51
|
Tibbo AJ, Baillie GS. Phosphodiesterase 4B: Master Regulator of Brain Signaling. Cells 2020; 9:cells9051254. [PMID: 32438615 PMCID: PMC7291338 DOI: 10.3390/cells9051254] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 05/13/2020] [Accepted: 05/14/2020] [Indexed: 12/25/2022] Open
Abstract
Phosphodiesterases (PDEs) are the only superfamily of enzymes that have the ability to break down cyclic nucleotides and, as such, they have a pivotal role in neurological disease and brain development. PDEs have a modular structure that allows targeting of individual isoforms to discrete brain locations and it is often the location of a PDE that shapes its cellular function. Many of the eleven different families of PDEs have been associated with specific diseases. However, we evaluate the evidence, which suggests the activity from a sub-family of the PDE4 family, namely PDE4B, underpins a range of important functions in the brain that positions the PDE4B enzymes as a therapeutic target for a diverse collection of indications, such as, schizophrenia, neuroinflammation, and cognitive function.
Collapse
|
52
|
Vázquez-Bourgon J, Ayesa-Arriola R, Fatjó-Vilas M, Roiz-Santiañez R, Fañanás L, Crespo-Facorro B. Effect of DISC1 Polymorphisms on the Long-term Course of Neurocognitive Deficits in Non-affective Psychosis. Eur Psychiatry 2020; 30:861-7. [DOI: 10.1016/j.eurpsy.2015.07.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 07/10/2015] [Accepted: 07/13/2015] [Indexed: 12/12/2022] Open
Abstract
AbstractNeurocognitive deficits are core symptoms of schizophrenia that determine a poorer outcome. High variability in the progression of neuropsychological deficits in schizophrenia has been described. It is still unknown whether genetic variations can affect the course of cognitive deficits. Variations in the Disrupted in Schizophrenia 1 (DISC1) gene have previously been associated with neurocognitive deficits. This study investigated the association between 3 DISC1 polymorphisms (rs6675281 (Leu607Phe), rs1000731, and rs821616 (Ser704Cys)) and long-term (3 years) cognitive performance. One-hundred-thirty-three Caucasian drug-naive patients experiencing a first episode of non-affective psychosis were genotyped. Cognitive function was assessed at baseline and after 3 years of initiating treatment. Other clinical and socio-demographic variables were recorded to eliminate potential confounding effects. Patients carrying the A allele of rs1000731 exhibited a significant improvement in Working Memory and Attention domains, and the homozygosity of the A allele of rs821616 showed a significant improvement in Motor Dexterity performance over 3 years of follow-up. In conclusion, DISC1 gene variations may affect the course of cognitive deficits found in patients suffering from the first episode of non-affective psychosis.
Collapse
|
53
|
Evidence of association of the DISC1 interactome gene set with schizophrenia from GWAS. Prog Neuropsychopharmacol Biol Psychiatry 2019; 95:109729. [PMID: 31398428 DOI: 10.1016/j.pnpbp.2019.109729] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 07/31/2019] [Accepted: 08/03/2019] [Indexed: 12/31/2022]
Abstract
DISC1 was discovered as a gene disrupted by a balanced translocation in a large pedigree that segregated with major mental disorders, including schizophrenia. Further attempts to find genetic association with schizophrenia were inconclusive. Most of the biology of DISC1 was inferred from the functionality of its protein partners. Recently, a gene set constituted by DISC1 and several of its partners has been associated with cognitive performance during development, a well-known schizophrenia endophenotype, by means of burden test of rare disruptive variants. Here, we performed a gene set analysis using common variants from the largest schizophrenia genome-wide association study of the Psychiatric Genomics Consortium to test if this gene set is associated with schizophrenia. The main test was based on the MAGMA software. Several additional tests were performed to analyze the robustness of the main findings. The DISC1 interactome gene set was associated with schizophrenia (P = .0056), confirmed by an additional method (INRICH). This association was robust to removal of the major histocompatibility complex region, different definitions of gene boundaries, or different statistical gene models. Conditional analysis revealed that the association was not solely explained by higher expression in brain. Three genes from the gene set, CLIC1, DST, and PDE4B, were associated with schizophrenia at the gene level. Consideration of other DISC1 interactome gene sets revealed the importance of gene set definition. Therefore, we present the first evidence from genome-wide association studies of the role of DISC1 and interacting partners in schizophrenia susceptibility, reconciling genetic and molecular biology data.
Collapse
|
54
|
Translating preclinical findings in clinically relevant new antipsychotic targets: focus on the glutamatergic postsynaptic density. Implications for treatment resistant schizophrenia. Neurosci Biobehav Rev 2019; 107:795-827. [DOI: 10.1016/j.neubiorev.2019.08.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 07/20/2019] [Accepted: 08/22/2019] [Indexed: 02/07/2023]
|
55
|
Baskaran R, Lai C, Li W, Tuan L, Wang C, Lee LJ, Liu C, Hwu H, Lee L. Characterization of striatal phenotypes in heterozygous
Disc1
mutant mice, a model of haploinsufficiency. J Comp Neurol 2019; 528:1157-1172. [DOI: 10.1002/cne.24813] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 08/26/2019] [Accepted: 11/08/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Rathinasamy Baskaran
- Graduate Institute of Anatomy and Cell BiologyNational Taiwan University Taipei Taiwan ROC
| | - Chuan‐Ching Lai
- Graduate Institute of Anatomy and Cell BiologyNational Taiwan University Taipei Taiwan ROC
| | - Wai‐Yu Li
- Graduate Institute of Anatomy and Cell BiologyNational Taiwan University Taipei Taiwan ROC
| | - Li‐Heng Tuan
- Graduate Institute of Anatomy and Cell BiologyNational Taiwan University Taipei Taiwan ROC
| | - Chia‐Chuan Wang
- School of MedicineFu Jen Catholic University New Taipei Taiwan ROC
| | - Lukas J.‐H. Lee
- Division of Environmental Health and Occupational MedicineNational Health Research Institutes Miaoli Taiwan ROC
| | - Chih‐Min Liu
- Department of PsychiatryNational Taiwan University Hospital and National Taiwan University College of Medicine Taipei Taiwan ROC
- Neurobiology and Cognitive Science CenterNational Taiwan University Taipei Taiwan ROC
| | - Hai‐Gwo Hwu
- Department of PsychiatryNational Taiwan University Hospital and National Taiwan University College of Medicine Taipei Taiwan ROC
- Neurobiology and Cognitive Science CenterNational Taiwan University Taipei Taiwan ROC
- Institute of Brain and Mind SciencesNational Taiwan University Taipei Taiwan ROC
| | - Li‐Jen Lee
- Graduate Institute of Anatomy and Cell BiologyNational Taiwan University Taipei Taiwan ROC
- Department of PsychiatryNational Taiwan University Hospital and National Taiwan University College of Medicine Taipei Taiwan ROC
- Neurobiology and Cognitive Science CenterNational Taiwan University Taipei Taiwan ROC
- Institute of Brain and Mind SciencesNational Taiwan University Taipei Taiwan ROC
| |
Collapse
|
56
|
Trantham-Davidson H, Lavin A. Loss of dysbindin-1 affects GABAergic transmission in the PFC. Psychopharmacology (Berl) 2019; 236:3291-3300. [PMID: 31201475 PMCID: PMC6832803 DOI: 10.1007/s00213-019-05285-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 05/24/2019] [Indexed: 10/26/2022]
Abstract
It has been shown that dystrobrevin-binding protein 1 gene that encodes the protein dysbindin-1 is associated with risk for cognitive deficits, and studies have shown decreases in glutamate and correlated decreases in dysbindin-1 protein in the prefrontal cortex (PFC) and hippocampus of post-mortem tissue from schizophrenia patients. The PFC and the hippocampus have been shown to play a fundamental role in cognition, and studies in dysbindin-1 null mice have shown alterations in NMDAR located in pyramidal neurons as well as perturbation in LTP and cognitive deficits. The balance between excitatory and inhibitory transmission is crucial for normal cognitive functions; however, there is a dearth of information regarding the effects of loss of dysbindin-1 in GABAergic transmission. Using in vitro whole-cell clamp recordings, Western blots, and immunohistochemistry, we report here that dysbindin-1-deficient mice exhibit a significant decrease in the frequency of sIPSCs and in the amplitude of mIPSCs and significant decreases in PV staining and protein level. These results suggest that loss of dysbindin-1 affects GABAergic transmission at pre- and postsynaptic level and decreases parvalbumin markers.
Collapse
Affiliation(s)
| | - A Lavin
- Department of Neuroscience, MUSC, Charleston, SC, 29425, USA.
| |
Collapse
|
57
|
Roche J, Potoyan DA. Disorder Mediated Oligomerization of DISC1 Proteins Revealed by Coarse-Grained Molecular Dynamics Simulations. J Phys Chem B 2019; 123:9567-9575. [PMID: 31614085 DOI: 10.1021/acs.jpcb.9b07467] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Disrupted-in-schizophrenia-1 (DISC1) is a scaffold protein of significant importance for neuro-development and a prominent candidate protein in the etiology of mental disorders. In this work, we investigate the role of conformational heterogeneity and local structural disorder in the oligomerization pathway of the full-length DISC1 and of two truncation variants. Through extensive coarse-grained molecular dynamics simulations with a predictive energy landscape-based model, we shed light on the interplay of local and global disorder which lead to different oligomerization pathways. We found that both global conformational heterogeneity and local structural disorder play an important role in shaping distinct oligomerization trends of DISC1. This study also sheds light on the differences in oligomerization pathways of the full-length protein compared to the truncated variants produced by a chromosomal translocation associated with schizophrenia. We report that oligomerization of full-length DISC1 sequence works in a nonadditive manner with respect to truncated fragments that do not mirror the conformational landscape or binding affinities of the full-length unit.
Collapse
Affiliation(s)
- Julien Roche
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology , Iowa State University , Ames , Iowa 50011 , United States
| | - Davit A Potoyan
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology , Iowa State University , Ames , Iowa 50011 , United States.,Department of Chemistry , Iowa State University , Ames , Iowa 50011 , United States.,Bioinformatics and Computational Biology Program , Iowa State University , Ames , Iowa 50011 , United States
| |
Collapse
|
58
|
Barnett BR, Anderson JM, Torres-Velázquez M, Yi SY, Rowley PA, Yu JPJ. Exercise ameliorates deficits in neural microstructure in a Disc1 model of psychiatric illness. Magn Reson Imaging 2019; 61:90-96. [PMID: 31103832 PMCID: PMC6663582 DOI: 10.1016/j.mri.2019.05.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 05/14/2019] [Accepted: 05/15/2019] [Indexed: 01/06/2023]
Abstract
Recent studies have investigated the effectiveness of aerobic exercise to improve physical and mental health outcomes in schizophrenia; however, few have explicitly explored the impact of aerobic exercise on neural microstructure, which is hypothesized to mediate the behavioral changes observed. Neural microstructure is influenced by numerous genetic factors including DISC1, which is a major molecular scaffold protein that interacts with partners like GSK3β, NDEL1, and PDE4. DISC1 has been shown to play a role in neurogenesis, neuronal migration, neuronal maturation, and synaptic signaling. As with other genetic variants that present an increased risk for disease, mutations of the DISC1 gene have been implicated in the molecular intersection of schizophrenia and numerous other major psychiatric illnesses. This study investigated whether short-term exercise recovers deficits in neural microstructure in a novel genetic Disc1 svΔ2 rat model. Disc1 svΔ2 animals and age- and sex-matched controls were subjected to a treadmill exercise protocol. Subsequent ex-vivo diffusion tensor imaging (DTI) and neurite orientation dispersion and density imaging (NODDI) compared neural microstructure in regions of interest (ROI) between sedentary and exercise wild-type animals and between sedentary and exercise Disc1 svΔ2 animals. Short-term exercise uncovered no significant differences in neural microstructure between sedentary and exercise control animals but did lead to significant differences between sedentary and exercise Disc1 svΔ2 animals in neocortex, basal ganglia, corpus callosum, and external capsule, suggesting a positive benefit derived from a short-term exercise regimen. Our findings suggest that Disc1 svΔ2 animals are more sensitive to the effects of short-term exercise and highlight the ameliorating potential of positive treatment interventions such as exercise on neural microstructure in genetic backgrounds of psychiatric disease susceptibility.
