1
|
Mutations in DISC1 alter IP 3R and voltage-gated Ca 2+ channel functioning, implications for major mental illness. Neuronal Signal 2021; 5:NS20180122. [PMID: 34956649 PMCID: PMC8663806 DOI: 10.1042/ns20180122] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 10/26/2021] [Accepted: 11/08/2021] [Indexed: 12/16/2022] Open
Abstract
Disrupted in Schizophrenia 1 (DISC1) participates in a wide variety of
developmental processes of central neurons. It also serves critical roles that
underlie cognitive functioning in adult central neurons. Here we summarize
DISC1’s general properties and discuss its use as a model system for
understanding major mental illnesses (MMIs). We then discuss the cellular
actions of DISC1 that involve or regulate Ca2+ signaling in adult
central neurons. In particular, we focus on the tethering role DISC1 plays in
transporting RNA particles containing Ca2+ channel subunit RNAs,
including IP3R1, CACNA1C and CACNA2D1, and in transporting mitochondria into
dendritic and axonal processes. We also review DISC1’s role in modulating
IP3R1 activity within mitochondria-associated ER membrane (MAM).
Finally, we discuss DISC1-glycogen synthase kinase 3β (GSK3β)
signaling that regulates functional expression of voltage-gated Ca2+
channels (VGCCs) at central synapses. In each case, DISC1 regulates the movement
of molecules that impact Ca2+ signaling in neurons.
Collapse
|
2
|
LIM-Kinases in Synaptic Plasticity, Memory, and Brain Diseases. Cells 2021; 10:cells10082079. [PMID: 34440848 PMCID: PMC8391678 DOI: 10.3390/cells10082079] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 07/30/2021] [Accepted: 08/04/2021] [Indexed: 12/12/2022] Open
Abstract
Learning and memory require structural and functional modifications of synaptic connections, and synaptic deficits are believed to underlie many brain disorders. The LIM-domain-containing protein kinases (LIMK1 and LIMK2) are key regulators of the actin cytoskeleton by affecting the actin-binding protein, cofilin. In addition, LIMK1 is implicated in the regulation of gene expression by interacting with the cAMP-response element-binding protein. Accumulating evidence indicates that LIMKs are critically involved in brain function and dysfunction. In this paper, we will review studies on the roles and underlying mechanisms of LIMKs in the regulation of long-term potentiation (LTP) and depression (LTD), the most extensively studied forms of long-lasting synaptic plasticity widely regarded as cellular mechanisms underlying learning and memory. We will also discuss the involvement of LIMKs in the regulation of the dendritic spine, the structural basis of synaptic plasticity, and memory formation. Finally, we will discuss recent progress on investigations of LIMKs in neurological and mental disorders, including Alzheimer’s, Parkinson’s, Williams–Beuren syndrome, schizophrenia, and autism spectrum disorders.
Collapse
|
3
|
Cuveillier C, Boulan B, Ravanello C, Denarier E, Deloulme JC, Gory-Fauré S, Delphin C, Bosc C, Arnal I, Andrieux A. Beyond Neuronal Microtubule Stabilization: MAP6 and CRMPS, Two Converging Stories. Front Mol Neurosci 2021; 14:665693. [PMID: 34025352 PMCID: PMC8131560 DOI: 10.3389/fnmol.2021.665693] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/09/2021] [Indexed: 12/21/2022] Open
Abstract
The development and function of the central nervous system rely on the microtubule (MT) and actin cytoskeletons and their respective effectors. Although the structural role of the cytoskeleton has long been acknowledged in neuronal morphology and activity, it was recently recognized to play the role of a signaling platform. Following this recognition, research into Microtubule Associated Proteins (MAPs) diversified. Indeed, historically, structural MAPs—including MAP1B, MAP2, Tau, and MAP6 (also known as STOP);—were identified and described as MT-binding and -stabilizing proteins. Extensive data obtained over the last 20 years indicated that these structural MAPs could also contribute to a variety of other molecular roles. Among multi-role MAPs, MAP6 provides a striking example illustrating the diverse molecular and cellular properties of MAPs and showing how their functional versatility contributes to the central nervous system. In this review, in addition to MAP6’s effect on microtubules, we describe its impact on the actin cytoskeleton, on neuroreceptor homeostasis, and its involvement in signaling pathways governing neuron development and maturation. We also discuss its roles in synaptic plasticity, brain connectivity, and cognitive abilities, as well as the potential relationships between the integrated brain functions of MAP6 and its molecular activities. In parallel, the Collapsin Response Mediator Proteins (CRMPs) are presented as examples of how other proteins, not initially identified as MAPs, fall into the broader MAP family. These proteins bind MTs as well as exhibiting molecular and cellular properties very similar to MAP6. Finally, we briefly summarize the multiple similarities between other classical structural MAPs and MAP6 or CRMPs.In summary, this review revisits the molecular properties and the cellular and neuronal roles of the classical MAPs, broadening our definition of what constitutes a MAP.
Collapse
|
4
|
Gory-Fauré S, Powell R, Jonckheere J, Lanté F, Denarier E, Peris L, Nguyen CH, Buisson A, Lafanechère L, Andrieux A. Pyr1-Mediated Pharmacological Inhibition of LIM Kinase Restores Synaptic Plasticity and Normal Behavior in a Mouse Model of Schizophrenia. Front Pharmacol 2021; 12:627995. [PMID: 33790791 PMCID: PMC8006432 DOI: 10.3389/fphar.2021.627995] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 02/04/2021] [Indexed: 12/14/2022] Open
Abstract
The search for effective treatments for neuropsychiatric disorders is ongoing, with progress being made as brain structure and neuronal function become clearer. The central roles played by microtubules (MT) and actin in synaptic transmission and plasticity suggest that the cytoskeleton and its modulators could be relevant targets for the development of new molecules to treat psychiatric diseases. In this context, LIM Kinase - which regulates both the actin and MT cytoskeleton especially in dendritic spines, the post-synaptic compartment of the synapse - might be a good target. In this study, we analyzed the consequences of blocking LIMK1 pharmacologically using Pyr1. We investigated synaptic plasticity defects and behavioral disorders in MAP6 KO mice, an animal model useful for the study of psychiatric disorders, particularly schizophrenia. Our results show that Pyr1 can modulate MT dynamics in neurons. In MAP6 KO mice, chronic LIMK inhibition by long-term treatment with Pyr1 can restore normal dendritic spine density and also improves long-term potentiation, both of which are altered in these mice. Pyr1 treatment improved synaptic plasticity, and also reduced social withdrawal and depressive/anxiety-like behavior in MAP6 KO mice. Overall, the results of this study validate the hypothesis that modulation of LIMK activity could represent a new therapeutic strategy for neuropsychiatric diseases.
Collapse
Affiliation(s)
- Sylvie Gory-Fauré
- Department of Molecular and Cellular Neurosciences, Grenoble Institute Neuroscience, Inserm U1216, Grenoble, France.,Université Grenoble Alpes, Grenoble, France
| | - Rebecca Powell
- Department of Molecular and Cellular Neurosciences, Grenoble Institute Neuroscience, Inserm U1216, Grenoble, France.,Université Grenoble Alpes, Grenoble, France
| | - Julie Jonckheere
- Department of Molecular and Cellular Neurosciences, Grenoble Institute Neuroscience, Inserm U1216, Grenoble, France.,Université Grenoble Alpes, Grenoble, France
| | - Fabien Lanté
- Department of Molecular and Cellular Neurosciences, Grenoble Institute Neuroscience, Inserm U1216, Grenoble, France.,Université Grenoble Alpes, Grenoble, France
| | - Eric Denarier
- Department of Molecular and Cellular Neurosciences, Grenoble Institute Neuroscience, Inserm U1216, Grenoble, France.,Université Grenoble Alpes, Grenoble, France.,Health Department, Interdisciplinary Research Institute of Grenoble, CEA, Grenoble, France
| | - Leticia Peris
- Department of Molecular and Cellular Neurosciences, Grenoble Institute Neuroscience, Inserm U1216, Grenoble, France.,Université Grenoble Alpes, Grenoble, France
| | - Chi Hung Nguyen
- Chimie et Modélisation pour la Biologie du Cancer, Institut Curie, PSL Research University, CNRS UMR9187, Inserm U1196, Orsay, France
| | - Alain Buisson
- Department of Molecular and Cellular Neurosciences, Grenoble Institute Neuroscience, Inserm U1216, Grenoble, France.,Université Grenoble Alpes, Grenoble, France
| | - Laurence Lafanechère
- Université Grenoble Alpes, Grenoble, France.,Microenvironment, Cell Plasticity and Signaling Department, Institute for Advanced Biosciences, CNRS UMR5309, Inserm U1209, Grenoble, France
| | - Annie Andrieux
- Department of Molecular and Cellular Neurosciences, Grenoble Institute Neuroscience, Inserm U1216, Grenoble, France.,Université Grenoble Alpes, Grenoble, France.,Health Department, Interdisciplinary Research Institute of Grenoble, CEA, Grenoble, France
| |
Collapse
|
5
|
Shen X, Yeung HT, Lai KO. Application of Human-Induced Pluripotent Stem Cells (hiPSCs) to Study Synaptopathy of Neurodevelopmental Disorders. Dev Neurobiol 2018; 79:20-35. [PMID: 30304570 DOI: 10.1002/dneu.22644] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 09/27/2018] [Accepted: 10/04/2018] [Indexed: 12/15/2022]
Abstract
Synapses are the basic structural and functional units for information processing and storage in the brain. Their diverse properties and functions ultimately underlie the complexity of human behavior. Proper development and maintenance of synapses are essential for normal functioning of the nervous system. Disruption in synaptogenesis and the consequent alteration in synaptic function have been strongly implicated to cause neurodevelopmental disorders such as autism spectrum disorders (ASDs) and schizophrenia (SCZ). The introduction of human-induced pluripotent stem cells (hiPSCs) provides a new path to elucidate disease mechanisms and potential therapies. In this review, we will discuss the advantages and limitations of using hiPSC-derived neurons to study synaptic disorders. Many mutations in genes encoding for proteins that regulate synaptogenesis have been identified in patients with ASDs and SCZ. We use Methyl-CpG binding protein 2 (MECP2), SH3 and multiple ankyrin repeat domains 3 (SHANK3) and Disrupted in schizophrenia 1 (DISC1) as examples to illustrate the promise of using hiPSCs as cellular models to elucidate the mechanisms underlying disease-related synaptopathy.
Collapse
Affiliation(s)
- Xuting Shen
- Faculty of Medicine, School of Biomedical Sciences, The University of Hong Kong, 21 Sassoon Road, Hong Kong, China
| | - Hoi Ting Yeung
- Faculty of Medicine, School of Biomedical Sciences, The University of Hong Kong, 21 Sassoon Road, Hong Kong, China
| | - Kwok-On Lai
- Faculty of Medicine, School of Biomedical Sciences, The University of Hong Kong, 21 Sassoon Road, Hong Kong, China.,State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, 21 Sassoon Road, Hong Kong, China
| |
Collapse
|
6
|
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
|
7
|
Muñoz-Estrada J, Lora-Castellanos A, Meza I, Alarcón Elizalde S, Benítez-King G. Primary cilia formation is diminished in schizophrenia and bipolar disorder: A possible marker for these psychiatric diseases. Schizophr Res 2018; 195:412-420. [PMID: 28927861 DOI: 10.1016/j.schres.2017.08.055] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 08/25/2017] [Accepted: 08/28/2017] [Indexed: 01/25/2023]
Abstract
Primary cilium (PC) is a microtubule-rich organelle that protrudes from the plasma membrane and acts as a cellular antenna sensing extracellular signals during brain development. DISC1 (Disrupted-in-Schizophrenia-1) is involved in PC formation and is considered a risk factor for neuropsychiatric disorders. We have previously described altered subcellular distribution of DISC1 and an aberrant microtubule organization in olfactory neuronal precursors (ONP) obtained from schizophrenia (SCZ) and bipolar disorder (BD) patients. Herein, we analyzed in vitro PC formation in healthy control subjects, SCZ and BD patients. The results indicated that 66.73±4.33% of ONP from control subjects showed immunostaining for the PC marker, acetylated α-tubulin. By contrast, only a small percentage of cells in culture from paranoid SCZ and BD patients showed PC staining (SCZ, 12.8±4.43%; BD, 12.32±5.86%). However, cells from an affected proband with disorganized SCZ and a subject with BD displayed a higher percentage of cells with cilia (SCZ, 42.20%; BD, 38.59%). Additionally, cilia elongation was observed in lithium-treated ONP derived from all groups, with a more evident response in cells from the BD group. The present study provides novel evidence that the molecular pathways involved in PC formation are defective in SCZ and BD, and impairment in these processes may be involved in the physiopathology of both diseases. Our observations also suggest that ONP is a patient-derived cell model with a potential use for diagnosis and high-throughput drug screening for brain diseases.
Collapse
Affiliation(s)
- Jesús Muñoz-Estrada
- Instituto Nacional de Psiquiatría Ramón de la Fuente Muñíz, Ciudad de México, Mexico; Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Ciudad de México, Mexico
| | | | - Isaura Meza
- Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Ciudad de México, Mexico
| | | | - Gloria Benítez-King
- Instituto Nacional de Psiquiatría Ramón de la Fuente Muñíz, Ciudad de México, Mexico.
| |
Collapse
|
8
|
Krause M, Theiss C, Brüne M. Ultrastructural Alterations of Von Economo Neurons in the Anterior Cingulate Cortex in Schizophrenia. Anat Rec (Hoboken) 2017; 300:2017-2024. [DOI: 10.1002/ar.23635] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 03/16/2017] [Accepted: 04/06/2017] [Indexed: 12/14/2022]
Affiliation(s)
- Martin Krause
- Department of Cytology, Institute of Anatomy; Ruhr-University Bochum; Bochum 44780 Germany
| | - Carsten Theiss
- Department of Cytology, Institute of Anatomy; Ruhr-University Bochum; Bochum 44780 Germany
| | - Martin Brüne
- Division of Cognitive Neuropsychiatry and Psychiatric Preventive Medicine, LWL University Hospital Bochum; Ruhr-University Bochum; Bochum 44791 Germany
| |
Collapse
|
9
|
Wang S, Chen YY, Li YP, Gu J, Gu SD, Shi H, Li XS, Lu XN, Li X, Zhang SL, Yu KJ, Liu K, Ji LL. DISC1 overexpression promotes non-small cell lung cancer cell proliferation. Oncotarget 2017; 8:65199-65210. [PMID: 29029423 PMCID: PMC5630323 DOI: 10.18632/oncotarget.18055] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 04/27/2017] [Indexed: 12/29/2022] Open
Abstract
Neuropsychiatric disorder-associated disrupted-in-schizophrenia-1 (DISC1) activates Wnt/β-catenin signaling by inhibiting glycogen synthase kinase 3 beta (GSK3β) phosphorylation, and may promote neural progenitor cell and pancreatic β-cell proliferation. The present study found that DISC1 promotes non-small cell lung cancer (NSCLC) cell growth. Western blotting and immunohistochemistry analyses showed that DISC1 was highly expressed in NSCLC cell lines and patient tissues. DISC1 expression was negatively associated with phosphorylated (p-) GSK3β, but positively correlated with a more invasive tumor phenotype and predicted poor NSCLC patient prognosis. siRNA-mediated DISC1 silencing increased p-GSK3β expression and decreased expression of β-catenin and Cyclin D1, while DISC1 upregulation produced the opposite results. DISC1 knockdown also reduced NSCLC cell proliferation rates in vitro. These results suggest that DISC1 promotes NSCLC growth, likely through GSK3β/β-catenin signaling, and that DISC1 may function as an oncogene and novel anti-NSCLC therapeutic target.
Collapse
Affiliation(s)
- Shuo Wang
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong, China.,Department of Pathology, Medical School of Nantong University, Nantong, China
| | - Ying-Ying Chen
- Department of Immunology, Medical School of Nantong University, Nantong, China.,Department of Oncology, Affiliated Hospital of Nantong University, Nantong, China
| | - Yu-Peng Li
- Department of Pediatrics, The People's Hospital of Rizhao, Rizhao, China
| | - Jun Gu
- Department of Respiratory, Affiliated Hospital of Nantong University, Nantong, China
| | - Shu-Dong Gu
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong, China
| | - Hai Shi
- Department of Cardiothoracic Surgery, The Third People's Hospital of Nantong, Nantong, China
| | - Xue-Song Li
- Department of Immunology, Medical School of Nantong University, Nantong, China
| | - Xiao-Ning Lu
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong, China.,Department of Pathology, Medical School of Nantong University, Nantong, China
| | - Xiang Li
- Department of Otorhinolaryngology, Maternal and Child Health Care Hospital of Nantong, Nantong, China
| | - Shuang-Long Zhang
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Kang-Jun Yu
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Kun Liu
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Li-Li Ji
- Department of Pathology, Medical School of Nantong University, Nantong, China
| |
Collapse
|
10
|
Trulioff A, Ermakov A, Malashichev Y. Primary Cilia as a Possible Link between Left-Right Asymmetry and Neurodevelopmental Diseases. Genes (Basel) 2017; 8:genes8020048. [PMID: 28125008 PMCID: PMC5333037 DOI: 10.3390/genes8020048] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 12/21/2016] [Accepted: 01/19/2017] [Indexed: 12/11/2022] Open
Abstract
Cilia have multiple functions in the development of the entire organism, and participate in the development and functioning of the central nervous system. In the last decade, studies have shown that they are implicated in the development of the visceral left-right asymmetry in different vertebrates. At the same time, some neuropsychiatric disorders, such as schizophrenia, autism, bipolar disorder, and dyslexia, are known to be associated with lateralization failure. In this review, we consider possible links in the mechanisms of determination of visceral asymmetry and brain lateralization, through cilia. We review the functions of seven genes associated with both cilia, and with neurodevelopmental diseases, keeping in mind their possible role in the establishment of the left-right brain asymmetry.
Collapse
Affiliation(s)
- Andrey Trulioff
- Department of Vertebrate Zoology, Faculty of Biology, Saint Petersburg State University, Universitetskaya nab., 7/9, Saint Petersburg 199034, Russia.
| | - Alexander Ermakov
- Department of Vertebrate Zoology, Faculty of Biology, Saint Petersburg State University, Universitetskaya nab., 7/9, Saint Petersburg 199034, Russia.
- Laboratory of Molecular Neurobiology, Department of Ecological Physiology, Institute of Experimental Medicine, ul. Akad. Pavlov, 12, Saint Petersburg 197376, Russia.
| | - Yegor Malashichev
- Department of Vertebrate Zoology, Faculty of Biology, Saint Petersburg State University, Universitetskaya nab., 7/9, Saint Petersburg 199034, Russia.
- Laboratory of Molecular Neurobiology, Department of Ecological Physiology, Institute of Experimental Medicine, ul. Akad. Pavlov, 12, Saint Petersburg 197376, Russia.
| |
Collapse
|
11
|
Nath S, Christian L, Tan SY, Ki S, Ehrlich LIR, Poenie M. Dynein Separately Partners with NDE1 and Dynactin To Orchestrate T Cell Focused Secretion. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 197:2090-101. [PMID: 27534551 PMCID: PMC5010990 DOI: 10.4049/jimmunol.1600180] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 07/14/2016] [Indexed: 11/19/2022]
Abstract
Helper and cytotoxic T cells accomplish focused secretion through the movement of vesicles toward the microtubule organizing center (MTOC) and translocation of the MTOC to the target contact site. In this study, using Jurkat cells and OT-I TCR transgenic primary murine CTLs, we show that the dynein-binding proteins nuclear distribution E homolog 1 (NDE1) and dynactin (as represented by p150(Glued)) form mutually exclusive complexes with dynein, exhibit nonoverlapping distributions in target-stimulated cells, and mediate different transport events. When Jurkat cells expressing a dominant negative form of NDE1 (NDE1-enhanced GFP fusion) were activated by Staphylococcus enterotoxin E-coated Raji cells, NDE1 and dynein failed to accumulate at the immunological synapse (IS) and MTOC translocation was inhibited. Knockdown of NDE1 in Jurkat cells or primary mouse CTLs also inhibited MTOC translocation and CTL-mediated killing. In contrast to NDE1, knockdown of p150(Glued), which depleted the alternative dynein/dynactin complex, resulted in impaired accumulation of CTLA4 and granzyme B-containing intracellular vesicles at the IS, whereas MTOC translocation was not affected. Depletion of p150(Glued) in CTLs also inhibited CTL-mediated lysis. We conclude that the NDE1/Lissencephaly 1 and dynactin complexes separately mediate two key components of T cell-focused secretion, namely translocation of the MTOC and lytic granules to the IS, respectively.
Collapse
Affiliation(s)
- Shubhankar Nath
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712; and
| | - Laura Christian
- Department of Molecular Cell and Developmental Biology, University of Texas at Austin, Austin, TX 78712
| | - Sarah Youngsun Tan
- Department of Molecular Cell and Developmental Biology, University of Texas at Austin, Austin, TX 78712
| | - Sanghee Ki
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712; and
| | - Lauren I R Ehrlich
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712; and
| | - Martin Poenie
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712; and
| |
Collapse
|
12
|
Shah K, Lahiri DK. A Tale of the Good and Bad: Remodeling of the Microtubule Network in the Brain by Cdk5. Mol Neurobiol 2016; 54:2255-2268. [PMID: 26944284 DOI: 10.1007/s12035-016-9792-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 02/11/2016] [Indexed: 10/22/2022]
Abstract
Cdk5, a cyclin-dependent kinase family member, is a global orchestrator of neuronal cytoskeletal dynamics. During embryogenesis, Cdk5 is indispensable for brain development. In adults, it is essential for numerous neuronal processes, including higher cognitive functions such as learning and memory formation, drug addiction, pain signaling, and long-term behavior changes through long-term potentiation and long-term depression, all of which rely on rapid alterations in the cytoskeleton. Cdk5 activity becomes deregulated in various brain disorders, including Alzheimer's disease, Parkinson's disease, Huntington's disease, attention-deficit hyperactivity disorder, epilepsy, schizophrenia, and ischemic stroke; these all result in profound remodeling of the neuronal cytoskeleton. This Commentary specifically focuses on the pleiotropic contribution of Cdk5 in regulating neuronal microtubule remodeling. Because the vast majority of the physiological substrates of Cdk5 are associated with the neuronal cytoskeleton, our emphasis is on the Cdk5 substrates, such as CRMP2, stathmin, drebrin, dixdc1, axin, MAP2, MAP1B, doublecortin, kinesin-5, and tau, that have allowed to unravel the molecular mechanisms through which Cdk5 exerts its divergent roles in regulating neuronal microtubule dynamics, both in healthy and disease states.
Collapse
Affiliation(s)
- Kavita Shah
- Department of Chemistry and Purdue University Center for Cancer Research, 560 Oval Drive, West Lafayette, IN, 47907, USA.
| | - Debomoy K Lahiri
- Departments of Psychiatry and Medical & Molecular Genetics, Institute of Psychiatric Research, Neuroscience Research Center, Indiana University School of Medicine, 320 W. 15th Street, Indianapolis, IN, 46202-2266, USA
| |
Collapse
|
13
|
Benítez-King G, Valdés-Tovar M, Trueta C, Galván-Arrieta T, Argueta J, Alarcón S, Lora-Castellanos A, Solís-Chagoyán H. The microtubular cytoskeleton of olfactory neurons derived from patients with schizophrenia or with bipolar disorder: Implications for biomarker characterization, neuronal physiology and pharmacological screening. Mol Cell Neurosci 2016; 73:84-95. [PMID: 26837043 DOI: 10.1016/j.mcn.2016.01.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 01/26/2016] [Accepted: 01/29/2016] [Indexed: 01/29/2023] Open
Abstract
Schizophrenia (SZ) and Bipolar Disorder (BD) are highly inheritable chronic mental disorders with a worldwide prevalence of around 1%. Despite that many efforts had been made to characterize biomarkers in order to allow for biological testing for their diagnoses, these disorders are currently detected and classified only by clinical appraisal based on the Diagnostic and Statistical Manual of Mental Disorders. Olfactory neuroepithelium-derived neuronal precursors have been recently proposed as a model for biomarker characterization. Because of their peripheral localization, they are amenable to collection and suitable for being cultured and propagated in vitro. Olfactory neuroepithelial cells can be obtained by a non-invasive brush-exfoliation technique from neuropsychiatric patients and healthy subjects. Neuronal precursors isolated from these samples undergo in vitro the cytoskeletal reorganization inherent to the neurodevelopment process which has been described as one important feature in the etiology of both diseases. In this paper, we will review the current knowledge on microtubular organization in olfactory neurons of patients with SZ and with BD that may constitute specific cytoskeletal endophenotypes and their relation with alterations in L-type voltage-activated Ca(2+) currents. Finally, the potential usefulness of neuronal precursors for pharmacological screening will be discussed.
Collapse
Affiliation(s)
- G Benítez-King
- Laboratorio de Neurofarmacología, Subdirección de Investigaciones Clínicas, Mexico.
| | - M Valdés-Tovar
- