further detected accumulation of immature BDNF precursors and opposite changes in α- and β-Syn levels in the dlPFC of MDD and PD patients compared to controls. Our findings suggest that alterations in proteostasis contribute to the pathophysiology of MDD, as previously described in PD. A deeper understanding of the pathways involved will identify other candidate proteins and new targets with therapeutic potential.
Collapse
Affiliation(s)
- Unai Sarriés-Serrano
- Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain; Systems Neuropharmacology Research Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain; Biomedical Research Networking Center for Mental Health (CIBERSAM), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain; Department of Pharmacology, University of the Basque Country UPV/EHU, E-48940 Leioa, Bizkaia, Spain
| | - Lluis Miquel-Rio
- Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain; Systems Neuropharmacology Research Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain; Biomedical Research Networking Center for Mental Health (CIBERSAM), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
| | - Noemí Santana
- Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain; Systems Neuropharmacology Research Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain; Biomedical Research Networking Center for Mental Health (CIBERSAM), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
| | - Verónica Paz
- Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain; Systems Neuropharmacology Research Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain; Biomedical Research Networking Center for Mental Health (CIBERSAM), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
| | - María Sancho-Alonso
- Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain; Systems Neuropharmacology Research Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain; Biomedical Research Networking Center for Mental Health (CIBERSAM), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
| | - Luis F Callado
- Biomedical Research Networking Center for Mental Health (CIBERSAM), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain; Department of Pharmacology, University of the Basque Country UPV/EHU, E-48940 Leioa, Bizkaia, Spain; Biobizkaia Health Research Institute, 48903 Barakaldo, Spain
| | - J Javier Meana
- Biomedical Research Networking Center for Mental Health (CIBERSAM), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain; Department of Pharmacology, University of the Basque Country UPV/EHU, E-48940 Leioa, Bizkaia, Spain; Biobizkaia Health Research Institute, 48903 Barakaldo, Spain
| | - Analia Bortolozzi
- Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain; Systems Neuropharmacology Research Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain; Biomedical Research Networking Center for Mental Health (CIBERSAM), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain.
| |
Collapse
|
2
|
Bergstrom JJD, Fu MM. Dysregulation of myelination-related genes in schizophrenia. J Neurochem 2024; 168:2227-2242. [PMID: 39086020 PMCID: PMC11449665 DOI: 10.1111/jnc.16152] [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: 02/29/2024] [Revised: 06/03/2024] [Accepted: 06/05/2024] [Indexed: 08/02/2024]
Abstract
Schizophrenic individuals display disrupted myelination patterns, altered oligodendrocyte distribution, and abnormal oligodendrocyte morphology. Schizophrenia is linked with dysregulation of a variety of genes involved in oligodendrocyte function and myelin production. Single-nucleotide polymorphisms (SNPs) and rare mutations in myelination-related genes are observed in certain schizophrenic populations, representing potential genetic risk factors. Downregulation of myelination-related RNAs and proteins, particularly in frontal and limbic regions, is consistently associated with the disorder across multiple studies. These findings support the notion that disruptions in myelination may contribute to the cognitive and behavioral impairments experienced in schizophrenia, although further evidence of causation is needed.
Collapse
Affiliation(s)
| | - Meng-Meng Fu
- Department of Molecular and Cell Biology, UC Berkeley, Berkeley, California, USA
| |
Collapse
|
3
|
Zaharija B, Bradshaw NJ. Aggregation of Disrupted in Schizophrenia 1 arises from a central region of the protein. Prog Neuropsychopharmacol Biol Psychiatry 2024; 130:110923. [PMID: 38135095 DOI: 10.1016/j.pnpbp.2023.110923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/12/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023]
Abstract
An emerging approach to studying major mental illness is through proteostasis, with the identification of several proteins that form insoluble aggregates in the brains of patients. One of these is Disrupted in Schizophrenia 1 (DISC1), a neurodevelopmentally-important scaffold protein, and product of a classic schizophrenia risk gene. DISC1 aggregates have been detected in post mortem brain tissue from patients with schizophrenia, bipolar disorder and major depressive disorder, as well as various model systems, although the mechanism by which it aggregates is still unclear. Aggregation of two other proteins implicated in mental illness, TRIOBP-1 and NPAS3, was shown to be dependent on very specific structural regions of the protein. We therefore looked at the domain structure of DISC1, and investigated which structural elements are key for its aggregation. While none of the known structured DISC1 regions (named D, I, S and C respectively) formed aggregates individually when expressed in neuroblastoma cells, the combination of the D and I regions, plus the linker region between them, formed visible aggregates. Further refinement revealed that a region of approximately 30 amino acids between these two regions is critical for aggregation, and deletion of this region is sufficient to abolish the aggregation propensity of DISC1. This finding from mammalian cell culture contrasts with the recent determination that the C-region of DISC1 can aggregate in vitro, although some variations of the C-terminal of DISC1 could aggregate in our system. It therefore appears likely that DISC1 aggregation, implicated in mental illness, can occur through at least two distinct mechanisms.
Collapse
Affiliation(s)
- Beti Zaharija
- Faculty of Biotechnology and Drug Development, University of Rijeka, Croatia
| | - Nicholas J Bradshaw
- Faculty of Biotechnology and Drug Development, University of Rijeka, Croatia.
| |
Collapse
|
4
|
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: 5] [Impact Index Per Article: 2.5] [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
|
5
|
Samardžija B, Juković M, Zaharija B, Renner É, Palkovits M, Bradshaw NJ. Co-Aggregation and Parallel Aggregation of Specific Proteins in Major Mental Illness. Cells 2023; 12:1848. [PMID: 37508512 PMCID: PMC10378145 DOI: 10.3390/cells12141848] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/12/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND Disrupted proteostasis is an emerging area of research into major depressive disorder. Several proteins have been implicated as forming aggregates specifically in the brains of subsets of patients with psychiatric illnesses. These proteins include CRMP1, DISC1, NPAS3 and TRIOBP-1. It is unclear, however, whether these proteins normally aggregate together in the same individuals and, if so, whether each protein aggregates independently of each other ("parallel aggregation") or if the proteins physically interact and aggregate together ("co-aggregation"). MATERIALS AND METHODS Post mortem insular cortex samples from major depressive disorder and Alzheimer's disease patients, suicide victims and control individuals had their insoluble fractions isolated and tested by Western blotting to determine which of these proteins are insoluble and, therefore, likely to be aggregating. The ability of the proteins to co-aggregate (directly interact and form common aggregate structures) was tested by systematic pairwise expression of the proteins in SH-SY5Y neuroblastoma cells, which were then examined by immunofluorescent microscopy. RESULTS Many individuals displayed multiple insoluble proteins in the brain, although not enough to imply interaction between the proteins. Cell culture analysis revealed that only a few of the proteins analyzed can consistently co-aggregate with each other: DISC1 with each of CRMP1 and TRIOBP-1. DISC1 was able to induce aggregation of full length TRIOBP-1, but not individual domains of TRIOBP-1 when they were expressed individually. CONCLUSIONS While specific proteins are capable of co-aggregating, and appear to do so in the brains of individuals with mental illness and potentially also with suicidal tendency, it is more common for such proteins to aggregate in a parallel manner, through independent mechanisms. This information aids in understanding the distribution of protein aggregates among mental illness patients and is therefore important for any future diagnostic or therapeutic approaches based on this aspect of mental illness pathology.
Collapse
Affiliation(s)
- Bobana Samardžija
- Department of Biotechnology, University of Rijeka, 51000 Rijeka, Croatia
| | - Maja Juković
- Department of Biotechnology, University of Rijeka, 51000 Rijeka, Croatia
| | - Beti Zaharija
- Department of Biotechnology, University of Rijeka, 51000 Rijeka, Croatia
| | - Éva Renner
- Human Brain Tissue Bank & Laboratory, Semmelweis University, 1094 Budapest, Hungary
| | - Miklós Palkovits
- Human Brain Tissue Bank & Laboratory, Semmelweis University, 1094 Budapest, Hungary
| | | |
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
|
Zaharija B, Odorčić M, Hart A, Samardžija B, Marreiros R, Prikulis I, Juković M, Hyde TM, Kleinman JE, Korth C, Bradshaw NJ. TRIOBP-1 Protein Aggregation Exists in Both Major Depressive Disorder and Schizophrenia, and Can Occur through Two Distinct Regions of the Protein. Int J Mol Sci 2022; 23:ijms231911048. [PMID: 36232351 PMCID: PMC9569677 DOI: 10.3390/ijms231911048] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 09/14/2022] [Accepted: 09/15/2022] [Indexed: 11/25/2022] Open
Abstract
The presence of proteinopathy, the accumulation of specific proteins as aggregates in neurons, is an emerging aspect of the pathology of schizophrenia and other major mental illnesses. Among the initial proteins implicated in forming such aggregates in these conditions is Trio and F-actin Binding Protein isoform 1 (TRIOBP-1), a ubiquitously expressed protein involved in the stabilization of the actin cytoskeleton. Here we investigate the insolubility of TRIOBP-1, as an indicator of aggregation, in brain samples from 25 schizophrenia patients, 25 major depressive disorder patients and 50 control individuals (anterior cingulate cortex, BA23). Strikingly, insoluble TRIOBP-1 is considerably more prevalent in both of these conditions than in controls, further implicating TRIOBP-1 aggregation in schizophrenia and indicating a role in major depressive disorder. These results were only seen using a high stringency insolubility assay (previously used to study DISC1 and other proteins), but not a lower stringency assay that would be expected to also detect functional, actin-bound TRIOBP-1. Previously, we have also determined that a region of 25 amino acids in the center of this protein is critical for its ability to form aggregates. Here we attempt to refine this further, through the expression of various truncated mutant TRIOBP-1 vectors in neuroblastoma cells and examining their aggregation. In this way, it was possible to narrow down the aggregation-critical region of TRIOBP-1 to just 8 amino acids (333–340 of the 652 amino acid-long TRIOBP-1). Surprisingly our results suggested that a second section of TRIOBP-1 is also capable of independently inducing aggregation: the optionally expressed 59 amino acids at the extreme N-terminus of the protein. As a result, the 597 amino acid long version of TRIOBP-1 (also referred to as “Tara” or “TAP68”) has reduced potential to form aggregates. The presence of insoluble TRIOBP-1 in brain samples from patients, combined with insight into the mechanism of aggregation of TRIOBP-1 and generation of an aggregation-resistant mutant TRIOBP-1 that lacks both these regions, will be of significant use in further investigating the mechanism and consequences of TRIOBP-1 aggregation in major mental illness.
Collapse
Affiliation(s)
- Beti Zaharija
- Department of Biotechnology, University of Rijeka, 51000 Rijeka, Croatia
| | - Maja Odorčić
- Department of Biotechnology, University of Rijeka, 51000 Rijeka, Croatia
| | - Anja Hart
- Department of Biotechnology, University of Rijeka, 51000 Rijeka, Croatia
| | - Bobana Samardžija
- Department of Biotechnology, University of Rijeka, 51000 Rijeka, Croatia
| | - Rita Marreiros
- Department of Neuropathology, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Ingrid Prikulis
- Department of Neuropathology, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Maja Juković
- Department of Biotechnology, University of Rijeka, 51000 Rijeka, Croatia
| | - Thomas M. Hyde
- Lieber Institute for Brain Development, Baltimore, MD 21295, USA
- Department of Psychiatry and Behavioral Sciences, John Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, John Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Joel E. Kleinman
- Lieber Institute for Brain Development, Baltimore, MD 21295, USA
- Department of Psychiatry and Behavioral Sciences, John Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Carsten Korth
- Department of Neuropathology, Heinrich Heine University, 40225 Düsseldorf, Germany
- Correspondence: (C.K.); (N.J.B.)
| | - Nicholas J. Bradshaw
- Department of Biotechnology, University of Rijeka, 51000 Rijeka, Croatia
- Department of Neuropathology, Heinrich Heine University, 40225 Düsseldorf, Germany
- Correspondence: (C.K.); (N.J.B.)
| |
Collapse
|
8
|
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
|
9
|
Hui KK, Endo R, Sawa A, Tanaka M. A Perspective on the Potential Involvement of Impaired Proteostasis in Neuropsychiatric Disorders. Biol Psychiatry 2022; 91:335-345. [PMID: 34836635 PMCID: PMC8792182 DOI: 10.1016/j.biopsych.2021.09.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 08/05/2021] [Accepted: 09/01/2021] [Indexed: 11/19/2022]
Abstract
Recent genetic approaches have demonstrated that genetic factors contribute to the pathologic origins of neuropsychiatric disorders. Nevertheless, the exact pathophysiological mechanism for most cases remains unclear. Recent studies have demonstrated alterations in pathways of protein homeostasis (proteostasis) and identified several proteins that are misfolded and/or aggregated in the brains of patients with neuropsychiatric disorders, thus providing early evidence that disrupted proteostasis may be a contributing factor to their pathophysiology. Unlike neurodegenerative disorders in which massive neuronal and synaptic losses are observed, proteostasis impairments in neuropsychiatric disorders do not lead to robust neuronal death, but rather likely act via loss- and gain-of-function effects to disrupt neuronal and synaptic functions. Furthermore, abnormal activation of or overwhelmed endoplasmic reticulum and mitochondrial quality control pathways may exacerbate the pathophysiological changes initiated by impaired proteostasis, as these organelles are critical for proper neuronal functions and involved in the maintenance of proteostasis. This perspective article reviews recent findings implicating proteostasis impairments in the pathophysiology of neuropsychiatric disorders and explores how neuronal and synaptic functions may be impacted by disruptions in protein homeostasis. A greater understanding of the contributions by proteostasis impairment in neuropsychiatric disorders will help guide future studies to identify additional candidate proteins and new targets for therapeutic development.
Collapse
Affiliation(s)
- Kelvin K Hui
- Center for Autophagy Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas; Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Ryo Endo
- Laboratory for Protein Conformation Diseases, RIKEN Center for Brain Science, Wako, Saitama, Japan
| | - Akira Sawa
- Department of Psychiatry, John Hopkins University School of Medicine, Baltimore, Maryland; Department of Neuroscience, John Hopkins University School of Medicine, Baltimore, Maryland; Department of Biomedical Engineering, John Hopkins University School of Medicine, Baltimore, Maryland; Department of Genetic Medicine, John Hopkins University School of Medicine, Baltimore, Maryland; Department of Mental Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland
| | - Motomasa Tanaka
- Laboratory for Protein Conformation Diseases, RIKEN Center for Brain Science, Wako, Saitama, Japan.
| |
Collapse
|
10
|
Protein Aggregation of NPAS3, Implicated in Mental Illness, Is Not Limited to the V304I Mutation. J Pers Med 2021; 11:jpm11111070. [PMID: 34834422 PMCID: PMC8623263 DOI: 10.3390/jpm11111070] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/15/2021] [Accepted: 10/20/2021] [Indexed: 01/01/2023] Open
Abstract
An emerging phenomenon in our understanding of the pathophysiology of mental illness is the idea that specific proteins may form insoluble aggregates in the brains of patients, in partial analogy to similar proteinopathies in neurodegenerative diseases. Several proteins have now been detected as forming such aggregates in the brains of patients, including DISC1, dysbindin-1 and TRIOBP-1. Recently, neuronal PAS domain protein 3 (NPAS3), a known genetic risk factor for schizophrenia, was implicated through a V304I point mutation in a family with major mental illness. Investigation of the mutation revealed that it may lead to aggregation of NPAS3. Here we investigated NPAS3 aggregation in insular cortex samples from 40 individuals, by purifying the insoluble fraction of these samples and testing them by Western blotting. Strikingly, full-length NPAS3 was found in the insoluble fraction of 70% of these samples, implying that aggregation is far more widely spread than can be accounted for by this rare mutation. We investigated the possible mechanism of aggregation further in neuroblastoma cells, finding that oxidative stress plays a larger role than the V304I mutation. Finally, we tested to see if NPAS3 aggregation could also be seen in blood serum, as a more accessible tissue than the human brain for future diagnosis. While no indication of NPAS3 aggregation was seen in the serum, soluble NPAS3 was detected, and was more prevalent in patients with schizophrenia than in those with major depressive disorder or controls. Aggregation of NPAS3 therefore appears to be a widespread and multifactorial phenomenon. Further research is now needed to determine whether it is specifically enhanced in schizophrenia or other mental illnesses.
Collapse
|
11
|
Attili D, Schill DJ, DeLong CJ, Lim KC, Jiang G, Campbell KF, Walker K, Laszczyk A, McInnis MG, O'Shea KS. Astrocyte-Derived Exosomes in an iPSC Model of Bipolar Disorder. ADVANCES IN NEUROBIOLOGY 2020; 25:219-235. [PMID: 32578149 DOI: 10.1007/978-3-030-45493-7_8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Bipolar I Disorder (BP) is a serious, recurrent mood disorder that is characterized by alternating episodes of mania and depression. To begin to identify novel approaches and pathways involved in BP, we have obtained skin samples from BP patients and undiagnosed control (C) individuals, reprogrammed them to form induced pluripotent stem cells (iPSC), and then differentiated the stem cells into astrocytes. RNAs from BP and C astrocytes were extracted and RNAseq analysis carried out. 501 differentially expressed genes were identified, including genes for cytoskeletal elements, extracellular matrix, signaling pathways, neurodegeneration, and notably transcripts that identify exosomes. When we compared highly expressed genes using hierarchial cluster analysis, "Exosome" was the first and most highly significant cluster identified, p < 5 × 10-13, Benjamini correction. Exosomes are membrane-bound vesicles that package and remove toxic proteins from cells and also enable cell to cell communication. They carry genetic material, including DNA, mRNA and microRNAs, proteins, and lipids to target cells throughout the body. Exosomes are released by cortical neurons and astrocytes in culture and are present in BP vs C postmortem brain tissue. Little is known about what transcripts and proteins are targeted to neurons, how they regulate biological functions of the acceptor cell, or how that may be altered in mood disorders. Since astrocyte-derived exosomes have been suggested to promote neuronal plasticity, as well as to remove toxic proteins in the brain, alterations in their function or content may be involved in neurodevelopmental, neuropathological, and neuropsychiatric conditions. To examine exosome cargos and interactions with neural precursor cells, astrocytes were differentiated from four bipolar disorder (BP) and four control (C) iPSC lines. Culture supernatants from these astrocytes were collected, and exosomes isolated by ultra-centrifugation. Western blot analysis demonstrated the presence of the exosome markers CD9, CD81, and Hsp70. Nanosight technology was used to characterize exosomes from each astrocyte cell line, suggesting that exosomes were slightly more concentrated in culture supernatants derived from BP compared with C astrocytes but there was no difference in the mean sizes of the exosomes. Analysis of their function in neuronal differentiation is being carried out by labeling exosomes derived from bipolar patient and control astrocytes and adding them to control neural progenitor cells. Given the current interest in clearing toxic proteins from brains of patients with neurodegenerative disorders, exosomes may present similar opportunities in BP.
Collapse
Affiliation(s)
- D Attili
- Department of Cell and Developmental Biology, The University of Michigan, Ann Arbor, MI, USA
| | - D J Schill
- Department of Cell and Developmental Biology, The University of Michigan, Ann Arbor, MI, USA
| | - C J DeLong
- Department of Cell and Developmental Biology, The University of Michigan, Ann Arbor, MI, USA
| | - K C Lim
- Department of Cell and Developmental Biology, The University of Michigan, Ann Arbor, MI, USA
| | - G Jiang
- Department of Cell and Developmental Biology, The University of Michigan, Ann Arbor, MI, USA
| | - K F Campbell
- Department of Cell and Developmental Biology, The University of Michigan, Ann Arbor, MI, USA
| | - K Walker
- Department of Cell and Developmental Biology, The University of Michigan, Ann Arbor, MI, USA
| | - A Laszczyk
- Department of Cell and Developmental Biology, The University of Michigan, Ann Arbor, MI, USA
| | - M G McInnis
- Department of Psychiatry, The University of Michigan, Ann Arbor, MI, USA
| | - K S O'Shea
- Department of Cell and Developmental Biology, The University of Michigan, Ann Arbor, MI, USA.
- Department of Psychiatry, The University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
12
|
Norkett R, Lesept F, Kittler JT. DISC1 Regulates Mitochondrial Trafficking in a Miro1-GTP-Dependent Manner. Front Cell Dev Biol 2020; 8:449. [PMID: 32637409 PMCID: PMC7317294 DOI: 10.3389/fcell.2020.00449] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 05/13/2020] [Indexed: 11/20/2022] Open
Abstract
The disrupted in schizophrenia 1 (DISC1) protein is implicated in major mental illnesses including schizophrenia and bipolar disorder. A key feature of psychiatric disease is aberrant synaptic communication. Correct synaptic transmission is dependent on spatiotemporally regulated energy provision and calcium buffering. This can be achieved by precise distribution of mitochondria throughout the elaborate architecture of the neuron. Central to this process is the calcium sensor and GTPase Miro1, which allows mitochondrial trafficking by molecular motors. While the role of Miro1-calcium binding in mitochondrial transport is well described, far less is known regarding the functions of the two GTPase domains. Here, we investigate the effects of a psychiatric disease-associated mutation in DISC1 on mitochondrial trafficking. We show that this DISC1 mutation impairs Miro1’s ability to transport mitochondria. We also demonstrate the necessity of the first Miro1 GTPase domain in determining direction of mitochondrial transport and the involvement of DISC1 in this process. Finally, we describe the effects of mutant DISC1 on positioning of mitochondria at synapses.
Collapse
Affiliation(s)
- Rosalind Norkett
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - Flavie Lesept
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - Josef T Kittler
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| |
Collapse
|
13
|
Disruption of cellular proteostasis by H1N1 influenza A virus causes α-synuclein aggregation. Proc Natl Acad Sci U S A 2020; 117:6741-6751. [PMID: 32152117 DOI: 10.1073/pnas.1906466117] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Neurodegenerative diseases feature specific misfolded or misassembled proteins associated with neurotoxicity. The precise mechanisms by which protein aggregates first arise in the majority of sporadic cases have remained unclear. Likely, a first critical mass of misfolded proteins starts a vicious cycle of a prion-like expansion. We hypothesize that viruses, having evolved to hijack the host cellular machinery for catalyzing their replication, lead to profound disturbances of cellular proteostasis, resulting in such a critical mass of protein aggregates. Here, we investigated the effect of influenza virus (H1N1) strains on proteostasis of proteins associated with neurodegenerative diseases in Lund human mesencephalic dopaminergic cells in vitro and infection of Rag knockout mice in vivo. We demonstrate that acute H1N1 infection leads to the formation of α-synuclein and Disrupted-in-Schizophrenia 1 (DISC1) aggregates, but not of tau or TDP-43 aggregates, indicating a selective effect on proteostasis. Oseltamivir phosphate, an antiinfluenza drug, prevented H1N1-induced α-synuclein aggregation. As a cell pathobiological mechanism, we identified H1N1-induced blocking of autophagosome formation and inhibition of autophagic flux. In addition, α-synuclein aggregates appeared in infected cell populations connected to the olfactory bulbs following intranasal instillation of H1N1 in Rag knockout mice. We propose that H1N1 virus replication in neuronal cells can induce seeds of aggregated α-synuclein or DISC1 that may be able to initiate further detrimental downstream events and should thus be considered a risk factor in the pathogenesis of synucleinopathies or a subset of mental disorders. More generally, aberrant proteostasis induced by viruses may be an underappreciated factor in initiating protein misfolding.
Collapse
|
14
|
Bradshaw NJ, Trossbach SV, Köber S, Walter S, Prikulis I, Weggen S, Korth C. Disrupted in Schizophrenia 1 regulates the processing of reelin in the perinatal cortex. Schizophr Res 2020; 215:506-513. [PMID: 28433501 DOI: 10.1016/j.schres.2017.04.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 04/03/2017] [Accepted: 04/04/2017] [Indexed: 02/06/2023]
Abstract
Disrupted in Schizophrenia 1 (DISC1) is a prominent gene in mental illness research, encoding a scaffold protein known to be of importance in the developing cerebral cortex. Reelin is a critical extracellular protein for development and lamination of the prenatal cortex and which has also been independently implicated in mental illness. Regulation of reelin activity occurs through processing by the metalloproteinases ADAMTS-4 and ADAMTS-5. Through cross-breeding of heterozygous transgenic DISC1 mice with heterozygous reeler mice, which have reduced reelin, pups heterozygous for both phenotypes were generated. From these, we determine that transgenic DISC1 leads to a reduction in the processing of reelin, with implications for its downstream signalling element Dab1. An effect of DISC1 on reelin processing was confirmed in vitro, and revealed that intracellular DISC1 affects ADAMTS-4 protein, which in turn is exported and affects processing of extracellular reelin. In transgenic rat cortical cultures, an effect of DISC1 on reelin processing could also be seen specifically in early, immature neurons, but was lost in calretinin and reelin-positive mature neurons, suggesting cell-type specificity. DISC1 therefore acts upstream of reelin in the perinatal cerebral cortex in a cell type/time specific manner, leading to regulation of its activity through altered proteolytic cleavage. Thus a functional link is demonstrated between two proteins, each of independent importance for both cortical development and associated cognitive functions leading to behavioural maladaptation and mental illness.
Collapse
Affiliation(s)
- Nicholas J Bradshaw
- Department of Neuropathology, Heinrich Heine University, 40225 Düsseldorf, Germany.
| | - Svenja V Trossbach
- Department of Neuropathology, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Sabrina Köber
- Department of Neuropathology, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Susanne Walter
- Department of Neuropathology, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Ingrid Prikulis
- Department of Neuropathology, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Sascha Weggen
- Department of Neuropathology, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Carsten Korth
- Department of Neuropathology, Heinrich Heine University, 40225 Düsseldorf, Germany.
| |
Collapse
|
15
|
Niemeyer C, Matosin N, Kaul D, Philipsen A, Gassen NC. The Role of Cathepsins in Memory Functions and the Pathophysiology of Psychiatric Disorders. Front Psychiatry 2020; 11:718. [PMID: 32793006 PMCID: PMC7393989 DOI: 10.3389/fpsyt.2020.00718] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 07/08/2020] [Indexed: 01/27/2023] Open
Abstract
Cathepsins are proteases with functions in cellular homeostasis, lysosomal degradation and autophagy. Their role in the development of neurodegenerative diseases has been extensively studied. It is well established that impairment of proper cathepsin function plays a crucial role in the pathophysiology of neurodegenerative diseases, and in recent years a role for cathepsins in mental disorders has emerged given the involvement of cathepsins in memory function, hyperactivity, and in depression- and anxiety-like behavior. Here we review putative cathepsin functions with a special focus on their role in the pathophysiology of psychiatric diseases. Specifically, cathepsins are crucial for maintaining cellular homeostasis, particularly as part of the autophagy machinery of neural strategies underlying acute stress response. Disruption of cathepsin functions can lead to psychiatric diseases such as major depressive disease (MDD), bipolar disorder, and schizophrenia. Specifically, cathepsins can be excreted via a process called secretory autophagy. Thereby, they are able to regulate extracellular factors such as brain-derived neurotrophic factor and perlecan c-terminal fragment LG3 providing maintenance of neuronal homeostasis and mediating neuronal plasticity in response to acute stress or trauma. In addition, impairment of proper cathepsin function can result in impaired synaptic transmission by compromised recycling and biogenesis of synaptic vesicles. Taken together, further investigations on cathepsin functions and stress response, neuroplasticity, and synaptic transmission will be of great interest in understanding the pathophysiology of psychiatric disorders.
Collapse
Affiliation(s)
- Christine Niemeyer
- Neurohomeostasis Research Group, Department of Psychiatry and Psychotherapy, University Hospital Bonn, Bonn, Germany
| | - Natalie Matosin
- Faculty of Science, Medicine and Health, Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia.,Molecular Horizons, School of Chemistry and Molecular Biosciences, University of Wollongong, Wollongong, NSW, Australia.,Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| | - Dominic Kaul
- Faculty of Science, Medicine and Health, Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia.,Molecular Horizons, School of Chemistry and Molecular Biosciences, University of Wollongong, Wollongong, NSW, Australia
| | - Alexandra Philipsen
- Neurohomeostasis Research Group, Department of Psychiatry and Psychotherapy, University Hospital Bonn, Bonn, Germany
| | - Nils C Gassen
- Neurohomeostasis Research Group, Department of Psychiatry and Psychotherapy, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
16
|
Nucifora LG, MacDonald ML, Lee BJ, Peters ME, Norris AL, Orsburn BC, Yang K, Gleason K, Margolis RL, Pevsner J, Tamminga CA, Sweet RA, Ross CA, Sawa A, Nucifora FC. Increased Protein Insolubility in Brains From a Subset of Patients With Schizophrenia. Am J Psychiatry 2019; 176:730-743. [PMID: 31055969 DOI: 10.1176/appi.ajp.2019.18070864] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE The mechanisms leading to schizophrenia are likely to be diverse. However, there may be common pathophysiological pathways for subtypes of the disease. The authors tested the hypothesis that increased protein insolubility and ubiquitination underlie the pathophysiology for a subtype of schizophrenia. METHODS Prefrontal cortex and superior temporal gyrus from postmortem brains of individuals with and without schizophrenia were subjected to cold sarkosyl fractionation, separating proteins into soluble and insoluble fractions. Protein insolubility and ubiquitin levels were quantified for each insoluble fraction, with normalization to total homogenate protein. Mass spectrometry analysis was then performed to identify the protein contents of the insoluble fractions. The potential biological relevance of the detected proteins was assessed using Gene Ontology enrichment analysis and Ingenuity Pathway Analysis. RESULTS A subset of the schizophrenia brains showed an increase in protein insolubility and ubiquitination in the insoluble fraction. Mass spectrometry of the insoluble fraction revealed that brains with increased insolubility and ubiquitination exhibited a similar peptide expression by principal component analysis. The proteins that were significantly altered in the insoluble fraction were enriched for pathways relating to axon target recognition as well as nervous system development and function. CONCLUSIONS This study suggests a pathological process related to protein insolubility for a subset of patients with schizophrenia. Determining the molecular mechanism of this subtype of schizophrenia could lead to a better understanding of the pathways underlying the clinical phenotype in some patients with major mental illness as well as to improved nosology and identification of novel therapeutic targets.
Collapse
Affiliation(s)
- Leslie G Nucifora
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Matthew L MacDonald
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Brian J Lee
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Matthew E Peters
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Alexis L Norris
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Benjamin C Orsburn
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Kun Yang
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Kelly Gleason
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Russell L Margolis
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Jonathan Pevsner
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Carol A Tamminga
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Robert A Sweet
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Christopher A Ross
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Akira Sawa
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Frederick C Nucifora
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| |
Collapse
|
17
|
Protein misassembly and aggregation as potential convergence points for non-genetic causes of chronic mental illness. Mol Psychiatry 2019; 24:936-951. [PMID: 30089789 DOI: 10.1038/s41380-018-0133-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 06/10/2018] [Accepted: 06/18/2018] [Indexed: 12/13/2022]
Abstract
Chronic mental illnesses (CMI), such as schizophrenia or recurrent affective disorders, are complex conditions with both genetic and non-genetic elements. In many other chronic brain conditions, including Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis and frontotemporal dementia, sporadic instances of the disease are more common than gene-driven familial cases. Yet, the pathology of these conditions can be characterized by the presence of aberrant protein homeostasis, proteostasis, resulting in misfolded or aggregated proteins in the brains of patients that predominantly do not derive from genetic mutations. While visible deposits of aggregated protein have not yet been detected in CMI patients, we propose the existence of more subtle protein misassembly in these conditions, which form a continuum with the psychiatric phenotypes found in the early stages of many neurodegenerative conditions. Such proteinopathies need not rely on genetic variation. In a similar manner to the established aberrant neurotransmitter homeostasis in CMI, aberrant homeostasis of proteins is a functional statement that can only partially be explained by, but is certainly complementary to, genetic approaches. Here, we review evidence for aberrant proteostasis signatures from post mortem human cases, in vivo animal work, and in vitro analysis of candidate proteins misassembled in CMI. The five best-characterized proteins in this respect are currently DISC1, dysbindin-1, CRMP1, TRIOBP-1, and NPAS3. Misassembly of these proteins with inherently unstructured domains is triggered by extracellular stressors and thus provides a converging point for non-genetic causes of CMI.
Collapse
|
18
|
Dysregulation of a specific immune-related network of genes biologically defines a subset of schizophrenia. Transl Psychiatry 2019; 9:156. [PMID: 31150013 PMCID: PMC6544656 DOI: 10.1038/s41398-019-0486-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 03/22/2019] [Accepted: 04/29/2019] [Indexed: 12/31/2022] Open
Abstract
Currently, the clinical diagnosis of schizophrenia relies solely on self-reporting and clinical interview, and likely comprises heterogeneous biological subsets. Such subsets may be defined by an underlying biology leading to solid biomarkers. A transgenic rat model modestly overexpressing the full-length, non-mutant Disrupted-in-Schizophrenia 1 (DISC1) protein (tgDISC1 rat) was generated that defines such a subset, inspired by our previous identification of insoluble DISC1 protein in post mortem brains from patients with chronic mental illness. Besides specific phenotypes such as DISC1 protein pathology, abnormal dopamine homeostasis, and changes in neuroanatomy and behavior, this animal model also shows subtle disturbances in overarching signaling pathways relevant for schizophrenia. In a reverse-translational approach, assuming that both the animal model and a patient subset share common disturbed signaling pathways, we identified differentially expressed transcripts from peripheral blood mononuclear cells of tgDISC1 rats that revealed an interconnected set of dysregulated genes, led by decreased expression of regulator of G-protein signaling 1 (RGS1), chemokine (C-C) ligand 4 (CCL4), and other immune-related transcripts enriched in T-cell and macrophage signaling and converging in one module after weighted gene correlation network analysis. Testing expression of this gene network in two independent cohorts of patients with schizophrenia versus healthy controls (n = 16/50 and n = 54/45) demonstrated similar expression changes. The two top markers RGS1 and CCL4 defined a subset of 27% of patients with 97% specificity. Thus, analogous aberrant signaling pathways can be identified by a blood test in an animal model and a corresponding schizophrenia patient subset, suggesting that in this animal model tailored pharmacotherapies for this patient subset could be achieved.
Collapse
|
19
|
Proteomic Studies Reveal Disrupted in Schizophrenia 1 as a Player in Both Neurodevelopment and Synaptic Function. Int J Mol Sci 2018; 20:ijms20010119. [PMID: 30597994 PMCID: PMC6337115 DOI: 10.3390/ijms20010119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 12/21/2018] [Accepted: 12/24/2018] [Indexed: 02/03/2023] Open
Abstract
A balanced chromosomal translocation disrupting DISC1 (Disrupted in Schizophrenia 1) gene has been linked to psychiatric diseases, such as major depression, bipolar disorder and schizophrenia. Since the discovery of this translocation, many studies have focused on understating the role of the truncated isoform of DISC1, hypothesizing that the gain of function of this protein could be behind the neurobiology of mental conditions, but not so many studies have focused in the mechanisms impaired due to its loss of function. For that reason, we performed an analysis on the cellular proteome of primary neurons in which DISC1 was knocked down with the goal of identifying relevant pathways directly affected by DISC1 loss of function. Using an unbiased proteomic approach, we found that the expression of 31 proteins related to neurodevelopment (e.g., CRMP-2, stathmin) and synaptic function (e.g., MUNC-18, NCS-1) is altered by DISC1 in primary mouse neurons. Hence, this study reinforces the idea that DISC1 is a unifying regulator of both neurodevelopment and synaptic function, thereby providing a link between these two key anatomical and cellular circuitries.
Collapse
|
20
|
Ryskalin L, Limanaqi F, Frati A, Busceti CL, Fornai F. mTOR-Related Brain Dysfunctions in Neuropsychiatric Disorders. Int J Mol Sci 2018; 19:ijms19082226. [PMID: 30061532 PMCID: PMC6121884 DOI: 10.3390/ijms19082226] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 07/26/2018] [Accepted: 07/27/2018] [Indexed: 12/12/2022] Open
Abstract
The mammalian target of rapamycin (mTOR) is an ubiquitously expressed serine-threonine kinase, which senses and integrates several intracellular and environmental cues to orchestrate major processes such as cell growth and metabolism. Altered mTOR signalling is associated with brain malformation and neurological disorders. Emerging evidence indicates that even subtle defects in the mTOR pathway may produce severe effects, which are evident as neurological and psychiatric disorders. On the other hand, administration of mTOR inhibitors may be beneficial for a variety of neuropsychiatric alterations encompassing neurodegeneration, brain tumors, brain ischemia, epilepsy, autism, mood disorders, drugs of abuse, and schizophrenia. mTOR has been widely implicated in synaptic plasticity and autophagy activation. This review addresses the role of mTOR-dependent autophagy dysfunction in a variety of neuropsychiatric disorders, to focus mainly on psychiatric syndromes including schizophrenia and drug addiction. For instance, amphetamines-induced addiction fairly overlaps with some neuropsychiatric disorders including neurodegeneration and schizophrenia. For this reason, in the present review, a special emphasis is placed on the role of mTOR on methamphetamine-induced brain alterations.
Collapse
Affiliation(s)
- Larisa Ryskalin
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy.
| | - Fiona Limanaqi
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy.
| | | | | | - Francesco Fornai
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy.
- I.R.C.C.S. Neuromed, Via Atinense 18, 86077 Isernia, Italy.
| |
Collapse
|
21
|
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
|
22
|
Sialana FJ, Wang AL, Fazari B, Kristofova M, Smidak R, Trossbach SV, Korth C, Huston JP, de Souza Silva MA, Lubec G. Quantitative Proteomics of Synaptosomal Fractions in a Rat Overexpressing Human DISC1 Gene Indicates Profound Synaptic Dysregulation in the Dorsal Striatum. Front Mol Neurosci 2018; 11:26. [PMID: 29467617 PMCID: PMC5808171 DOI: 10.3389/fnmol.2018.00026] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 01/18/2018] [Indexed: 12/12/2022] Open
Abstract
Disrupted-in-schizophrenia 1 (DISC1) is a key protein involved in behavioral processes and various mental disorders, including schizophrenia and major depression. A transgenic rat overexpressing non-mutant human DISC1, modeling aberrant proteostasis of the DISC1 protein, displays behavioral, biochemical and anatomical deficits consistent with aspects of mental disorders, including changes in the dorsal striatum, an anatomical region critical in the development of behavioral disorders. Herein, dorsal striatum of 10 transgenic DISC1 (tgDISC1) and 10 wild type (WT) littermate control rats was used for synaptosomal preparations and for performing liquid chromatography-tandem mass spectrometry (LC-MS)-based quantitative proteomics, using isobaric labeling (TMT10plex). Functional enrichment analysis was generated from proteins with level changes. The increase in DISC1 expression leads to changes in proteins and synaptic-associated processes including membrane trafficking, ion transport, synaptic organization and neurodevelopment. Canonical pathway analysis assigned proteins with level changes to actin cytoskeleton, Gαq, Rho family GTPase and Rho GDI, axonal guidance, ephrin receptor and dopamine-DARPP32 feedback in cAMP signaling. DISC1-regulated proteins proposed in the current study are also highly associated with neurodevelopmental and mental disorders. Bioinformatics analyses from the current study predicted that the following biological processes may be activated by overexpression of DISC1, i.e., regulation of cell quantities, neuronal and axonal extension and long term potentiation. Our findings demonstrate that the effects of overexpression of non-mutant DISC1 or its misassembly has profound consequences on protein networks essential for behavioral control. These results are also relevant for the interpretation of previous as well as for the design of future studies on DISC1.
Collapse
Affiliation(s)
- Fernando J Sialana
- Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - An-Li Wang
- Center for Behavioral Neuroscience, University of Düsseldorf, Düsseldorf, Germany
| | - Benedetta Fazari
- Center for Behavioral Neuroscience, University of Düsseldorf, Düsseldorf, Germany
| | - Martina Kristofova
- Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Roman Smidak
- Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Svenja V Trossbach
- Department of Neuropathology, Heinrich-Heine University of Düsseldorf, Düsseldorf, Germany
| | - Carsten Korth
- Department of Neuropathology, Heinrich-Heine University of Düsseldorf, Düsseldorf, Germany
| | - Joseph P Huston
- Center for Behavioral Neuroscience, University of Düsseldorf, Düsseldorf, Germany
| | | | - Gert Lubec
- Department of Neuroproteomics, Paracelsus Private Medical University, Salzburg, Austria
| |
Collapse
|
23
|
Shao L, Lu B, Wen Z, Teng S, Wang L, Zhao Y, Wang L, Ishizuka K, Xu X, Sawa A, Song H, Ming G, Zhong Y. Disrupted-in-Schizophrenia-1 (DISC1) protein disturbs neural function in multiple disease-risk pathways. Hum Mol Genet 2018; 26:2634-2648. [PMID: 28472294 DOI: 10.1093/hmg/ddx147] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 04/13/2017] [Indexed: 12/20/2022] Open
Abstract
Although the genetic contribution is under debate, biological studies in multiple mouse models have suggested that the Disrupted-in-Schizophrenia-1 (DISC1) protein may contribute to susceptibility to psychiatric disorders. In the present study, we took the advantages of the Drosophila model to dissect the molecular pathways that can be affected by DISC1 in the context of pathology-related phenotypes. We found that three pathways that include the homologs of Drosophila Dys, Trio, and Shot were downregulated by introducing a C-terminal truncated mutant DISC1. Consistently, these three molecules were downregulated in the induced pluripotent stem cell-derived forebrain neurons from the subjects carrying a frameshift deletion in DISC1 C-terminus. Importantly, the three pathways were underscored in the pathophysiology of psychiatric disorders in bioinformatics analysis. Taken together, our findings are in line with the polygenic theory of psychiatric disorders.
Collapse
Affiliation(s)
- Lisha Shao
- Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, MOE Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China.,Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Binyan Lu
- Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, MOE Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China.,State Key Laboratory of Systematic and Evolutionary Botany, Institute of Botany, The Chinese Academy of Sciences, Beijing 100093, P.R. China
| | - Zhexing Wen
- Departments of Psychiatry and Behavioral Sciences, Cell Biology, and Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Shaolei Teng
- Department of Biology, Howard University, Washington, DC 20059, USA
| | - Lingling Wang
- Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, MOE Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China
| | - Yi Zhao
- Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, MOE Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China
| | - Liyuan Wang
- Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, MOE Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China
| | - Koko Ishizuka
- Molecular Psychiatry Program, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Xiufeng Xu
- Department of Psychiatry, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Akira Sawa
- Molecular Psychiatry Program, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Hongjun Song
- Departments of Psychiatry and Behavioral Sciences, Cell Biology, and Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Guoli Ming
- Departments of Psychiatry and Behavioral Sciences, Cell Biology, and Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Yi Zhong
- Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, MOE Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China.,Cold Spring Harbor Lab, Cold Spring Harbor, NY 11724, USA
| |
Collapse
|
24
|
Yerabham ASK, Müller-Schiffmann A, Ziehm T, Stadler A, Köber S, Indurkhya X, Marreiros R, Trossbach SV, Bradshaw NJ, Prikulis I, Willbold D, Weiergräber OH, Korth C. Biophysical insights from a single chain camelid antibody directed against the Disrupted-in-Schizophrenia 1 protein. PLoS One 2018; 13:e0191162. [PMID: 29324815 PMCID: PMC5764400 DOI: 10.1371/journal.pone.0191162] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 12/31/2017] [Indexed: 01/17/2023] Open
Abstract
Accumulating evidence suggests an important role for the Disrupted-in-Schizophrenia 1 (DISC1) protein in neurodevelopment and chronic mental illness. In particular, the C-terminal 300 amino acids of DISC1 have been found to mediate important protein-protein interactions and to harbor functionally important phosphorylation sites and disease-associated polymorphisms. However, long disordered regions and oligomer-forming subdomains have so far impeded structural analysis. VHH domains derived from camelid heavy chain only antibodies are minimal antigen binding modules with appreciable solubility and stability, which makes them well suited for the stabilizing proteins prior to structural investigation. Here, we report on the generation of a VHH domain derived from an immunized Lama glama, displaying high affinity for the human DISC1 C region (aa 691-836), and its characterization by surface plasmon resonance, size exclusion chromatography and immunological techniques. The VHH-DISC1 (C region) complex was also used for structural investigation by small angle X-ray scattering analysis. In combination with molecular modeling, these data support predictions regarding the three-dimensional fold of this DISC1 segment as well as its steric arrangement in complex with our VHH antibody.
Collapse
Affiliation(s)
- Antony S. K. Yerabham
- Department of Neuropathology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | | | - Tamar Ziehm
- Institute of Complex Systems (ICS-6: Structural Biochemistry), Forschungszentrum Jülich, Jülich, Germany
| | - Andreas Stadler
- Jülich Centre for Neutron Science JCNS and Institute for Complex Systems ICS, Forschungszentrum Jülich, Jülich, Germany
| | - Sabrina Köber
- Department of Neuropathology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Xela Indurkhya
- Department of Neuropathology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Rita Marreiros
- Department of Neuropathology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Svenja V. Trossbach
- Department of Neuropathology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Nicholas J. Bradshaw
- Department of Neuropathology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Ingrid Prikulis
- Department of Neuropathology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Dieter Willbold
- Institute of Complex Systems (ICS-6: Structural Biochemistry), Forschungszentrum Jülich, Jülich, Germany
- Institute for Physical Biology and BMFZ, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Oliver H. Weiergräber
- Institute of Complex Systems (ICS-6: Structural Biochemistry), Forschungszentrum Jülich, Jülich, Germany
| | - Carsten Korth
- Department of Neuropathology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
25
|
Kaneta H, Ukai W, Tsujino H, Furuse K, Kigawa Y, Tayama M, Ishii T, Hashimoto E, Kawanishi C. Antipsychotics promote GABAergic interneuron genesis in the adult rat brain: Role of heat-shock protein production. J Psychiatr Res 2017; 92:108-118. [PMID: 28414930 DOI: 10.1016/j.jpsychires.2017.03.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 03/07/2017] [Accepted: 03/08/2017] [Indexed: 02/07/2023]
Abstract
Current antipsychotics reduce positive symptoms and reverse negative symptoms in conjunction with cognitive behavioral issues with the goal of restoring impaired occupational and social functioning. However, limited information is available on their influence on gliogenesis or their neurogenic properties in adult schizophrenia brains, particularly on GABAergic interneuron production. In the present study, we used young adult subventricular zone (SVZ)-derived progenitor cells expressing proteoglycan NG2 cultures to examine the oligodendrocyte and GABAergic interneuron genesis effects of several kinds of antipsychotics on changes in differentiation function induced by exposure to the NMDA receptor antagonist MK-801. We herein demonstrated that antipsychotics promoted or restored changes in the oligodendrocyte/GABAergic interneuron differentiation functions of NG2(+) cells induced by the exposure to MK-801, which was considered to be one of the drug-induced schizophrenia model. We also demonstrated that antipsychotics restored heat-shock protein (HSP) production in NG2(+) cells with differentiation impairment. The antipsychotics olanzapine, aripiprazole, and blonanserin, but not haloperidol increased HSP90 levels, which were reduced by the exposure to MK-801. Our results showed that antipsychotics, particularly those recently synthesized, exerted similar GABAergic interneuron genesis effects on NG2(+) neuronal/glial progenitor cells in the adult rat brain by increasing cellular HSP production, and also suggest that HSP90 may play a crucial role in the pathophysiology of schizophrenia and is a key target for next drug development.
Collapse
Affiliation(s)
- Hiroo Kaneta
- Department of Neuropsychiatry, Sapporo Medical University, School of Medicine, S-1, W-16, Chuo-ku, Sapporo, 0608543, Japan.
| | - Wataru Ukai
- Department of Neuropsychiatry, Sapporo Medical University, School of Medicine, S-1, W-16, Chuo-ku, Sapporo, 0608543, Japan.
| | - Hanako Tsujino
- Department of Neuropsychiatry, Sapporo Medical University, School of Medicine, S-1, W-16, Chuo-ku, Sapporo, 0608543, Japan.
| | - Kengo Furuse
- Department of Neuropsychiatry, Sapporo Medical University, School of Medicine, S-1, W-16, Chuo-ku, Sapporo, 0608543, Japan.
| | - Yoshiyasu Kigawa
- Department of Neuropsychiatry, Sapporo Medical University, School of Medicine, S-1, W-16, Chuo-ku, Sapporo, 0608543, Japan.
| | - Masaya Tayama
- Department of Neuropsychiatry, Sapporo Medical University, School of Medicine, S-1, W-16, Chuo-ku, Sapporo, 0608543, Japan.
| | - Takao Ishii
- Department of Neuropsychiatry, Sapporo Medical University, School of Medicine, S-1, W-16, Chuo-ku, Sapporo, 0608543, Japan.
| | - Eri Hashimoto
- Department of Neuropsychiatry, Sapporo Medical University, School of Medicine, S-1, W-16, Chuo-ku, Sapporo, 0608543, Japan.
| | - Chiaki Kawanishi
- Department of Neuropsychiatry, Sapporo Medical University, School of Medicine, S-1, W-16, Chuo-ku, Sapporo, 0608543, Japan.
| |
Collapse
|
26
|
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
|
27
|
Tanaka M, Ishizuka K, Nekooki-Machida Y, Endo R, Takashima N, Sasaki H, Komi Y, Gathercole A, Huston E, Ishii K, Hui KKW, Kurosawa M, Kim SH, Nukina N, Takimoto E, Houslay MD, Sawa A. Aggregation of scaffolding protein DISC1 dysregulates phosphodiesterase 4 in Huntington's disease. J Clin Invest 2017; 127:1438-1450. [PMID: 28263187 DOI: 10.1172/jci85594] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 01/11/2017] [Indexed: 01/19/2023] Open
Abstract
Huntington's disease (HD) is a polyglutamine (polyQ) disease caused by aberrant expansion of the polyQ tract in Huntingtin (HTT). While motor impairment mediated by polyQ-expanded HTT has been intensively studied, molecular mechanisms for nonmotor symptoms in HD, such as psychiatric manifestations, remain elusive. Here we have demonstrated that HTT forms a ternary protein complex with the scaffolding protein DISC1 and cAMP-degrading phosphodiesterase 4 (PDE4) to regulate PDE4 activity. We observed pathological cross-seeding between DISC1 and mutant HTT aggregates in the brains of HD patients as well as in a murine model that recapitulates the polyQ pathology of HD (R6/2 mice). In R6/2 mice, consequent reductions in soluble DISC1 led to dysregulation of DISC1-PDE4 complexes, aberrantly increasing the activity of PDE4. Importantly, exogenous expression of a modified DISC1, which binds to PDE4 but not mutant HTT, normalized PDE4 activity and ameliorated anhedonia in the R6/2 mice. We propose that cross-seeding of mutant HTT and DISC1 and the resultant changes in PDE4 activity may underlie the pathology of a specific subset of mental manifestations of HD, which may provide an insight into molecular signaling in mental illness in general.
Collapse
|
28
|
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
|
29
|
Misassembly of full-length Disrupted-in-Schizophrenia 1 protein is linked to altered dopamine homeostasis and behavioral deficits. Mol Psychiatry 2016; 21:1561-1572. [PMID: 26754951 PMCID: PMC5078859 DOI: 10.1038/mp.2015.194] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 10/16/2015] [Accepted: 10/22/2015] [Indexed: 12/21/2022]
Abstract
Disrupted-in-schizophrenia 1 (DISC1) is a mental illness gene first identified in a Scottish pedigree. So far, DISC1-dependent phenotypes in animal models have been confined to expressing mutant DISC1. Here we investigated how pathology of full-length DISC1 protein could be a major mechanism in sporadic mental illness. We demonstrate that a novel transgenic rat model, modestly overexpressing the full-length DISC1 transgene, showed phenotypes consistent with a significant role of DISC1 misassembly in mental illness. The tgDISC1 rat displayed mainly perinuclear DISC1 aggregates in neurons. Furthermore, the tgDISC1 rat showed a robust signature of behavioral phenotypes that includes amphetamine supersensitivity, hyperexploratory behavior and rotarod deficits, all pointing to changes in dopamine (DA) neurotransmission. To understand the etiology of the behavioral deficits, we undertook a series of molecular studies in the dorsal striatum of tgDISC1 rats. We observed an 80% increase in high-affinity DA D2 receptors, an increased translocation of the dopamine transporter to the plasma membrane and a corresponding increase in DA inflow as observed by cyclic voltammetry. A reciprocal relationship between DISC1 protein assembly and DA homeostasis was corroborated by in vitro studies. Elevated cytosolic dopamine caused an increase in DISC1 multimerization, insolubility and complexing with the dopamine transporter, suggesting a physiological mechanism linking DISC1 assembly and dopamine homeostasis. DISC1 protein pathology and its interaction with dopamine homeostasis is a novel cellular mechanism that is relevant for behavioral control and may have a role in mental illness.
Collapse
|
30
|
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
|
31
|
Umeda K, Iritani S, Fujishiro H, Sekiguchi H, Torii Y, Habuchi C, Kuroda K, Kaibuchi K, Ozaki N. Immunohistochemical evaluation of the GABAergic neuronal system in the prefrontal cortex of a DISC1 knockout mouse model of schizophrenia. Synapse 2016; 70:508-518. [PMID: 27421906 DOI: 10.1002/syn.21924] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 06/27/2016] [Accepted: 07/11/2016] [Indexed: 01/23/2023]
Abstract
The etiology of schizophrenia remains unknown. However, using molecular biological techniques, some candidate genes have been identified that might be associated with the disease. One of these candidate genes, disrupted-in-schizophrenia 1 (DISC1), was found in a large Scottish family with multiple mental illnesses. The function of DISC1 is considered to be associated with axon elongation and neuron migration in the central nervous system, but the functional consequences of defects in this gene have not been fully clarified in brain neuronal systems. Dysfunction of the gamma-aminobutyric acid (GABA)ergic neuronal system is also considered to contribute to the pathogenesis of schizophrenia. Thus, to clarify the neuropathological changes associated with DISC1 dysfunction, we investigated the number and distribution of GABAergic neurons in the prefrontal cortex of DISC1 knockout mice. We immunohistochemically quantified the laminar density of GABAergic neurons using anti-parvalbumin and anti-calbindin D28k antibodies (markers of GABAergic neuronal subpopulations). We found that the densities of both parvalbumin- and calbindin-immunoreactive neurons in the anterior cingulate, medial prefrontal, and orbitofrontal cortices were markedly lower in DISC1 knockout mice than in wild-type mice. In addition, reductions in cell density were observed in layers II and III and the deep layers of the cortex. This reduction in GABAergic neuronal density was not associated with alterations in neuronal size. These findings suggest that disrupted GABAergic neuronal network formation due to a DISC1 deficit might be involved in the pathophysiology of schizophrenia.
Collapse
Affiliation(s)
- Kentaro Umeda
- Department of Psychiatry, Graduate School of Medicine, Nagoya University, 65 Tsurumai, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Shuji Iritani
- Department of Psychiatry, Graduate School of Medicine, Nagoya University, 65 Tsurumai, Showa-ku, Nagoya, Aichi, 466-8550, Japan.
| | - Hiroshige Fujishiro
- Department of Psychiatry, Graduate School of Medicine, Nagoya University, 65 Tsurumai, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Hirotaka Sekiguchi
- Department of Psychiatry, Graduate School of Medicine, Nagoya University, 65 Tsurumai, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Youta Torii
- Department of Psychiatry, Graduate School of Medicine, Nagoya University, 65 Tsurumai, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Chikako Habuchi
- Department of Psychiatry, Graduate School of Medicine, Nagoya University, 65 Tsurumai, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Keisuke Kuroda
- Department of Cell Pharmacology, Graduate School of Medicine, Nagoya University, 65 Tsurumai, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Kozo Kaibuchi
- Department of Cell Pharmacology, Graduate School of Medicine, Nagoya University, 65 Tsurumai, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Norio Ozaki
- Department of Psychiatry, Graduate School of Medicine, Nagoya University, 65 Tsurumai, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| |
Collapse
|
32
|
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
|
33
|
Tomoda T, Sumitomo A, Jaaro-Peled H, Sawa A. Utility and validity of DISC1 mouse models in biological psychiatry. Neuroscience 2016; 321:99-107. [PMID: 26768401 PMCID: PMC4803604 DOI: 10.1016/j.neuroscience.2015.12.061] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Revised: 12/31/2015] [Accepted: 12/31/2015] [Indexed: 11/26/2022]
Abstract
We have seen an era of explosive progress in translating neurobiology into etiological understanding of mental disorders for the past 10-15 years. The discovery of Disrupted-in-schizophrenia 1 (DISC1) gene was one of the major driving forces that have contributed to the progress. The finding that DISC1 plays crucial roles in neurodevelopment and synapse regulation clearly underscored the utility and validity of DISC1-related biology in advancing our understanding of pathophysiological processes underlying psychiatric conditions. Despite recent genetic studies that failed to identify DISC1 as a risk gene for sporadic cases of schizophrenia, DISC1 mutant mice, coupled with various environmental stressors, have proven successful in satisfying face validity as models of a wide range of human psychiatric conditions. Investigating mental disorders using these models is expected to further contribute to the circuit-level understanding of the pathological mechanisms, as well as to the development of novel therapeutic strategies in the future.
Collapse
Affiliation(s)
- T Tomoda
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan.
| | - A Sumitomo
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - H Jaaro-Peled
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - A Sawa
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
34
|
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
|
35
|
Norkett R, Modi S, Birsa N, Atkin TA, Ivankovic D, Pathania M, Trossbach SV, Korth C, Hirst WD, Kittler JT. DISC1-dependent Regulation of Mitochondrial Dynamics Controls the Morphogenesis of Complex Neuronal Dendrites. J Biol Chem 2015; 291:613-29. [PMID: 26553875 PMCID: PMC4705382 DOI: 10.1074/jbc.m115.699447] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Indexed: 01/09/2023] Open
Abstract
The DISC1 protein is implicated in major mental illnesses including schizophrenia, depression, bipolar disorder, and autism. Aberrant mitochondrial dynamics are also associated with major mental illness. DISC1 plays a role in mitochondrial transport in neuronal axons, but its effects in dendrites have yet to be studied. Further, the mechanisms of this regulation and its role in neuronal development and brain function are poorly understood. Here we have demonstrated that DISC1 couples to the mitochondrial transport and fusion machinery via interaction with the outer mitochondrial membrane GTPase proteins Miro1 and Miro2, the TRAK1 and TRAK2 mitochondrial trafficking adaptors, and the mitochondrial fusion proteins (mitofusins). Using live cell imaging, we show that disruption of the DISC1-Miro-TRAK complex inhibits mitochondrial transport in neurons. We also show that the fusion protein generated from the originally described DISC1 translocation (DISC1-Boymaw) localizes to the mitochondria, where it similarly disrupts mitochondrial dynamics. We also show by super resolution microscopy that DISC1 is localized to endoplasmic reticulum contact sites and that the DISC1-Boymaw fusion protein decreases the endoplasmic reticulum-mitochondria contact area. Moreover, disruption of mitochondrial dynamics by targeting the DISC1-Miro-TRAK complex or upon expression of the DISC1-Boymaw fusion protein impairs the correct development of neuronal dendrites. Thus, DISC1 acts as an important regulator of mitochondrial dynamics in both axons and dendrites to mediate the transport, fusion, and cross-talk of these organelles, and pathological DISC1 isoforms disrupt this critical function leading to abnormal neuronal development.
Collapse
Affiliation(s)
- Rosalind Norkett
- From the Department of Neuroscience, Physiology, and Pharmacology, University College London, Gower Street, London WC1E 6BT, United Kingdom
| | - Souvik Modi
- From the Department of Neuroscience, Physiology, and Pharmacology, University College London, Gower Street, London WC1E 6BT, United Kingdom
| | - Nicol Birsa
- From the Department of Neuroscience, Physiology, and Pharmacology, University College London, Gower Street, London WC1E 6BT, United Kingdom
| | - Talia A Atkin
- From the Department of Neuroscience, Physiology, and Pharmacology, University College London, Gower Street, London WC1E 6BT, United Kingdom
| | - Davor Ivankovic
- From the Department of Neuroscience, Physiology, and Pharmacology, University College London, Gower Street, London WC1E 6BT, United Kingdom
| | - Manav Pathania
- From the Department of Neuroscience, Physiology, and Pharmacology, University College London, Gower Street, London WC1E 6BT, United Kingdom
| | - Svenja V Trossbach
- the Department of Neuropathology, Heinrich Heine University, Moorenstrasse 5, 40225 Dusseldorf, Germany
| | - Carsten Korth
- the Department of Neuropathology, Heinrich Heine University, Moorenstrasse 5, 40225 Dusseldorf, Germany
| | - Warren D Hirst
- the Neuroscience Research Unit, Pfizer, Cambridge, Massachusetts 02139, and
| | - Josef T Kittler
- From the Department of Neuroscience, Physiology, and Pharmacology, University College London, Gower Street, London WC1E 6BT, United Kingdom,
| |
Collapse
|
36
|
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]
|
37
|
Lee SA, Kim SM, Suh BK, Sun HY, Park YU, Hong JH, Park C, Nguyen MD, Nagata KI, Yoo JY, Park SK. Disrupted-in-schizophrenia 1 (DISC1) regulates dysbindin function by enhancing its stability. J Biol Chem 2015; 290:7087-96. [PMID: 25635053 DOI: 10.1074/jbc.m114.614750] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Dysbindin and DISC1 are schizophrenia susceptibility factors playing roles in neuronal development. Here we show that the physical interaction between dysbindin and DISC1 is critical for the stability of dysbindin and for the process of neurite outgrowth. We found that DISC1 forms a complex with dysbindin and increases its stability in association with a reduction in ubiquitylation. Furthermore, knockdown of DISC1 or expression of a deletion mutant, DISC1 lacking amino acid residues 403-504 of DISC1 (DISC1(Δ403-504)), effectively decreased levels of endogenous dysbindin. Finally, the neurite outgrowth defect induced by knockdown of DISC1 was partially reversed by coexpression of dysbindin. Taken together, these results indicate that dysbindin and DISC1 form a physiologically functional complex that is essential for normal neurite outgrowth.
Collapse
Affiliation(s)
- Seol-Ae Lee
- From the Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Seong-Mo Kim
- From the Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Bo Kyoung Suh
- From the Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Hwa-Young Sun
- From the Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Young-Un Park
- From the Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Ji-Ho Hong
- From the Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Cana Park
- From the Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Minh Dang Nguyen
- the Hotchkiss Brain Institute, Departments of Clinical Neurosciences, Cell Biology and Anatomy, and Biochemistry and Molecular Biology, University of Calgary, Calgary T2N 4N1, Canada, and
| | - Koh-Ichi Nagata
- the Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Human Service Center, 713-8 Kamiya, Kasugai 480-0392, Japan
| | - Joo-Yeon Yoo
- From the Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Sang Ki Park
- From the Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea,
| |
Collapse
|
38
|
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]
|
39
|
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
|
40
|
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
|
41
|
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
|
42
|
Roles of heat shock factor 1 in neuronal response to fetal environmental risks and its relevance to brain disorders. Neuron 2014; 82:560-72. [PMID: 24726381 DOI: 10.1016/j.neuron.2014.03.002] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2014] [Indexed: 12/25/2022]
Abstract
Prenatal exposure of the developing brain to various environmental challenges increases susceptibility to late onset of neuropsychiatric dysfunction; still, the underlying mechanisms remain obscure. Here we show that exposure of embryos to a variety of environmental factors such as alcohol, methylmercury, and maternal seizure activates HSF1 in cerebral cortical cells. Furthermore, Hsf1 deficiency in the mouse cortex exposed in utero to subthreshold levels of these challenges causes structural abnormalities and increases seizure susceptibility after birth. In addition, we found that human neural progenitor cells differentiated from induced pluripotent stem cells derived from schizophrenia patients show higher variability in the levels of HSF1 activation induced by environmental challenges compared to controls. We propose that HSF1 plays a crucial role in the response of brain cells to prenatal environmental insults and may be a key component in the pathogenesis of late-onset neuropsychiatric disorders.
Collapse
|
43
|
Regulation of N-methyl-D-aspartate receptors by disrupted-in-schizophrenia-1. Biol Psychiatry 2014; 75:414-424. [PMID: 23906531 PMCID: PMC3864617 DOI: 10.1016/j.biopsych.2013.06.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 06/12/2013] [Accepted: 06/19/2013] [Indexed: 02/06/2023]
Abstract
BACKGROUND Genetic studies have implicated disrupted-in-schizophrenia-1 (DISC1) as a risk factor for a wide range of mental conditions, including schizophrenia. Because N-methyl-D-aspartate receptor (NMDAR) dysfunction has been strongly linked to the pathophysiology of these conditions, we examined whether the NMDAR is a potential target of DISC1. METHODS DISC1 was knocked down with a small inference RNA. NMDAR-mediated currents were recorded and NMDAR expression was measured. RESULTS We found that cellular knockdown of DISC1 significantly increased NMDAR currents in cortical cultures, which were accompanied by an increase in the expression of NMDAR subunit, GluN2A. NMDAR-mediated synaptic response in prefrontal cortical pyramidal neurons was also increased by DISC1 knockdown in vivo. The effect of DISC1 knockdown on NMDAR currents in cortical cultures was blocked by protein kinase A (PKA) inhibitor, occluded by PKA activator, and prevented by phosphodiesterase 4 inhibitor. Knockdown of DISC1 caused a significant increase of cyclic adenosine monophosphate response element-binding protein (CREB) activity. Inhibiting CREB prevented the DISC1 deficiency-induced increase of NMDAR currents and GluN2A clusters. CONCLUSIONS Our results suggest that DISC1 exerts an important impact on NMDAR expression and function through a phosphodiesterase 4/PKA/CREB-dependent mechanism, which provides a potential molecular basis for the role of DISC1 in influencing NMDAR-dependent cognitive and emotional processes.
Collapse
|
44
|
Orozco IJ, Koppensteiner P, Ninan I, Arancio O. The schizophrenia susceptibility gene DTNBP1 modulates AMPAR synaptic transmission and plasticity in the hippocampus of juvenile DBA/2J mice. Mol Cell Neurosci 2013; 58:76-84. [PMID: 24321452 DOI: 10.1016/j.mcn.2013.12.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2013] [Revised: 10/31/2013] [Accepted: 12/02/2013] [Indexed: 10/25/2022] Open
Abstract
The dystrobrevin binding protein (DTNBP) 1 gene has emerged over the last decade as a potential susceptibility locus for schizophrenia. While no causative mutations have been found, reduced expression of the encoded protein, dysbindin, was reported in patients. Dysbindin likely plays a role in the neuronal trafficking of proteins including receptors. One important pathway suspected to be affected in schizophrenia is the fast excitatory glutamatergic transmission mediated by AMPA receptors. Here, we investigated excitatory synaptic transmission and plasticity in hippocampal neurons from dysbindin-deficient sandy mice bred on the DBA/2J strain. In cultured neurons an enhancement of AMPAR responses was observed. The enhancement of AMPAR-mediated transmission was confirmed in hippocampal CA3-CA1 synapses, and was not associated with changes in the expression of GluA1-4 subunits or an increase in GluR2-lacking receptor complexes. Lastly, an enhancement in LTP was also found in these mice. These data provide compelling evidence that dysbindin, a widely suspected susceptibility protein in schizophrenia, is important for AMPAR-mediated synaptic transmission and plasticity in the developing hippocampus.
Collapse
Affiliation(s)
- Ian J Orozco
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA; Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA.
| | - Peter Koppensteiner
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA; Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Ipe Ninan
- Department of Psychiatry, New York University Langone Medical Center, New York, NY, USA
| | - Ottavio Arancio
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA; Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA.
| |
Collapse
|
45
|
Ottis P, Topic B, Loos M, Li KW, de Souza A, Schulz D, Smit AB, Huston JP, Korth C. Aging-induced proteostatic changes in the rat hippocampus identify ARP3, NEB2 and BRAG2 as a molecular circuitry for cognitive impairment. PLoS One 2013; 8:e75112. [PMID: 24069387 PMCID: PMC3777897 DOI: 10.1371/journal.pone.0075112] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Accepted: 08/10/2013] [Indexed: 11/28/2022] Open
Abstract
Disturbed proteostasis as a particular phenotype of the aging organism has been advanced in C. elegans experiments and is also conceived to underlie neurodegenerative diseases in humans. Here, we investigated whether particular changes in non-disease related proteostasis can be identified in the aged mammalian brain, and whether a particular signature of aberrant proteostasis is related to behavioral performance of learning and memory. Young (adult, n = 30) and aged (2 years, n = 50) Wistar rats were tested in the Morris Water Maze (MWM) to distinguish superior and inferior performers. For both young and old rats, the best and worst performers in the MWM were selected and the insoluble proteome, termed aggregome, was purified from the hippocampus as evidence for aberrant proteostasis. Quantitative proteomics (iTRAQ) was performed. The aged inferior performers were considered as a model for spontaneous, age-associated cognitive impairment. Whereas variability of the insoluble proteome increased with age, absolute changes in the levels of insoluble proteins were small compared to the findings in the whole C. elegans insoluble proteome. However, we identified proteins with aberrant proteostasis in aging. For the cognitively impaired rats, we identified a changed molecular circuitry of proteins selectively involved in F-actin remodeling, synapse building and long-term depression: actin related protein 3 (ARP3), neurabin II (NEB2) and IQ motif and SEC7 domain-containing protein 1 (BRAG2). We demonstrate that aberrant proteostasis is a specific phenotype of brain aging in mammals. We identify a distinct molecular circuitry where changes in proteostasis are characteristic for poor learning and memory performance in the wild type, aged rat. Our findings 1. establish the search for aberrant proteostasis as a successful strategy to identify neuronal dysfunction in deficient cognitive behavior, 2. reveal a previously unknown functional network of proteins (ARP3, NEB2, BRAG2) involved in age-associated cognitive dysfunction.
Collapse
Affiliation(s)
- Philipp Ottis
- Department of Neuropathology, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Bianca Topic
- Center for Behavioral Neuroscience, Department Experimental Psychology, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Maarten Loos
- Department of Molecular and Cellular Neurobiology, Faculty of Earth and Life Sciences, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
- Synaptologics B.V., Amsterdam, The Netherlands
| | - Ka Wan Li
- Department of Molecular and Cellular Neurobiology, Faculty of Earth and Life Sciences, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
| | - Angelica de Souza
- Center for Behavioral Neuroscience, Department Experimental Psychology, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Daniela Schulz
- Center for Behavioral Neuroscience, Department Experimental Psychology, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - August B. Smit
- Department of Molecular and Cellular Neurobiology, Faculty of Earth and Life Sciences, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
| | - Joseph P. Huston
- Center for Behavioral Neuroscience, Department Experimental Psychology, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Carsten Korth
- Department of Neuropathology, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
- * E-mail:
| |
Collapse
|
46
|
Kelsom C, Lu W. Development and specification of GABAergic cortical interneurons. Cell Biosci 2013; 3:19. [PMID: 23618463 PMCID: PMC3668182 DOI: 10.1186/2045-3701-3-19] [Citation(s) in RCA: 124] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Accepted: 03/28/2013] [Indexed: 12/21/2022] Open
Abstract
GABAergic interneurons are inhibitory neurons of the nervous system that play a vital role in neural circuitry and activity. They are so named due to their release of the neurotransmitter gamma-aminobutyric acid (GABA), and occupy different areas of the brain. This review will focus primarily on GABAergic interneurons of the mammalian cerebral cortex from a developmental standpoint. There is a diverse amount of cortical interneuronal subtypes that may be categorized by a number of characteristics; this review will classify them largely by the protein markers they express. The developmental origins of GABAergic interneurons will be discussed, as well as factors that influence the complex migration routes that these interneurons must take in order to ultimately localize in the cerebral cortex where they will integrate with the neural circuitry set in place. This review will also place an emphasis on the transcriptional network of genes that play a role in the specification and maintenance of GABAergic interneuron fate. Gaining an understanding of the different aspects of cortical interneuron development and specification, especially in humans, has many useful clinical applications that may serve to treat various neurological disorders linked to alterations in interneuron populations.
Collapse
Affiliation(s)
- Corey Kelsom
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Department of Biochemistry and Molecular Biology, University of Southern California, 1425 San Pablo Street, Los Angeles, CA 90033, USA.
| | | |
Collapse
|
47
|
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
|
48
|
Seshadri S, Zeledon M, Sawa A. Synapse-specific contributions in the cortical pathology of schizophrenia. Neurobiol Dis 2013; 53:26-35. [PMID: 23336981 DOI: 10.1016/j.nbd.2013.01.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Revised: 01/06/2013] [Accepted: 01/10/2013] [Indexed: 12/22/2022] Open
Abstract
Schizophrenia (SZ) is often described as a disease of neuronal connectivity. Cognitive processes such as working memory, which are particularly dependent on the proper functioning of complex cortical circuitry, are disturbed in the disease. Reciprocal connections between pyramidal neurons and interneurons, as well as dopaminergic innervations, form the basis for higher cognition in the cortex. Nonetheless, only a few review articles are available which address how each synapse operates, and is possibly disturbed in SZ, at least in part by the mechanisms involving genetic susceptibility factors for SZ. In this review, we provide an overview of cortical glutamatergic, GABAergic, and dopaminergic circuitry, review SZ-associated deficits at each of these synapses, and discuss how genetic factors for SZ may contribute to SZ-related phenotype deficits in a synapse-specific manner. Pinpointing the spatially and temporally distinct sites of action of putative SZ susceptibility factors may help us better understand the pathological mechanisms of SZ, especially those associated with synaptic functioning and neuronal connectivity.
Collapse
Affiliation(s)
- Saurav Seshadri
- Department of Psychiatry, Johns Hopkins University, Baltimore, MD 21287, USA
| | | | | |
Collapse
|
49
|
Coughlin JM, Ishizuka K, Kano SI, Edwards JA, Seifuddin FT, Shimano MA, Daley EL, Zandi PP, Leweke FM, Cascella NG, Pomper MG, Yolken RH, Sawa A. Marked reduction of soluble superoxide dismutase-1 (SOD1) in cerebrospinal fluid of patients with recent-onset schizophrenia. Mol Psychiatry 2013; 18:10-1. [PMID: 22349781 PMCID: PMC4113962 DOI: 10.1038/mp.2012.6] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- JM Coughlin
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - K Ishizuka
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - SI Kano
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - JA Edwards
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - FT Seifuddin
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - MA Shimano
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - EL Daley
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - PP Zandi
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA,Bloomberg School of Public Health, Baltimore, MD, USA
| | - FM Leweke
- Central Institute of Mental Health, University of Heidelberg, Mannheim, Germany
| | - NG Cascella
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - MG Pomper
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - RH Yolken
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA,The Stanley Medical Research Institute, Chevy Chase, MD, USA
| | - A Sawa
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
50
|
Bader V, Tomppo L, Trossbach SV, Bradshaw NJ, Prikulis I, Leliveld SR, Lin CY, Ishizuka K, Sawa A, Ramos A, Rosa I, García Á, Requena JR, Hipolito M, Rai N, Nwulia E, Henning U, Ferrea S, Luckhaus C, Ekelund J, Veijola J, Järvelin MR, Hennah W, Korth C. Proteomic, genomic and translational approaches identify CRMP1 for a role in schizophrenia and its underlying traits. Hum Mol Genet 2012; 21:4406-18. [PMID: 22798627 PMCID: PMC3529585 DOI: 10.1093/hmg/dds273] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Revised: 07/05/2012] [Accepted: 07/07/2012] [Indexed: 01/06/2023] Open
Abstract
Schizophrenia is a chronic illness of heterogenous biological origin. We hypothesized that, similar to chronic progressive brain conditions, persistent functional disturbances of neurons would result in disturbed proteostasis in the brains of schizophrenia patients, leading to increased abundance of specific misfolded, insoluble proteins. Identification of such proteins would facilitate the elucidation of molecular processes underlying these devastating conditions. We therefore generated antibodies against pooled insoluble proteome of post-mortem brains from schizophrenia patients in order to identify unique, disease-specific epitopes. We successfully identified such an epitope to be present on collapsin-response mediator protein 1 (CRMP1) in biochemically purified, insoluble brain fractions. A genetic association analysis for the CRMP1 gene in a large Finnish population cohort (n = 4651) corroborated the association of physical and social anhedonia with the CRMP1 locus in a DISC1 (Disrupted-in-schizophrenia 1)-dependent manner. Physical and social anhedonia are heritable traits, present as chronic, negative symptoms of schizophrenia and severe major depression, thus constituting serious vulnerability factors for mental disease. Strikingly, lymphoblastoid cell lines derived from schizophrenia patients mirrored aberrant CRMP1 immunoreactivity by showing an increase of CRMP1 expression, suggesting its potential role as a blood-based diagnostic marker. CRMP1 is a novel candidate protein for schizophrenia traits at the intersection of the reelin and DISC1 pathways that directly and functionally interacts with DISC1. We demonstrate the impact of an interdisciplinary approach where the identification of a disease-associated epitope in post-mortem brains, powered by a genetic association study, is rapidly translated into a potential blood-based diagnostic marker.
Collapse
Affiliation(s)
- Verian Bader
- Department of Neuropathology, Heinrich Heine University Düsseldorf, Germany
| | - Liisa Tomppo
- Institute for Molecular Medicine Finland FIMM, University of Helsinki, Helsinki, Finland
| | | | | | - Ingrid Prikulis
- Department of Neuropathology, Heinrich Heine University Düsseldorf, Germany
| | - S. Rutger Leliveld
- Department of Neuropathology, Heinrich Heine University Düsseldorf, Germany
| | - Chi-Ying Lin
- Department of Psychiatry, Johns Hopkins University Medical School, Baltimore, MD, USA
| | - Koko Ishizuka
- Department of Psychiatry, Johns Hopkins University Medical School, Baltimore, MD, USA
| | - Akira Sawa
- Department of Psychiatry, Johns Hopkins University Medical School, Baltimore, MD, USA
| | - Adriana Ramos
- Department of Medicine and
- CIMUS Biomedical Research Institute, University of Santiago de Compostela-IDIS, Santiago de Compostela, Spain
| | - Isaac Rosa
- Department of Pharmacology, University of Santiago de Compostela, Santiago de Compostela, Spain
- CIMUS Biomedical Research Institute, University of Santiago de Compostela-IDIS, Santiago de Compostela, Spain
| | - Ángel García
- Department of Pharmacology, University of Santiago de Compostela, Santiago de Compostela, Spain
- CIMUS Biomedical Research Institute, University of Santiago de Compostela-IDIS, Santiago de Compostela, Spain
| | - Jesús R. Requena
- Department of Medicine and
- CIMUS Biomedical Research Institute, University of Santiago de Compostela-IDIS, Santiago de Compostela, Spain
| | - Maria Hipolito
- Department of Psychiatry, Howard University, Washington, DC, USA
| | - Narayan Rai
- Department of Psychiatry, Howard University, Washington, DC, USA
| | - Evaristus Nwulia
- Department of Psychiatry, Howard University, Washington, DC, USA
| | - Uwe Henning
- Department of Psychiatry, Heinrich Heine University Düsseldorf, Germany
| | - Stefano Ferrea
- Department of Psychiatry, Heinrich Heine University Düsseldorf, Germany
| | | | - Jesper Ekelund
- National Institute for Health and Welfare, Helsinki, Finland
- Department of Psychiatry, University of Helsinki, Helsinki, Finland
- Vaasa Hospital District, Vaasa, Finland
| | - Juha Veijola
- Department of Psychiatry, University of Oulu Central Hospital, Oulu, Finland
| | - Marjo-Riitta Järvelin
- School of Public Health and MRC Health Protection Agency, Centre for Environment and Health, Imperial College London, London, UK
- Institute of Health Sciences and Biocenter Oulu, University of Oulu, Oulu, Finland and
| | - William Hennah
- Institute for Molecular Medicine Finland FIMM, University of Helsinki, Helsinki, Finland
- National Institute for Health and Welfare, Helsinki, Finland
- Medical Genetics Section, University of Edinburgh, Edinburgh, UK
| | - Carsten Korth
- Department of Neuropathology, Heinrich Heine University Düsseldorf, Germany
| |
Collapse
|