Collapse
Affiliation(s)
- Brian R Barnett
- Neuroscience Training Program, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Jacqueline M Anderson
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Maribel Torres-Velázquez
- Department of Biomedical Engineering, College of Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Sue Y Yi
- Neuroscience Training Program, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Paul A Rowley
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - John-Paul J Yu
- Neuroscience Training Program, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; Department of Biomedical Engineering, College of Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA; Department of Psychiatry, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA.
| |
Collapse
|
59
|
Monte GG, Nani JV, de Almeida Campos MR, Dal Mas C, Marins LAN, Martins LG, Tasic L, Mori MA, Hayashi MAF. Impact of nuclear distribution element genes in the typical and atypical antipsychotics effects on nematode Caenorhabditis elegans: Putative animal model for studying the pathways correlated to schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry 2019; 92:19-30. [PMID: 30578843 DOI: 10.1016/j.pnpbp.2018.12.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 12/17/2018] [Accepted: 12/19/2018] [Indexed: 12/18/2022]
Abstract
The nuclear distribution element genes are conserved from fungus to humans. The nematode Caenorhabditis elegans expresses two isoforms of nuclear distribution element genes, namely nud-1 and nud-2. While nud-1 was functionally demonstrated to be the worm nudC ortholog, bioinformatic analysis revealed that the nud-2 gene encodes the worm ortholog of the mammalian NDE1 (Nuclear Distribution Element 1 or NudE) and NDEL1 (NDE-Like 1 or NudEL) genes, which share overlapping roles in brain development in mammals and also mediate the axon guidance in mammalian and C. elegans neurons. A significantly higher NDEL1 enzyme activity was shown in treatment non-resistant compared to treatment resistant SCZ patients, who essentially present response to the therapy with atypical clozapine but not with typical antipsychotics. Using C. elegans as a model, we tested the consequence of nud genes suppression in the effects of typical and atypical antipsychotics. To assess the role of nud genes and antipsychotic drugs over C. elegans behavior, we measured body bend frequency, egg laying and pharyngeal pumping, which traits are controlled by specific neurons and neurotransmitters known to be involved in SCZ, as dopamine and serotonin. Evaluation of metabolic and behavioral response to the pharmacotherapy with these antipsychotics demonstrates an important unbalance in serotonin pathway in both nud-1 and nud-2 knockout worms, with more significant effects for nud-2 knockout. The present data also show an interesting trend of mutant knockout worm strains to present a metabolic profile closer to that observed for the wild-type animals after the treatment with the typical antipsychotic haloperidol, but which was not observed for the treatment with the atypical antipsychotic clozapine. Paradoxically, behavioral assays showed more evident effects for clozapine than for haloperidol, which is in line with previous studies with rodent animal models and clinical evaluations with SCZ patients. In addition, the validity and reliability of using this experimental animal model to further explore the convergence between the dopamine/serotonin pathways and neurodevelopmental processes was demonstrated here, and the potential usefulness of this model for evaluating the metabolic consequences of treatments with antipsychotics is also suggested.
Collapse
Affiliation(s)
- Gabriela Guilherme Monte
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP) São Paulo, Brazil
| | - João V Nani
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP) São Paulo, Brazil
| | | | - Caroline Dal Mas
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP) São Paulo, Brazil
| | - Lucas Augusto Negri Marins
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP) São Paulo, Brazil
| | - Lucas Gelain Martins
- Chemical Biology Laboratory, Department of Organic Chemistry, Institute of Chemistry, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil
| | - Ljubica Tasic
- Chemical Biology Laboratory, Department of Organic Chemistry, Institute of Chemistry, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil
| | - Marcelo A Mori
- Departament of Biochemistry and Tissue Biology, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil
| | - Mirian A F Hayashi
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP) São Paulo, Brazil.
| |
Collapse
|
60
|
Ndel1 oligopeptidase activity as a potential biomarker of early stages of schizophrenia. Schizophr Res 2019; 208:202-208. [PMID: 30857875 DOI: 10.1016/j.schres.2019.02.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 02/22/2019] [Accepted: 02/25/2019] [Indexed: 02/01/2023]
Abstract
Our previous studies showed reduced Ndel1 enzyme activity in patients with chronic schizophrenia (SCZ), and only a subtle NDEL1 mRNA increases in antipsychotic-naïve first-episode psychosis (FEP) individuals compared to matched healthy controls (HC). Aiming to refine the evaluation of Ndel1 enzyme activity in early stages of psychosis, we compared 3 groups composed by (1) subjects at ultra-high-risk (UHR) for psychosis, (2) a cohort comprising antipsychotic-naïve FEP individuals (assessed in three moments, at baseline (FEP-0), and after 2 months (FEP-2 M) and one year (FEP-1Y) of treatment with risperidone), and (3) a HC group. There was no significant difference in Ndel1 enzyme activity between UHR and HC, but this activity was significantly lower in FEP compared to HC. Conversely, Ndel1 activity in HC groups was higher than in FEP even before (FEP-0) or after the treatment with risperidone (FEP-2 M and FEP-1Y), and with progressive decrease of Ndel1 activity and significant improvement of symptoms observed after this treatment. In addition, a positive correlation was observed for Ndel1 activity with clinical symptoms as assessed by PANSS, while a negative correlation was seen for GAF scores. Our results suggest that reductions in Ndel1 activity in FEP may be possibly related to responses to the illness, rather than to the pharmacological effects of antipsychotics, which might be acting essentially in the symptoms suppression. This hypothesis might be further evaluated in prospective long-term follow-up studies with a larger sample cohort.
Collapse
|
61
|
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
|
62
|
Oulas A, Minadakis G, Zachariou M, Sokratous K, Bourdakou MM, Spyrou GM. Systems Bioinformatics: increasing precision of computational diagnostics and therapeutics through network-based approaches. Brief Bioinform 2019; 20:806-824. [PMID: 29186305 PMCID: PMC6585387 DOI: 10.1093/bib/bbx151] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 10/17/2017] [Indexed: 02/01/2023] Open
Abstract
Systems Bioinformatics is a relatively new approach, which lies in the intersection of systems biology and classical bioinformatics. It focuses on integrating information across different levels using a bottom-up approach as in systems biology with a data-driven top-down approach as in bioinformatics. The advent of omics technologies has provided the stepping-stone for the emergence of Systems Bioinformatics. These technologies provide a spectrum of information ranging from genomics, transcriptomics and proteomics to epigenomics, pharmacogenomics, metagenomics and metabolomics. Systems Bioinformatics is the framework in which systems approaches are applied to such data, setting the level of resolution as well as the boundary of the system of interest and studying the emerging properties of the system as a whole rather than the sum of the properties derived from the system's individual components. A key approach in Systems Bioinformatics is the construction of multiple networks representing each level of the omics spectrum and their integration in a layered network that exchanges information within and between layers. Here, we provide evidence on how Systems Bioinformatics enhances computational therapeutics and diagnostics, hence paving the way to precision medicine. The aim of this review is to familiarize the reader with the emerging field of Systems Bioinformatics and to provide a comprehensive overview of its current state-of-the-art methods and technologies. Moreover, we provide examples of success stories and case studies that utilize such methods and tools to significantly advance research in the fields of systems biology and systems medicine.
Collapse
Affiliation(s)
- Anastasis Oulas
- Bioinformatics European Research Area Chair, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - George Minadakis
- Bioinformatics European Research Area Chair, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Margarita Zachariou
- Bioinformatics European Research Area Chair, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Kleitos Sokratous
- Bioinformatics European Research Area Chair, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Marilena M Bourdakou
- Bioinformatics European Research Area Chair, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - George M Spyrou
- Bioinformatics European Research Area Chair, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| |
Collapse
|
63
|
Read J, Collie IT, Nguyen-McCarty M, Lucaj C, Robinson J, Conway L, Mukherjee J, McCall E, Donohoe G, Flavell E, Peciak K, Warwicker J, Dix C, Van den Hoven BG, Madin A, Brown DG, Moss S, Haggarty SJ, Brandon NJ, Bürli RW. Tool inhibitors and assays to interrogate the biology of the TRAF2 and NCK interacting kinase. Bioorg Med Chem Lett 2019; 29:1962-1967. [PMID: 31153805 DOI: 10.1016/j.bmcl.2019.05.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 05/16/2019] [Accepted: 05/17/2019] [Indexed: 01/21/2023]
Abstract
The TRAF2 and NCK interacting kinase (TNIK) has been proposed to play a role in cytoskeletal organization and synaptic plasticity and has been linked, among others, to neurological disorders. However, target validation efforts for TNIK have been hampered by the limited kinase selectivity of small molecule probes and possible functional compensation in mouse models. Both issues are at least in part due to its close homology to the kinases MINK1 (or MAP4K6) and MAP4K4 (or HGK). As part of our interest in validating TNIK as a therapeutic target for neurological diseases, we set up a panel of biochemical and cellular assays, which are described herein. We then examined the activity of known amino-pyridine-based TNIK inhibitors (1, 3) and prepared structurally very close analogs that lack the ability to inhibit the target. We also developed a structurally orthogonal, naphthyridine-based TNIK inhibitor (9) and an inactive control molecule of the same chemical series. These validated small-molecule probes will enable dissection of the function of TNIK family in the context of human disease biology.
Collapse
Affiliation(s)
- Jon Read
- Discovery Sciences, IMED Biotech Unit, AstraZeneca, Cambridge CB4 0WG, UK
| | - Iain T Collie
- Discovery Sciences, IMED Biotech Unit, AstraZeneca, Cambridge CB4 0WG, UK
| | - Michelle Nguyen-McCarty
- Departments of Neurology and Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Christopher Lucaj
- AstraZeneca Tufts Laboratory for Basic and Translational Neuroscience, Boston, MA 02111, USA
| | - James Robinson
- Discovery Sciences, IMED Biotech Unit, AstraZeneca, Cambridge CB4 0WG, UK
| | - Leslie Conway
- AstraZeneca Tufts Laboratory for Basic and Translational Neuroscience, Boston, MA 02111, USA
| | - Jayanta Mukherjee
- AstraZeneca Tufts Laboratory for Basic and Translational Neuroscience, Boston, MA 02111, USA
| | - Eileen McCall
- Discovery Sciences, IMED Biotech Unit, AstraZeneca, Cambridge CB4 0WG, UK
| | - Gerard Donohoe
- Discovery Sciences, IMED Biotech Unit, AstraZeneca, Cambridge CB4 0WG, UK
| | - Elizabeth Flavell
- Discovery Sciences, IMED Biotech Unit, AstraZeneca, Alderley Park, SK10 4TG, UK
| | - Karolina Peciak
- Discovery Sciences, IMED Biotech Unit, AstraZeneca, Alderley Park, SK10 4TG, UK
| | - Juli Warwicker
- Discovery Sciences, IMED Biotech Unit, AstraZeneca, Alderley Park, SK10 4TG, UK
| | - Carly Dix
- Discovery Sciences, IMED Biotech Unit, AstraZeneca, Cambridge CB4 0WG, UK
| | | | - Andrew Madin
- Discovery Sciences, IMED Biotech Unit, AstraZeneca, Cambridge CB4 0WG, UK
| | - Dean G Brown
- Discovery Sciences, IMED Biotech Unit, AstraZeneca Boston, Waltham, MA 02451, USA
| | - Stephen Moss
- AstraZeneca Tufts Laboratory for Basic and Translational Neuroscience, Boston, MA 02111, USA
| | - Stephen J Haggarty
- Departments of Neurology and Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Nicholas J Brandon
- Neuroscience, IMED Biotech Unit, AstraZeneca Boston, Waltham, MA 02451, USA.
| | - Roland W Bürli
- Neuroscience, IMED Biotech Unit, AstraZeneca, Cambridge CB21 6GH, UK.
| |
Collapse
|
64
|
Han MR, Han KM, Kim A, Kang W, Kang Y, Kang J, Won E, Tae WS, Cho Y, Ham BJ. Whole-exome sequencing identifies variants associated with structural MRI markers in patients with bipolar disorders. J Affect Disord 2019; 249:159-168. [PMID: 30772743 DOI: 10.1016/j.jad.2019.02.028] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 01/29/2019] [Accepted: 02/10/2019] [Indexed: 12/23/2022]
Abstract
BACKGROUND Bipolar disorder (BD) is one of the most heritable psychiatric disorders. A growing number of whole-exome sequencing (WES) studies for BD has been performed, however, no research has examined the association between single nucleotide variants (SNVs) from WES and structural magnetic resonance imaging (MRI) data. METHODS We sequenced whole-exomes in 53 patients with BD and 82 healthy control participants at an initial discovery stage and investigated the impacts of SNVs in risk genes from WES analysis on the cortical gray-matter thickness and integrity of white matter tracts and in the following stage. Cortical thickness and white matter integrity were investigated using the FreeSurfer and TRACULA (Tracts Constrained by UnderLying Anatomy). RESULTS We identified 122 BD-related genes including KMT2C, AHNAK, CDH23, DCHS1, FRAS1, MACF1 and RYR3 and observed 27 recurrent copy number alteration regions including gain on 8p23.1 and loss on 15q11.1 - q11.2. Among them, single nucleotide polymorphism (SNP) rs4639425 in KMT2C gene, which regulates histone H3 lysine 4 (H3K4) methylation involved in chromatin remodeling, was associated with widespread alterations of white matter integrity including the cingulum, uncinate fasciculus, cortico-spinal tract, and superior longitudinal fasciculus. LIMITATION The small sample size of patients with BD in the genome data may cause our study to be underpowered when searching for putative rare mutations. CONCLUSION This study first combined a WES approach and neuroimaging findings in psychiatric disorders. We postulate the rs4639425 may be associated with BD-related microstructural changes of white matter tracts.
Collapse
Affiliation(s)
- Mi-Ryung Han
- Department of Laboratory Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Kyu-Man Han
- Department of Psychiatry, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Aram Kim
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Wooyoung Kang
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Youbin Kang
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - June Kang
- Department of Brain and Cognitive Engineering, Korea University, Seoul, Republic of Korea
| | - Eunsoo Won
- Department of Psychiatry, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Woo-Suk Tae
- Brain Convergence Research Center, Korea University Anam Hospital, Seoul, Republic of Korea
| | - Yunjung Cho
- Department of Laboratory Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Byung-Joo Ham
- Department of Psychiatry, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea; Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea; Brain Convergence Research Center, Korea University Anam Hospital, Seoul, Republic of Korea.
| |
Collapse
|
65
|
Wilkinson B, Evgrafov O, Zheng D, Hartel N, Knowles JA, Graham NA, Ichida J, Coba MP. Endogenous Cell Type-Specific Disrupted in Schizophrenia 1 Interactomes Reveal Protein Networks Associated With Neurodevelopmental Disorders. Biol Psychiatry 2019; 85:305-316. [PMID: 29961565 PMCID: PMC6251761 DOI: 10.1016/j.biopsych.2018.05.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 04/03/2018] [Accepted: 05/03/2018] [Indexed: 11/17/2022]
Abstract
BACKGROUND Disrupted in schizophrenia 1 (DISC1) has been implicated in a number of psychiatric diseases along with neurodevelopmental phenotypes such as the proliferation and differentiation of neural progenitor cells. While there has been significant effort directed toward understanding the function of DISC1 through the determination of its protein-protein interactions within an in vitro setting, endogenous interactions involving DISC1 within a cell type-specific setting relevant to neural development remain unclear. METHODS Using CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/CRISPR-associated protein 9) genome engineering technology, we inserted an endogenous 3X-FLAG tag at the C-terminus of the canonical DISC1 gene in human induced pluripotent stem cells (iPSCs). We further differentiated these cells and used affinity purification to determine protein-protein interactions involving DISC1 in iPSC-derived neural progenitor cells and astrocytes. RESULTS We were able to determine 151 novel cell type-specific proteins present in DISC1 endogenous interactomes. The DISC1 interactomes can be clustered into several subcomplexes that suggest novel DISC1 cell-specific functions. In addition, the DISC1 interactome in iPSC-derived neural progenitor cells associates in a connected network containing proteins found to harbor de novo mutations in patients affected by schizophrenia and contains a subset of novel interactions that are known to harbor syndromic mutations in neurodevelopmental disorders. CONCLUSIONS Endogenous DISC1 interactomes within iPSC-derived human neural progenitor cells and astrocytes are able to provide context to DISC1 function in a cell type-specific setting relevant to neural development and enables the integration of psychiatric disease risk factors within a set of defined molecular functions.
Collapse
Affiliation(s)
- Brent Wilkinson
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Oleg Evgrafov
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY 11203, USA
| | - DongQing Zheng
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90033, USA
| | - Nicolas Hartel
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90033, USA
| | - James A. Knowles
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY 11203, USA
| | - Nicholas A. Graham
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90033, USA
| | - Justin Ichida
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA,Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA,Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC
| | - Marcelo P. Coba
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA,Department of Psychiatry and Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA,Corresponding Author: Marcelo P. Coba, Keck School of Medicine, University of Southern California, Zilkha Neurogenetic Institute, 1501 San Pablo St, Los Angeles, CA 90033, USA. Phone: 323-442-4345.
| |
Collapse
|
66
|
Sawa A. DISC1 and Its Protein Interactomes for Mental Function. Biol Psychiatry 2019; 85:283-284. [PMID: 30665501 DOI: 10.1016/j.biopsych.2018.12.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 12/13/2018] [Accepted: 12/13/2018] [Indexed: 12/30/2022]
Affiliation(s)
- Akira Sawa
- Departments of Psychiatry, Neuroscience, Mental Health, and Biomedical Engineering, Johns Hopkins University School of Medicine and Bloomberg School of Public Health, Baltimore, Maryland.
| |
Collapse
|
67
|
Barnett BR, Torres-Velázquez M, Yi SY, Rowley PA, Sawin EA, Rubinstein CD, Krentz K, Anderson JM, Bakshi VP, Yu JPJ. Sex-specific deficits in neurite density and white matter integrity are associated with targeted disruption of exon 2 of the Disc1 gene in the rat. Transl Psychiatry 2019; 9:82. [PMID: 30745562 PMCID: PMC6370885 DOI: 10.1038/s41398-019-0429-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 01/24/2019] [Accepted: 01/26/2019] [Indexed: 02/06/2023] Open
Abstract
Diffusion tensor imaging (DTI) has provided remarkable insight into our understanding of white matter microstructure and brain connectivity across a broad spectrum of psychiatric disease. While DTI and other diffusion weighted magnetic resonance imaging (MRI) methods have clarified the axonal contribution to the disconnectivity seen in numerous psychiatric diseases, absent from these studies are quantitative indices of neurite density and orientation that are especially important features in regions of high synaptic density that would capture the synaptic contribution to the psychiatric disease state. Here we report the application of neurite orientation dispersion and density imaging (NODDI), an emerging microstructure imaging technique, to a novel Disc1 svΔ2 rat model of psychiatric illness and demonstrate the complementary and more specific indices of tissue microstructure found in NODDI than those reported by DTI. Our results demonstrate global and sex-specific changes in white matter microstructural integrity and deficits in neurite density as a consequence of the Disc1 svΔ2 genetic variation and highlight the application of NODDI and quantitative measures of neurite density and neurite dispersion in psychiatric disease.
Collapse
Affiliation(s)
- Brian R Barnett
- Neuroscience Training Program, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Maribel Torres-Velázquez
- Department of Biomedical Engineering, College of Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Sue Y Yi
- Neuroscience Training Program, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Paul A Rowley
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
| | - Emily A Sawin
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
| | - C Dustin Rubinstein
- Biotechnology Center, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Kathleen Krentz
- Biotechnology Center, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Jacqueline M Anderson
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
| | - Vaishali P Bakshi
- Department of Psychiatry, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
| | - John-Paul J Yu
- Neuroscience Training Program, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, Madison, WI, 53705, USA.
- Department of Biomedical Engineering, College of Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA.
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA.
- Department of Psychiatry, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA.
| |
Collapse
|
68
|
Park DI, Turck CW. Interactome Studies of Psychiatric Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1118:163-173. [PMID: 30747422 DOI: 10.1007/978-3-030-05542-4_8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
High comorbidity and complexity have precluded reliable diagnostic assessment and treatment of psychiatric disorders. Impaired molecular interactions may be relevant for underlying mechanisms of psychiatric disorders but by and large remain unknown. With the help of a number of publicly available databases and various technological tools, recent research has filled the paucity of information by generating a novel dataset of psychiatric interactomes. Different technological platforms including yeast two-hybrid screen, co-immunoprecipitation-coupled with mass spectrometry-based proteomics, and transcriptomics have been widely used in combination with cellular and molecular techniques to interrogate the psychiatric interactome. Novel molecular interactions have been identified in association with different psychiatric disorders including autism spectrum disorders, schizophrenia, bipolar disorder, and major depressive disorder. However, more extensive and sophisticated interactome research needs to be conducted to overcome the current limitations such as incomplete interactome databases and a lack of functional information among components. Ultimately, integrated psychiatric interactome databases will contribute to the implementation of biomarkers and therapeutic intervention.
Collapse
Affiliation(s)
- Dong Ik Park
- Danish Research Institute of Translational Neuroscience (DANDRITE), Department of Biomedicine, Aarhus University, Aarhus, Denmark.
| | - Christoph W Turck
- Proteomics and Biomarkers, Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| |
Collapse
|
69
|
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
|
70
|
Srikanth P, Lagomarsino VN, Pearse RV, Liao M, Ghosh S, Nehme R, Seyfried N, Eggan K, Young-Pearse TL. Convergence of independent DISC1 mutations on impaired neurite growth via decreased UNC5D expression. Transl Psychiatry 2018; 8:245. [PMID: 30410030 PMCID: PMC6224395 DOI: 10.1038/s41398-018-0281-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 07/16/2018] [Indexed: 12/29/2022] Open
Abstract
The identification of convergent phenotypes in different models of psychiatric illness highlights robust phenotypes that are more likely to be implicated in disease pathophysiology. Here, we utilize human iPSCs harboring distinct mutations in DISC1 that have been found in families with major mental illness. One mutation was engineered to mimic the consequences on DISC1 protein of a balanced translocation linked to mental illness in a Scottish pedigree; the other mutation was identified in an American pedigree with a high incidence of mental illness. Directed differentiation of these iPSCs using NGN2 expression shows rapid conversion to a homogenous population of mature excitatory neurons. Both DISC1 mutations result in reduced DISC1 protein expression, and show subtle effects on certain presynaptic proteins. In addition, RNA sequencing and qPCR showed decreased expression of UNC5D, DPP10, PCDHA6, and ZNF506 in neurons with both DISC1 mutations. Longitudinal analysis of neurite outgrowth revealed decreased neurite outgrowth in neurons with each DISC1 mutation, which was mimicked by UNC5D knockdown and rescued by transient upregulation of endogenous UNC5D. This study shows a narrow range of convergent phenotypes of two mutations found in families with major mental illness, and implicates dysregulated netrin signaling in DISC1 biology.
Collapse
Affiliation(s)
- Priya Srikanth
- 0000 0004 0378 8294grid.62560.37Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA USA
| | - Valentina N. Lagomarsino
- 0000 0004 0378 8294grid.62560.37Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA USA
| | - Richard V. Pearse
- 0000 0004 0378 8294grid.62560.37Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA USA
| | - Meichen Liao
- 0000 0004 0378 8294grid.62560.37Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA USA
| | - Sulagna Ghosh
- 000000041936754Xgrid.38142.3cHarvard Stem Cell Institute, Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138 USA ,grid.66859.34Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Ralda Nehme
- 000000041936754Xgrid.38142.3cHarvard Stem Cell Institute, Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138 USA ,grid.66859.34Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Nicholas Seyfried
- 0000 0001 0941 6502grid.189967.8Department of Biochemistry, Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322 USA
| | - Kevin Eggan
- 000000041936754Xgrid.38142.3cHarvard Stem Cell Institute, Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138 USA ,grid.66859.34Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Tracy L. Young-Pearse
- 0000 0004 0378 8294grid.62560.37Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA USA
| |
Collapse
|
71
|
Endo R, Takashima N, Nekooki-Machida Y, Komi Y, Hui KKW, Takao M, Akatsu H, Murayama S, Sawa A, Tanaka M. TAR DNA-Binding Protein 43 and Disrupted in Schizophrenia 1 Coaggregation Disrupts Dendritic Local Translation and Mental Function in Frontotemporal Lobar Degeneration. Biol Psychiatry 2018; 84:509-521. [PMID: 29752072 PMCID: PMC6123275 DOI: 10.1016/j.biopsych.2018.03.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 02/06/2018] [Accepted: 03/07/2018] [Indexed: 12/26/2022]
Abstract
BACKGROUND Neurodegenerative diseases involving protein aggregation often accompany psychiatric symptoms. Frontotemporal lobar degeneration (FTLD) associated with TAR DNA-binding protein 43 (TDP-43) aggregation is characterized by progressive neuronal atrophy in frontal and temporal lobes of cerebral cortex. Furthermore, patients with FTLD display mental dysfunction in multiple behavioral dimensions. Nevertheless, their molecular origin for psychiatric symptoms remains unclear. METHODS In FTLD neurons and mouse models with TDP-43 aggregates, we examined coaggregation between TDP-43 and disrupted in schizophrenia 1 (DISC1), a key player in the pathology of mental conditions and its effects on local translation in dendrites and psychiatric behaviors. The protein coaggregation and the expression level of synaptic proteins were also investigated with postmortem brains from patients with FTLD (n = 6). RESULTS We found cytosolic TDP-43/DISC1 coaggregates in brains of both FTLD mouse model and patients with FTLD. At the mechanistic levels, the TDP-43/DISC1 coaggregates disrupted the activity-dependent dendritic local translation through impairment of translation initiation and, in turn, reduced synaptic protein expression. Behavioral deficits detected in FTLD model mice were ameliorated by exogenous DISC1 expression. CONCLUSIONS Our findings reveal a novel role of the aggregate-prone TDP-43/DISC1 protein complex in regulating local translation, which affects aberrant behaviors relevant to multiple psychiatric dimensions.
Collapse
Affiliation(s)
- Ryo Endo
- Laboratory for Protein Conformation Diseases, RIKEN Brain Science Institute, Japan
| | - Noriko Takashima
- Laboratory for Protein Conformation Diseases, RIKEN Brain Science Institute, Japan
| | - Yoko Nekooki-Machida
- Laboratory for Protein Conformation Diseases, RIKEN Brain Science Institute, Japan
| | - Yusuke Komi
- Laboratory for Protein Conformation Diseases, RIKEN Brain Science Institute, Japan
| | - Kelvin Kai-Wan Hui
- Laboratory for Protein Conformation Diseases, RIKEN Brain Science Institute, Japan
| | - Masaki Takao
- Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Japan,Department of Neurology, Saitama Medical University, Japan
| | - Hiroyasu Akatsu
- Choju Medical Institute, Fukushimura Hospital, Japan,Department of Medicine for Aging in Place and Community-Based Medical Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Shigeo Murayama
- Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Japan
| | - Akira Sawa
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD.
| | - Motomasa Tanaka
- Laboratory for Protein Conformation Diseases, RIKEN Brain Science Institute, Wako, Japan.
| |
Collapse
|
72
|
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
|
73
|
Ka M, Moffat JJ, Kim WY. MACF1 Controls Migration and Positioning of Cortical GABAergic Interneurons in Mice. Cereb Cortex 2018; 27:5525-5538. [PMID: 27756764 DOI: 10.1093/cercor/bhw319] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 09/23/2016] [Indexed: 01/08/2023] Open
Abstract
GABAergic interneurons develop in the ganglionic eminence in the ventral telencephalon and tangentially migrate into the cortical plate during development. However, key molecules controlling interneuron migration remain poorly identified. Here, we show that microtubule-actin cross-linking factor 1 (MACF1) regulates GABAergic interneuron migration and positioning in the developing mouse brain. To investigate the role of MACF1 in developing interneurons, we conditionally deleted the MACF1 gene in mouse interneuron progenitors and their progeny using Dlx5/6-Cre-IRES-EGFP and Nkx2.1-Cre drivers. We found that MACF1 deletion results in a marked reduction and defective positioning of interneurons in the mouse cerebral cortex and hippocampus, suggesting abnormal interneuron migration. Indeed, the speed and mode of interneuron migration were abnormal in the MACF1-mutant brain, compared with controls. Additionally, MACF1-deleted interneurons showed a significant reduction in the length of their leading processes and dendrites in the mouse brain. Finally, loss of MACF1 decreased microtubule stability in cortical interneurons. Our findings suggest that MACF1 plays a critical role in cortical interneuron migration and positioning in the developing mouse brain.
Collapse
Affiliation(s)
- Minhan Ka
- Department of Developmental Neuroscience, Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Jeffrey J Moffat
- Department of Developmental Neuroscience, Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Woo-Yang Kim
- Department of Developmental Neuroscience, Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
74
|
Tropea D, Hardingham N, Millar K, Fox K. Mechanisms underlying the role of DISC1 in synaptic plasticity. J Physiol 2018; 596:2747-2771. [PMID: 30008190 PMCID: PMC6046077 DOI: 10.1113/jp274330] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 02/02/2018] [Indexed: 12/11/2022] Open
Abstract
Disrupted in schizophrenia 1 (DISC1) is an important hub protein, forming multimeric complexes by self-association and interacting with a large number of synaptic and cytoskeletal molecules. The synaptic location of DISC1 in the adult brain suggests a role in synaptic plasticity, and indeed, a number of studies have discovered synaptic plasticity impairments in a variety of different DISC1 mutants. This review explores the possibility that DISC1 is an important molecule for organizing proteins involved in synaptic plasticity and examines why mutations in DISC1 impair plasticity. It concentrates on DISC1's role in interacting with synaptic proteins, controlling dendritic structure and cellular trafficking of mRNA, synaptic vesicles and mitochondria. N-terminal directed mutations appear to impair synaptic plasticity through interactions with phosphodiesterase 4B (PDE4B) and hence protein kinase A (PKA)/GluA1 and PKA/cAMP response element-binding protein (CREB) signalling pathways, and affect spine structure through interactions with kalirin 7 (Kal-7) and Rac1. C-terminal directed mutations also impair plasticity possibly through altered interactions with lissencephaly protein 1 (LIS1) and nuclear distribution protein nudE-like 1 (NDEL1), thereby affecting developmental processes such as dendritic structure and spine maturation. Many of the same molecules involved in DISC1's cytoskeletal interactions are also involved in intracellular trafficking, raising the possibility that impairments in intracellular trafficking affect cytoskeletal development and vice versa. While the multiplicity of DISC1 protein interactions makes it difficult to pinpoint a single causal signalling pathway, we suggest that the immediate-term effects of N-terminal influences on GluA1, Rac1 and CREB, coupled with the developmental effects of C-terminal influences on trafficking and the cytoskeleton make up the two main branches of DISC1's effect on synaptic plasticity and dendritic spine stability.
Collapse
Affiliation(s)
- Daniela Tropea
- Neurospychiatric GeneticsTrinity Center for Health Sciences and Trinity College Institute of Neuroscience (TCIN)Trinity College DublinDublinIreland
| | - Neil Hardingham
- School of BiosciencesMuseum AvenueCardiff UniversityCardiffUK
| | - Kirsty Millar
- Centre for Genomic & Experimental MedicineMRC Institute of Genetics & Molecular MedicineWestern General HospitalUniversity of EdinburghCrewe RoadEdinburghUK
| | - Kevin Fox
- School of BiosciencesMuseum AvenueCardiff UniversityCardiffUK
| |
Collapse
|
75
|
Baltussen LL, Rosianu F, Ultanir SK. Kinases in synaptic development and neurological diseases. Prog Neuropsychopharmacol Biol Psychiatry 2018; 84:343-352. [PMID: 29241837 DOI: 10.1016/j.pnpbp.2017.12.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 12/08/2017] [Accepted: 12/09/2017] [Indexed: 10/18/2022]
Abstract
Neuronal morphogenesis and synapse development is essential for building a functioning nervous system, and defects in these processes are associated with neurological disorders. Our understanding of molecular components and signalling events that contribute to neuronal development and pathogenesis is limited. Genes associated with neurodevelopmental and neurodegenerative diseases provide entry points for elucidating molecular events that contribute to these conditions. Several protein kinases, enzymes that regulate protein function by phosphorylating their substrates, are genetically linked to neurological disorders. Identifying substrates of these kinases is key to discovering their function and providing insight for possible therapies. In this review, we describe how various methods for kinase-substrate identification helped elucidate kinase signalling pathways important for neuronal development and function. We describe recent advances on roles of kinases TAOK2, TNIK and CDKL5 in neuronal development and the converging pathways of LRRK2, PINK1 and GAK in Parkinson's Disease.
Collapse
Affiliation(s)
- Lucas L Baltussen
- Kinases and Brain Development Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom
| | - Flavia Rosianu
- Kinases and Brain Development Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom
| | - Sila K Ultanir
- Kinases and Brain Development Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom.
| |
Collapse
|
76
|
Faundez V, De Toma I, Bardoni B, Bartesaghi R, Nizetic D, de la Torre R, Cohen Kadosh R, Herault Y, Dierssen M, Potier MC. Translating molecular advances in Down syndrome and Fragile X syndrome into therapies. Eur Neuropsychopharmacol 2018; 28:675-690. [PMID: 29887288 DOI: 10.1016/j.euroneuro.2018.03.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 02/19/2018] [Accepted: 03/12/2018] [Indexed: 12/27/2022]
Abstract
Ongoing treatments for genetic developmental disorders of the central nervous system are mostly symptomatic and do not correct the genetic cause. Recent identification of common mechanisms between diseases has suggested that new therapeutic targets could be applied across intellectual disabilities with potential disease-modifying properties. The European Down syndrome and other genetic developmental disorders (DSG2D) network joined basic and clinical scientists to foster this research and carry out clinical trials. Here we discuss common mechanisms between several intellectual disabilities from genetic origin including Down's and Fragile X syndromes: i) how to model these complex diseases using neuronal cells and brain organoids derived from induced pluripotent stem cells; ii) how to integrate genomic, proteomic and interactome data to help defining common mechanisms and boundaries between diseases; iii) how to target common pathways for designing clinical trials and assessing their efficacy; iv) how to bring new neuro-therapies, such as noninvasive brain stimulations and cognitive training to clinical research. The basic and translational research efforts of the last years have utterly transformed our understanding of the molecular pathology of these diseases but much is left to be done to bring them to newborn babies and children to improve their quality of life.
Collapse
Affiliation(s)
- Victor Faundez
- Department of Cell Biology, Emory University, Atlanta, GA, USA
| | - Ilario De Toma
- Cellular and Systems Neurobiology, Center for Genomic Regulation, The Barcelona Institute of Science and Technology, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; Centro de Investigación Biomédica en Red CIBERER, Spain
| | - Barbara Bardoni
- Université Côte d'Azur, INSERM, CNRS, Institute of Molecular and Cellular Pharmacology, Valbonne, France
| | - Renata Bartesaghi
- University of Bologna, Department of Biomedical and Neuromotor Sciences, Bologna, Italy
| | - Dean Nizetic
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore; Barts and The London School of Medicine, Queen Mary University of London, United Kingdom
| | - Rafael de la Torre
- Integrated Pharmacology and Neurosciences Systems Research Group, IMIM-Hospital del Mar Medical Research Institute, Barcelona, Spain; CIBEROBN, Madrid, Spain
| | - Roi Cohen Kadosh
- Department of Experimental Psychology, University of Oxford, Oxford, United Kingdom
| | - Yann Herault
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Université de Strasbourg, Illkirch, France; Centre National de la Recherche Scientifique, UMR7104, Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France
| | - Mara Dierssen
- Cellular and Systems Neurobiology, Center for Genomic Regulation, The Barcelona Institute of Science and Technology, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; Centro de Investigación Biomédica en Red CIBERER, Spain.
| | - Marie-Claude Potier
- Institut du Cerveau et de la Moelle épinière, CNRS UMR7225, INSERM U1127, UPMC, Hôpital de la Pitié-Salpêtrière, 47 Bd de l'Hôpital, Paris, France.
| |
Collapse
|
77
|
Yalla K, Elliott C, Day JP, Findlay J, Barratt S, Hughes ZA, Wilson L, Whiteley E, Popiolek M, Li Y, Dunlop J, Killick R, Adams DR, Brandon NJ, Houslay MD, Hao B, Baillie GS. FBXW7 regulates DISC1 stability via the ubiquitin-proteosome system. Mol Psychiatry 2018; 23:1278-1286. [PMID: 28727686 PMCID: PMC5984089 DOI: 10.1038/mp.2017.138] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 05/12/2017] [Accepted: 05/15/2017] [Indexed: 01/27/2023]
Abstract
Disrupted in schizophrenia 1 (DISC1) is a multi-functional scaffolding protein that has been associated with neuropsychiatric disease. The role of DISC1 is to assemble protein complexes that promote neural development and signaling, hence tight control of the concentration of cellular DISC1 in neurons is vital to brain function. Using structural and biochemical techniques, we show for we believe the first time that not only is DISC1 turnover elicited by the ubiquitin proteasome system (UPS) but that it is orchestrated by the F-Box protein, FBXW7. We present the structure of FBXW7 bound to the DISC1 phosphodegron motif and exploit this information to prove that disruption of the FBXW7-DISC1 complex results in a stabilization of DISC1. This action can counteract DISC1 deficiencies observed in neural progenitor cells derived from induced pluripotent stem cells from schizophrenia patients with a DISC1 frameshift mutation. Thus manipulation of DISC1 levels via the UPS may provide a novel method to explore DISC1 function.
Collapse
Affiliation(s)
- K Yalla
- College of Veterinary Medical and Life Sciences, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - C Elliott
- College of Veterinary Medical and Life Sciences, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
- Institute of Psychiatry, Psychology and Neuroscience, King’s College, London, UK
| | - J P Day
- College of Veterinary Medical and Life Sciences, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - J Findlay
- College of Veterinary Medical and Life Sciences, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - S Barratt
- College of Veterinary Medical and Life Sciences, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Z A Hughes
- Neuroscience Research Unit, Pfizer Inc, Cambridge, MA, USA
| | - L Wilson
- Neuroscience Research Unit, Pfizer Inc, Cambridge, MA, USA
| | - E Whiteley
- College of Veterinary Medical and Life Sciences, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - M Popiolek
- Neuroscience Research Unit, Pfizer Inc, Cambridge, MA, USA
| | - Y Li
- Department of Molecular Biology and Biophysics, University of Connecticut Health Centre, Farmington, CT, USA
| | - J Dunlop
- AstraZeneca, Neuroscience, Innovative Medicines & Early Development, Waltham, MA, USA
| | - R Killick
- Institute of Psychiatry, Psychology and Neuroscience, King’s College, London, UK
| | - D R Adams
- Institute of Chemical Sciences, Heriot-Watt University, Edinburgh, UK
| | - N J Brandon
- AstraZeneca, Neuroscience, Innovative Medicines & Early Development, Waltham, MA, USA
| | - M D Houslay
- Institute of Pharmaceutical Science, King’s College, London, UK
| | - B Hao
- Department of Molecular Biology and Biophysics, University of Connecticut Health Centre, Farmington, CT, USA
| | - G S Baillie
- College of Veterinary Medical and Life Sciences, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
78
|
Teng S, Thomson PA, McCarthy S, Kramer M, Muller S, Lihm J, Morris S, Soares DC, Hennah W, Harris S, Camargo LM, Malkov V, McIntosh AM, Millar JK, Blackwood DH, Evans KL, Deary IJ, Porteous DJ, McCombie WR. Rare disruptive variants in the DISC1 Interactome and Regulome: association with cognitive ability and schizophrenia. Mol Psychiatry 2018; 23:1270-1277. [PMID: 28630456 PMCID: PMC5984079 DOI: 10.1038/mp.2017.115] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 03/20/2017] [Accepted: 03/27/2017] [Indexed: 12/20/2022]
Abstract
Schizophrenia (SCZ), bipolar disorder (BD) and recurrent major depressive disorder (rMDD) are common psychiatric illnesses. All have been associated with lower cognitive ability, and show evidence of genetic overlap and substantial evidence of pleiotropy with cognitive function and neuroticism. Disrupted in schizophrenia 1 (DISC1) protein directly interacts with a large set of proteins (DISC1 Interactome) that are involved in brain development and signaling. Modulation of DISC1 expression alters the expression of a circumscribed set of genes (DISC1 Regulome) that are also implicated in brain biology and disorder. Here we report targeted sequencing of 59 DISC1 Interactome genes and 154 Regulome genes in 654 psychiatric patients and 889 cognitively-phenotyped control subjects, on whom we previously reported evidence for trait association from complete sequencing of the DISC1 locus. Burden analyses of rare and singleton variants predicted to be damaging were performed for psychiatric disorders, cognitive variables and personality traits. The DISC1 Interactome and Regulome showed differential association across the phenotypes tested. After family-wise error correction across all traits (FWERacross), an increased burden of singleton disruptive variants in the Regulome was associated with SCZ (FWERacross P=0.0339). The burden of singleton disruptive variants in the DISC1 Interactome was associated with low cognitive ability at age 11 (FWERacross P=0.0043). These results identify altered regulation of schizophrenia candidate genes by DISC1 and its core Interactome as an alternate pathway for schizophrenia risk, consistent with the emerging effects of rare copy number variants associated with intellectual disability.
Collapse
Affiliation(s)
- S Teng
- Stanley Institute for Cognitive Genomics, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
- Department of Biology, Howard University, Washington DC, USA
| | - P A Thomson
- Centre for Genomic and Experimental Medicine, MRC/University of Edinburgh Institute of Genetics & Molecular Medicine, Western General Hospital, Edinburgh, UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, Edinburgh, UK
| | - S McCarthy
- Stanley Institute for Cognitive Genomics, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - M Kramer
- Stanley Institute for Cognitive Genomics, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - S Muller
- Stanley Institute for Cognitive Genomics, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - J Lihm
- Stanley Institute for Cognitive Genomics, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - S Morris
- Centre for Genomic and Experimental Medicine, MRC/University of Edinburgh Institute of Genetics & Molecular Medicine, Western General Hospital, Edinburgh, UK
| | - D C Soares
- Centre for Genomic and Experimental Medicine, MRC/University of Edinburgh Institute of Genetics & Molecular Medicine, Western General Hospital, Edinburgh, UK
| | - W Hennah
- Institute for Molecular Medicine, Finland FIMM, University of Helsinki, Helsinki, Finland
| | - S Harris
- Centre for Genomic and Experimental Medicine, MRC/University of Edinburgh Institute of Genetics & Molecular Medicine, Western General Hospital, Edinburgh, UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, Edinburgh, UK
| | - L M Camargo
- UCB New Medicines, One Broadway, Cambridge, MA, USA
| | - V Malkov
- Genetics and Pharmacogenomics, MRL, Merck & Co, Boston, MA, USA
| | - A M McIntosh
- Division of Psychiatry, University of Edinburgh, Royal Edinburgh Hospital, Edinburgh, UK
| | - J K Millar
- Centre for Genomic and Experimental Medicine, MRC/University of Edinburgh Institute of Genetics & Molecular Medicine, Western General Hospital, Edinburgh, UK
| | - D H Blackwood
- Division of Psychiatry, University of Edinburgh, Royal Edinburgh Hospital, Edinburgh, UK
| | - K L Evans
- Centre for Cognitive Ageing and Cognitive Epidemiology, Edinburgh, UK
| | - I J Deary
- Centre for Cognitive Ageing and Cognitive Epidemiology, Edinburgh, UK
- Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - D J Porteous
- Centre for Genomic and Experimental Medicine, MRC/University of Edinburgh Institute of Genetics & Molecular Medicine, Western General Hospital, Edinburgh, UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, Edinburgh, UK
| | - W R McCombie
- Stanley Institute for Cognitive Genomics, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| |
Collapse
|
79
|
LaMarca EA, Powell SK, Akbarian S, Brennand KJ. Modeling Neuropsychiatric and Neurodegenerative Diseases With Induced Pluripotent Stem Cells. Front Pediatr 2018; 6:82. [PMID: 29666786 PMCID: PMC5891587 DOI: 10.3389/fped.2018.00082] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 03/15/2018] [Indexed: 12/19/2022] Open
Abstract
Human-induced pluripotent stem cells (hiPSCs) have revolutionized our ability to model neuropsychiatric and neurodegenerative diseases, and recent progress in the field is paving the way for improved therapeutics. In this review, we discuss major advances in generating hiPSC-derived neural cells and cutting-edge techniques that are transforming hiPSC technology, such as three-dimensional "mini-brains" and clustered, regularly interspersed short palindromic repeats (CRISPR)-Cas systems. We examine specific examples of how hiPSC-derived neural cells are being used to uncover the pathophysiology of schizophrenia and Parkinson's disease, and consider the future of this groundbreaking research.
Collapse
Affiliation(s)
- Elizabeth A. LaMarca
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Samuel K. Powell
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Medical Scientist Training Program, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Schahram Akbarian
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Kristen J. Brennand
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
80
|
Disrupted in schizophrenia 1 (DISC1) inhibits glioblastoma development by regulating mitochondria dynamics. Oncotarget 2018; 7:85963-85974. [PMID: 27852062 PMCID: PMC5349889 DOI: 10.18632/oncotarget.13290] [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: 08/15/2016] [Accepted: 11/07/2016] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma(GBM) is one of the most common and aggressive malignant primary tumors of the central nervous system and mitochondria have been proposed to participate in GBM tumorigenesis. Previous studies have identified a potential role of Disrupted in Schizophrenia 1 (DISC1), a multi-compartmentalized protein, in mitochondria. But whether DISC1 could regulate GBM tumorigenesis via mitochondria is still unknown. We determined the expression level of DISC1 by both bioinformatics analysis and tissue analysis, and found that DISC1 was highly expressed in GBM. Knocking down of DISC1 by shRNA in GBM cells significantly inhibited cell proliferation both in vitro and in vivo. In addition, down-regulation of DISC1 decreased cell migration and invasion of GBM and self renewal capacity of glioblastoma stem-like cells. Furthermore, multiple independent rings or spheres could be observed in mitochondria in GBM depleted of DISC1, while normal filamentous morphology was observed in control cells, demonstrating that DISC1 affected the mitochondrial dynamic. Dynamin-related protein 1 (Drp1) was reported to contribute to mitochondrial dynamic regulation and influence glioma cells proliferation and invasion by RHOA/ ROCK1 pathway. Our data showed a significant decrease of Drp1 both in mRNA and protein level in GBM lack of DISC1, indicating that DISC1 maybe affect the mitochondrial dynamic by regulating Drp1. Taken together, our findings reveal that DISC1 affects glioblastoma cell development via mitochondria dynamics partly by down regulation of Drp1.
Collapse
|
81
|
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
|
82
|
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
|
83
|
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
|
84
|
Recabarren-Leiva D, Alarcón M. New insights into the gene expression associated to amyotrophic lateral sclerosis. Life Sci 2018; 193:110-123. [DOI: 10.1016/j.lfs.2017.12.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Revised: 12/01/2017] [Accepted: 12/10/2017] [Indexed: 12/11/2022]
|
85
|
Wild AR, Dell'Acqua ML. Potential for therapeutic targeting of AKAP signaling complexes in nervous system disorders. Pharmacol Ther 2017; 185:99-121. [PMID: 29262295 DOI: 10.1016/j.pharmthera.2017.12.004] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
A common feature of neurological and neuropsychiatric disorders is a breakdown in the integrity of intracellular signal transduction pathways. Dysregulation of ion channels and receptors in the cell membrane and the enzymatic mediators that link them to intracellular effectors can lead to synaptic dysfunction and neuronal death. However, therapeutic targeting of these ubiquitous signaling elements can lead to off-target side effects due to their widespread expression in multiple systems of the body. A-kinase anchoring proteins (AKAPs) are multivalent scaffolding proteins that compartmentalize a diverse range of receptor and effector proteins to streamline signaling within nanodomain signalosomes. A number of essential neurological processes are known to critically depend on AKAP-directed signaling and an understanding of the role AKAPs play in nervous system disorders has emerged in recent years. Selective targeting of AKAP protein-protein interactions may be a means to uncouple pathologically active signaling pathways in neurological disorders with a greater degree of specificity. In this review we will discuss the role of AKAPs in both regulating normal nervous system function and dysfunction associated with disease, and the potential for therapeutic targeting of AKAP signaling complexes.
Collapse
Affiliation(s)
- Angela R Wild
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Mark L Dell'Acqua
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA.
| |
Collapse
|
86
|
Riemann D, Wallrafen R, Dresbach T. The Kohlschütter-Tönz syndrome associated gene Rogdi encodes a novel presynaptic protein. Sci Rep 2017; 7:15791. [PMID: 29150638 PMCID: PMC5693994 DOI: 10.1038/s41598-017-16004-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 11/06/2017] [Indexed: 11/09/2022] Open
Abstract
Mutations in the human homolog of the Drosophila gene Rogdi cause Kohlschütter-Tönz syndrome. This disorder is characterised by amelogenesis imperfecta, as well as severe neurological symptoms including epilepsy and psychomotor delay. However, little is known about the protein encoded by Rogdi, and hence the pathogenic mechanisms underlying Kohlschütter-Tönz syndrome have remained elusive. Using immunofluorescence of rat cultured hippocampal neurons and brain sections we find that Rogdi is enriched at synaptic sites. In addition, recombinant GFP-Rogdi expressed in cultured neurons was efficiently targeted to presynaptic sites, where it colocalised with the presynaptic scaffolding protein Bassoon and the synaptic vesicle markers Synaptophysin, Synapsin-1, VAMP2/Synaptobrevin and Mover. Our data indicate that GFP-Rogdi harbours efficient signals for presynaptic targeting, and that Rogdi is a presynaptic protein. Thus, the neurological symptoms associated with Kohlschütter-Tönz syndrome may arise from presynaptic dysfunction.
Collapse
Affiliation(s)
- Donatus Riemann
- Institute for Anatomy and Embryology, University Medical Centre Göttingen, Kreuzbergring 36, 37075, Göttingen, Germany
| | - Rebecca Wallrafen
- Institute for Anatomy and Embryology, University Medical Centre Göttingen, Kreuzbergring 36, 37075, Göttingen, Germany
| | - Thomas Dresbach
- Institute for Anatomy and Embryology, University Medical Centre Göttingen, Kreuzbergring 36, 37075, Göttingen, Germany.
| |
Collapse
|
87
|
DISC1 Regulates Neurogenesis via Modulating Kinetochore Attachment of Ndel1/Nde1 during Mitosis. Neuron 2017; 96:1041-1054.e5. [PMID: 29103808 DOI: 10.1016/j.neuron.2017.10.010] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Revised: 09/18/2017] [Accepted: 10/05/2017] [Indexed: 02/08/2023]
Abstract
Mutations of DISC1 (disrupted-in-schizophrenia 1) have been associated with major psychiatric disorders. Despite the hundreds of DISC1-binding proteins reported, almost nothing is known about how DISC1 interacts with other proteins structurally to impact human brain development. Here we solved the high-resolution structure of DISC1 C-terminal tail in complex with its binding domain of Ndel1. Mechanistically, DISC1 regulates Ndel1's kinetochore attachment, but not its centrosome localization, during mitosis. Functionally, disrupting DISC1/Ndel1 complex formation prolongs mitotic length and interferes with cell-cycle progression in human cells, and it causes cell-cycle deficits of radial glial cells in the embryonic mouse cortex and human forebrain organoids. We also observed similar deficits in organoids derived from schizophrenia patient induced pluripotent stem cells (iPSCs) with a DISC1 mutation that disrupts its interaction with Ndel1. Our study uncovers a new mechanism of action for DISC1 based on its structure, and it has implications for how genetic insults may contribute to psychiatric disorders.
Collapse
|
88
|
Pandey H, Bourahmoune K, Honda T, Honjo K, Kurita K, Sato T, Sawa A, Furukubo-Tokunaga K. Genetic interaction of DISC1 and Neurexin in the development of fruit fly glutamatergic synapses. NPJ SCHIZOPHRENIA 2017; 3:39. [PMID: 29079805 PMCID: PMC5660244 DOI: 10.1038/s41537-017-0040-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 09/19/2017] [Accepted: 09/27/2017] [Indexed: 12/31/2022]
Abstract
Originally identified at the breakpoint of a (1;11)(q42.1; q14.3) chromosomal translocation in a Scottish family with a wide range of mental disorders, the DISC1 gene has been a focus of intensive investigations as an entry point to study the molecular mechanisms of diverse mental dysfunctions. Perturbations of the DISC1 functions lead to behavioral changes in animal models, which are relevant to psychiatric conditions in patients. In this work, we have expressed the human DISC1 gene in the fruit fly (Drosophila melanogaster) and performed a genetic screening for the mutations of psychiatric risk genes that cause modifications of DISC1 synaptic phenotypes at the neuromuscular junction. We found that DISC1 interacts with dnrx1, the Drosophila homolog of the human Neurexin (NRXN1) gene, in the development of glutamatergic synapses. While overexpression of DISC1 suppressed the total bouton area on the target muscles and stimulated active zone density in wild-type background, a partial reduction of the dnrx1 activity negated the DISC1–mediated synaptic alterations. Likewise, overexpression of DISC1 stimulated the expression of a glutamate receptor component, DGLURIIA, in wild-type background but not in the dnrx1 heterozygous background. In addition, DISC1 caused mislocalization of Discs large, the Drosophila PSD-95 homolog, in the dnrx1 heterozygous background. Analyses with a series of domain deletions have revealed the importance of axonal localization of the DISC1 protein for efficient suppression of DNRX1 in synaptic boutons. These results thus suggest an intriguing converging mechanism controlled by the interaction of DISC1 and Neurexin in the developing glutamatergic synapses. Fruit fly models uncover a potential new mechanism by which two schizophrenia risk factor genes interact to alter synaptic junctions. DISC1 gene alterations have previously been linked to psychiatric anomalies, although the gene has not been formally recognized as a schizophrenia risk factor. A US-Japan research collaboration led by the University of Tsukuba’s Katsuo Furukubo-Tokunaga expressed human DISC1 in fruit fly synapses to better understand the changes that take place when gene disruption leads to overexpression. The team found that overexpression of DISC1 affected the expression of the fruit fly counterpart to human ‘neurexin,’ a known risk factor for conditions including schizophrenia and autism spectrum disorders. The interaction between neurexin and DISC1 also influenced other synapse-altering genes. Further research is warranted to explore the roles of DISC1 and neurexin in psychiatric disease.
Collapse
Affiliation(s)
- Himani Pandey
- Life and Environmental Sciences, University of Tsukuba, Tsukuba, 305-8572, Japan
| | - Katia Bourahmoune
- Life and Environmental Sciences, University of Tsukuba, Tsukuba, 305-8572, Japan
| | - Takato Honda
- Life and Environmental Sciences, University of Tsukuba, Tsukuba, 305-8572, Japan
| | - Ken Honjo
- Life and Environmental Sciences, University of Tsukuba, Tsukuba, 305-8572, Japan
| | - Kazuki Kurita
- Life and Environmental Sciences, University of Tsukuba, Tsukuba, 305-8572, Japan
| | - Tomohito Sato
- Life and Environmental Sciences, University of Tsukuba, Tsukuba, 305-8572, Japan
| | - Akira Sawa
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | |
Collapse
|
89
|
Hartwig C, Monis WJ, Chen X, Dickman DK, Pazour GJ, Faundez V. Neurodevelopmental disease mechanisms, primary cilia, and endosomes converge on the BLOC-1 and BORC complexes. Dev Neurobiol 2017; 78:311-330. [PMID: 28986965 DOI: 10.1002/dneu.22542] [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: 08/11/2017] [Revised: 09/08/2017] [Accepted: 09/15/2017] [Indexed: 12/12/2022]
Abstract
The biogenesis of lysosome-related organelles complex-1 (BLOC-1) and the bloc-one-related complex (BORC) are the cytosolic protein complexes required for specialized membrane protein traffic along the endocytic route and the spatial distribution of endosome-derived compartments, respectively. BLOC-1 and BORC complex subunits and components of their interactomes have been associated with the risk and/or pathomechanisms of neurodevelopmental disorders. Thus, cellular processes requiring BLOC-1 and BORC interactomes have the potential to offer novel insight into mechanisms underlying behavioral defects. We focus on interactions between BLOC-1 or BORC subunits with the actin and microtubule cytoskeleton, membrane tethers, and SNAREs. These interactions highlight requirements for BLOC-1 and BORC in membrane movement by motors, control of actin polymerization, and targeting of membrane proteins to specialized cellular domains such as the nerve terminal and the primary cilium. We propose that the endosome-primary cilia pathway is an underappreciated hub in the genesis and mechanisms of neurodevelopmental disorders. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 78: 311-330, 2018.
Collapse
Affiliation(s)
- Cortnie Hartwig
- Department of Cell Biology, Emory University, Atlanta, Georgia, 30322
| | - William J Monis
- Program in Molecular Medicine, University of Massachusetts Medical School, Biotech II, Worcester, Massachusetts, 01605
| | - Xun Chen
- Department of Biology, Neurobiology Section, University of Southern California, Los Angeles, California, 90089
| | - Dion K Dickman
- Department of Biology, Neurobiology Section, University of Southern California, Los Angeles, California, 90089
| | - Gregory J Pazour
- Program in Molecular Medicine, University of Massachusetts Medical School, Biotech II, Worcester, Massachusetts, 01605
| | - Victor Faundez
- Department of Cell Biology, Emory University, Atlanta, Georgia, 30322
| |
Collapse
|
90
|
Norkett R, Modi S, Kittler JT. Mitochondrial roles of the psychiatric disease risk factor DISC1. Schizophr Res 2017; 187:47-54. [PMID: 28087269 DOI: 10.1016/j.schres.2016.12.025] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Revised: 12/17/2016] [Accepted: 12/22/2016] [Indexed: 12/31/2022]
Abstract
Ion transport during neuronal signalling utilizes the majority of the brain's energy supply. Mitochondria are key sites for energy provision through ATP synthesis and play other important roles including calcium buffering. Thus, tightly regulated distribution and function of these organelles throughout the intricate architecture of the neuron is essential for normal synaptic communication. Therefore, delineating mechanisms coordinating mitochondrial transport and function is essential for understanding nervous system physiology and pathology. While aberrant mitochondrial transport and dynamics have long been associated with neurodegenerative disease, they have also more recently been linked to major mental illness including schizophrenia, autism and depression. However, the underlying mechanisms have yet to be elucidated, due to an incomplete understanding of the combinations of genetic and environmental factors contributing to these conditions. Consequently, the DISC1 gene has undergone intense study since its discovery at the site of a balanced chromosomal translocation, segregating with mental illness in a Scottish pedigree. The precise molecular functions of DISC1 remain elusive. Reported functions of DISC1 include regulation of intracellular signalling pathways, neuronal migration and dendritic development. Intriguingly, a role for DISC1 in mitochondrial homeostasis and transport is fast emerging. Therefore, a major function of DISC1 in regulating mitochondrial distribution, ATP synthesis and calcium buffering may be disrupted in psychiatric disease. In this review, we discuss the links between DISC1 and mitochondria, considering both trafficking of these organelles and their function, and how, via these processes, DISC1 may contribute to the regulation of neuronal behavior in normal and psychiatric disease states.
Collapse
Affiliation(s)
- R Norkett
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, UK
| | - S Modi
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, UK
| | - J T Kittler
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, UK.
| |
Collapse
|
91
|
|
92
|
Murphy LC, Millar JK. Regulation of mitochondrial dynamics by DISC1, a putative risk factor for major mental illness. Schizophr Res 2017; 187:55-61. [PMID: 28082141 DOI: 10.1016/j.schres.2016.12.027] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Revised: 12/22/2016] [Accepted: 12/23/2016] [Indexed: 12/22/2022]
Abstract
Mitochondria are dynamic organelles that are essential to power the process of neurotransmission. Neurons must therefore ensure that mitochondria maintain their functional integrity and are efficiently transported along the full extent of the axons and dendrites, from soma to synapses. Mitochondrial dynamics (trafficking, fission and fusion) co-ordinately regulate mitochondrial quality control and function. DISC1 is a component of the mitochondrial transport machinery and regulates mitochondrial dynamics. DISC1's role in this is adversely affected by sequence variants connected to brain structure/function and disease risk, and by mutant truncation. The DISC1 interactors NDE1 and GSK3β are also involved, indicating a convergence of putative risk factors for psychiatric illness upon mitochondrial dynamics.
Collapse
Affiliation(s)
- Laura C Murphy
- Centre for Genomic and Experimental Medicine, MRC Institute of Genetic and Molecular Medicine at the University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, UK
| | - J Kirsty Millar
- Centre for Genomic and Experimental Medicine, MRC Institute of Genetic and Molecular Medicine at the University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, UK.
| |
Collapse
|
93
|
Wu Q, Tang W, Luo Z, Li Y, Shu Y, Yue Z, Xiao B, Feng L. DISC1 Regulates the Proliferation and Migration of Mouse Neural Stem/Progenitor Cells through Pax5, Sox2, Dll1 and Neurog2. Front Cell Neurosci 2017; 11:261. [PMID: 28900388 PMCID: PMC5581844 DOI: 10.3389/fncel.2017.00261] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 08/11/2017] [Indexed: 11/27/2022] Open
Abstract
Background: Disrupted-in-schizophrenia 1 (DISC1) regulates neurogenesis and is a genetic risk factor for major psychiatric disorders. However, how DISC1 dysfunction affects neurogenesis and cell cycle progression at the molecular level is still unknown. Here, we investigated the role of DISC1 in regulating proliferation, migration, cell cycle progression and apoptosis in mouse neural stem/progenitor cells (MNSPCs) in vitro. Methods: MNSPCs were isolated and cultured from mouse fetal hippocampi. Retroviral vectors or siRNAs were used to manipulate DISC1 expression in MNSPCs. Proliferation, migration, cell cycle progression and apoptosis of altered MNSPCs were analyzed in cell proliferation assays (MTS), transwell system and flow cytometry. A neurogenesis specific polymerase chain reaction (PCR) array was used to identify genes downstream of DISC1, and functional analysis was performed through transfection of expression plasmids and siRNAs. Results: Loss of DISC1 reduced proliferation and migration of MNSPCs, while an increase in DISC1 led to increased proliferation and migration. Meanwhile, an increase in the proportion of cells in G0/G1 phase was concomitant with reduced levels of DISC1, but significant changes were not observed in the number MNSPCs undergoing apoptosis. Paired box gene 5 (Pax5), sex determining region Y-box 2 (Sox2), delta-like1 (Dll1) and Neurogenin2 (Neurog2) emerged as candidate molecules downstream of DISC1, and rescue experiments demonstrated that increased or decreased expression of either molecule regulated proliferation and migration in DISC1-altered MNSPCs. Conclusion: These results suggest that Pax5, Sox2, Dll1 and Neurog2 mediate DISC1 activity in MNSPC proliferation and migration.
Collapse
Affiliation(s)
- Qian Wu
- Department of Neurology, First Affiliated Hospital, Kunming Medical UniversityKunming, China
- Department of Neurology, Xiangya Hospital, Central South UniversityChangsha, China
| | - Weiting Tang
- Department of Neurology, Xiangya Hospital, Central South UniversityChangsha, China
| | - Zhaohui Luo
- Department of Neurology, Xiangya Hospital, Central South UniversityChangsha, China
| | - Yi Li
- Department of Neurology, University of Massachusetts Medical SchoolWorcester, MA, United States
| | - Yi Shu
- Department of Neurology, The Second Xiangya Hospital, Central South UniversityChangsha, China
| | - Zongwei Yue
- Department of Neurology, Xiangya Hospital, Central South UniversityChangsha, China
- Department of Neurology, Yale University School of MedicineNew Haven, CT, United States
| | - Bo Xiao
- Department of Neurology, Xiangya Hospital, Central South UniversityChangsha, China
| | - Li Feng
- Department of Neurology, Xiangya Hospital, Central South UniversityChangsha, China
- Department of Neurology, Yale University School of MedicineNew Haven, CT, United States
| |
Collapse
|
94
|
Tomoda T, Hikida T, Sakurai T. Role of DISC1 in Neuronal Trafficking and its Implication in Neuropsychiatric Manifestation and Neurotherapeutics. Neurotherapeutics 2017; 14:623-629. [PMID: 28664299 PMCID: PMC5509643 DOI: 10.1007/s13311-017-0556-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Disrupted-in-schizophrenia 1 (DISC1) was initially identified as a gene disrupted by a translocation mutation co-segregating with a variety of psychotic and mood disorders in a Scottish pedigree. In agreement with this original finding, mouse models that perturb Disc1 display deficits of behaviors in specific dimensions, such as cognition and emotion, but not a motor dimension. Although DISC1 is not a risk gene for sporadic cases of specific psychiatric disorders defined by categorical diagnostic criteria (e.g., schizophrenia and major depressive disorder), DISC1 is now regarded as an important molecular lead to decipher molecular pathology for specific dimensions relevant to major mental illnesses. Emerging evidence points to the role of DISC1 in the regulation of intracellular trafficking of a wide range of neuronal cargoes. We will review recent progress in this aspect of DISC1 biology and discuss how we could utilize this body of knowledge to better understand the pathophysiology of mental illnesses.
Collapse
Affiliation(s)
- Toshifumi Tomoda
- Centre for Addiction and Mental Health, University of Toronto, Toronto, Canada.
| | - Takatoshi Hikida
- Laboratory for Advanced Brain Functions, Institute for Protein Research, Osaka University, Osaka, Japan
| | - Takeshi Sakurai
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
95
|
Moffat JJ, Ka M, Jung EM, Smith AL, Kim WY. The role of MACF1 in nervous system development and maintenance. Semin Cell Dev Biol 2017; 69:9-17. [PMID: 28579452 DOI: 10.1016/j.semcdb.2017.05.020] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 05/12/2017] [Accepted: 05/29/2017] [Indexed: 12/14/2022]
Abstract
Microtubule-actin crosslinking factor 1 (MACF1), also known as actin crosslinking factor 7 (ACF7), is essential for proper modulation of actin and microtubule cytoskeletal networks. Most MACF1 isoforms are expressed broadly in the body, but some are exclusively found in the nervous system. Consequentially, MACF1 is integrally involved in multiple neural processes during development and in adulthood, including neurite outgrowth and neuronal migration. Furthermore, MACF1 participates in several signaling pathways, including the Wnt/β-catenin and GSK-3 signaling pathways, which regulate key cellular processes, such as proliferation and cell migration. Genetic mutation or dysregulation of the MACF1 gene has been associated with neurodevelopmental and neurodegenerative diseases, specifically schizophrenia and Parkinson's disease. MACF1 may also play a part in neuromuscular disorders and have a neuroprotective role in the optic nerve. In this review, the authors seek to synthesize recent findings relating to the roles of MACF1 within the nervous system and explore potential novel functions of MACF1 not yet examined.
Collapse
Affiliation(s)
- Jeffrey J Moffat
- Department of Developmental Neuroscience, Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Minhan Ka
- Department of Developmental Neuroscience, Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Eui-Man Jung
- Department of Developmental Neuroscience, Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Amanda L Smith
- Department of Developmental Neuroscience, Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Woo-Yang Kim
- Department of Developmental Neuroscience, Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
96
|
Blanco‐Suárez E, Caldwell ALM, Allen NJ. Role of astrocyte-synapse interactions in CNS disorders. J Physiol 2017; 595:1903-1916. [PMID: 27381164 PMCID: PMC5350444 DOI: 10.1113/jp270988] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 06/08/2016] [Indexed: 12/18/2022] Open
Abstract
Astrocytes comprise half of the cells in the brain. Although astrocytes have traditionally been described as playing a supportive role for neurons, they have recently been recognized as active participants in the development and plasticity of dendritic spines and synapses. Astrocytes can eliminate dendritic spines, induce synapse formation, and regulate neurotransmission and plasticity. Dendritic spine and synapse impairments are features of many neurological disorders, including autism spectrum disorder, schizophrenia, and Alzheimer's disease. In this review we will present evidence from multiple neurological disorders demonstrating that changes in astrocyte-synapse interaction contribute to the pathologies. Genomic analysis has connected altered astrocytic gene expression with synaptic deficits in a number of neurological disorders. Alterations in astrocyte-secreted factors have been implicated in the neuronal morphology and synaptic changes seen in neurodevelopmental disorders, while alteration in astrocytic glutamate uptake is a core feature of multiple neurodegenerative disorders. This evidence clearly demonstrates that maintaining astrocyte-synapse interaction is crucial for normal central nervous system functioning. Obtaining a better understanding of the role of astrocytes at synapses in health and disease will provide a new avenue for future therapeutic targeting.
Collapse
Affiliation(s)
- Elena Blanco‐Suárez
- Salk Institute for Biological StudiesMolecular Neuroscience Laboratory10010 North Torrey Pines RdLa JollaCA92037USA
| | - Alison L. M. Caldwell
- Salk Institute for Biological StudiesMolecular Neuroscience Laboratory10010 North Torrey Pines RdLa JollaCA92037USA
| | - Nicola J. Allen
- Salk Institute for Biological StudiesMolecular Neuroscience Laboratory10010 North Torrey Pines RdLa JollaCA92037USA
| |
Collapse
|
97
|
Emerging Synaptic Molecules as Candidates in the Etiology of Neurological Disorders. Neural Plast 2017; 2017:8081758. [PMID: 28331639 PMCID: PMC5346360 DOI: 10.1155/2017/8081758] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 02/06/2017] [Indexed: 01/06/2023] Open
Abstract
Synapses are complex structures that allow communication between neurons in the central nervous system. Studies conducted in vertebrate and invertebrate models have contributed to the knowledge of the function of synaptic proteins. The functional synapse requires numerous protein complexes with specialized functions that are regulated in space and time to allow synaptic plasticity. However, their interplay during neuronal development, learning, and memory is poorly understood. Accumulating evidence links synapse proteins to neurodevelopmental, neuropsychiatric, and neurodegenerative diseases. In this review, we describe the way in which several proteins that participate in cell adhesion, scaffolding, exocytosis, and neurotransmitter reception from presynaptic and postsynaptic compartments, mainly from excitatory synapses, have been associated with several synaptopathies, and we relate their functions to the disease phenotype.
Collapse
|
98
|
Kassan A, Egawa J, Zhang Z, Almenar-Queralt A, Nguyen QM, Lajevardi Y, Kim K, Posadas E, Jeste DV, Roth DM, Patel PM, Patel HH, Head BP. Caveolin-1 regulation of disrupted-in-schizophrenia-1 as a potential therapeutic target for schizophrenia. J Neurophysiol 2017; 117:436-444. [PMID: 27832597 PMCID: PMC5253400 DOI: 10.1152/jn.00481.2016] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 10/31/2016] [Indexed: 02/08/2023] Open
Abstract
Schizophrenia is a debilitating psychiatric disorder manifested in early adulthood. Disrupted-in-schizophrenia-1 (DISC1) is a susceptible gene for schizophrenia (Hodgkinson et al. 2004; Millar et al. 2000; St Clair et al. 1990) implicated in neuronal development, brain maturation, and neuroplasticity (Brandon and Sawa 2011; Chubb et al. 2008). Therefore, DISC1 is a promising candidate gene for schizophrenia, but the molecular mechanisms underlying its role in the pathogenesis of the disease are still poorly understood. Interestingly, caveolin-1 (Cav-1), a cholesterol binding and scaffolding protein, regulates neuronal signal transduction and promotes neuroplasticity. In this study we examined the role of Cav-1 in mediating DISC1 expression in neurons in vitro and the hippocampus in vivo. Overexpressing Cav-1 specifically in neurons using a neuron-specific synapsin promoter (SynCav1) increased expression of DISC1 and proteins involved in synaptic plasticity (PSD95, synaptobrevin, synaptophysin, neurexin, and syntaxin 1). Similarly, SynCav1-transfected differentiated human neurons derived from induced pluripotent stem cells (hiPSCs) exhibited increased expression of DISC1 and markers of synaptic plasticity. Conversely, hippocampi from Cav-1 knockout (KO) exhibited decreased expression of DISC1 and proteins involved in synaptic plasticity. Finally, SynCav1 delivery to the hippocampus of Cav-1 KO mice and Cav-1 KO neurons in culture restored expression of DISC1 and markers of synaptic plasticity. Furthermore, we found that Cav-1 coimmunoprecipitated with DISC1 in brain tissue. These findings suggest an important role by which neuron-targeted Cav-1 regulates DISC1 neurobiology with implications for synaptic plasticity. Therefore, SynCav1 might be a potential therapeutic target for restoring neuronal function in schizophrenia. NEW & NOTEWORTHY The present study is the first to demonstrate that caveolin-1 can regulate DISC1 expression in neuronal models. Furthermore, the findings are consistent across three separate neuronal models that include rodent neurons (in vitro and in vivo) and human differentiated neurons derived from induced pluripotent stem cells. These findings justify further investigation regarding the modulatory role by caveolin on synaptic function and as a potential therapeutic target for the treatment of schizophrenia.
Collapse
Affiliation(s)
- Adam Kassan
- Department of Anesthesiology, University of California San Diego, La Jolla, California
- VA San Diego Healthcare System, San Diego, California
- Department of Psychiatry and the Sam and Rose Stein Institute for Research on Aging, University of California, San Diego, La Jolla, California
| | - Junji Egawa
- VA San Diego Healthcare System, San Diego, California
| | - Zheng Zhang
- VA San Diego Healthcare System, San Diego, California
| | - Angels Almenar-Queralt
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California; and
| | | | | | - Kaitlyn Kim
- VA San Diego Healthcare System, San Diego, California
| | | | - Dilip V Jeste
- Department of Psychiatry and the Sam and Rose Stein Institute for Research on Aging, University of California, San Diego, La Jolla, California
| | - David M Roth
- Department of Anesthesiology, University of California San Diego, La Jolla, California
- VA San Diego Healthcare System, San Diego, California
| | - Piyush M Patel
- Department of Anesthesiology, University of California San Diego, La Jolla, California
- VA San Diego Healthcare System, San Diego, California
| | - Hemal H Patel
- Department of Anesthesiology, University of California San Diego, La Jolla, California
- VA San Diego Healthcare System, San Diego, California
| | - Brian P Head
- Department of Anesthesiology, University of California San Diego, La Jolla, California;
- VA San Diego Healthcare System, San Diego, California
- Sanford Consortium for Regenerative Medicine, La Jolla, California
| |
Collapse
|
99
|
Chen CY, Liu HY, Hsueh YP. TLR3 downregulates expression of schizophrenia gene Disc1 via MYD88 to control neuronal morphology. EMBO Rep 2016; 18:169-183. [PMID: 27979975 PMCID: PMC5210159 DOI: 10.15252/embr.201642586] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 11/08/2016] [Accepted: 11/09/2016] [Indexed: 01/28/2023] Open
Abstract
Viral infection during fetal or neonatal stages increases the risk of developing neuropsychiatric disorders such as schizophrenia and autism spectrum disorders. Although neurons express several key regulators of innate immunity, the role of neuronal innate immunity in psychiatric disorders is still unclear. Using cultured neurons and in vivo mouse brain studies, we show here that Toll‐like receptor 3 (TLR3) acts through myeloid differentiation primary response gene 88 (MYD88) to negatively control Disrupted in schizophrenia 1 (Disc1) expression, resulting in impairment of neuronal development. Cytokines are not involved in TLR3‐mediated inhibition of dendrite outgrowth. Instead, TLR3 signaling suppresses expression of several psychiatric disorder‐related genes, including Disc1. The impaired dendritic arborization caused by TLR3 activation is rescued by MYD88 deficiency or DISC1 overexpression. In addition, TLR3 activation at the neonatal stage increases dendritic spine density, but narrows spine heads at postnatal day 21 (P21), suggesting a long‐lasting effect of TLR3 activation on spinogenesis. Our study reveals a novel mechanism of TLR3 in regulation of dendritic morphology and provides an explanation for how environmental factors influence mental health.
Collapse
Affiliation(s)
- Chiung-Ya Chen
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Hsin-Yu Liu
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Yi-Ping Hsueh
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
100
|
Xie Z, Li J, Baker J, Eagleson KL, Coba MP, Levitt P. Receptor Tyrosine Kinase MET Interactome and Neurodevelopmental Disorder Partners at the Developing Synapse. Biol Psychiatry 2016; 80:933-942. [PMID: 27086544 PMCID: PMC5001930 DOI: 10.1016/j.biopsych.2016.02.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Revised: 07/15/2015] [Accepted: 02/15/2016] [Indexed: 12/26/2022]
Abstract
BACKGROUND Atypical synapse development and plasticity are implicated in many neurodevelopmental disorders (NDDs). NDD-associated, high-confidence risk genes have been identified, yet little is known about functional relationships at the level of protein-protein interactions, which are the dominant molecular bases responsible for mediating circuit development. METHODS Proteomics in three independent developing neocortical synaptosomal preparations identified putative interacting proteins of the ligand-activated MET receptor tyrosine kinase, an autism risk gene that mediates synapse development. The candidates were translated into interactome networks and analyzed bioinformatically. Additionally, three independent quantitative proximity ligation assays in cultured neurons and four independent immunoprecipitation analyses of synaptosomes validated protein interactions. RESULTS Approximately 11% (8/72) of MET-interacting proteins, including SHANK3, SYNGAP1, and GRIN2B, are associated with NDDs. Proteins in the MET interactome were translated into a novel MET interactome network based on human protein-protein interaction databases. High-confidence genes from different NDD datasets that encode synaptosomal proteins were analyzed for being enriched in MET interactome proteins. This was found for autism but not schizophrenia, bipolar disorder, major depressive disorder, or attention-deficit/hyperactivity disorder. There is correlated gene expression between MET and its interactive partners in developing human temporal and visual neocortices but not with highly expressed genes that are not in the interactome. Proximity ligation assays and biochemical analyses demonstrate that MET-protein partner interactions are dynamically regulated by receptor activation. CONCLUSIONS The results provide a novel molecular framework for deciphering the functional relations of key regulators of synaptogenesis that contribute to both typical cortical development and to NDDs.
Collapse
Affiliation(s)
- Zhihui Xie
- Department of Pediatrics and The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California
| | - Jing Li
- Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Jonathan Baker
- College of Science, University of Notre Dame, South Bend, Indiana
| | - Kathie L Eagleson
- Department of Pediatrics, Children's Hospital Los Angeles and the Keck School of Medicine of the University of Southern California; Los Angeles, California
| | - Marcelo P Coba
- Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Pat Levitt
- Department of Pediatrics, Children's Hospital Los Angeles and the Keck School of Medicine of the University of Southern California; Los Angeles, California; Program in Developmental Neurogenetics, Institute for the Developing Mind and The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California.
| |
Collapse
|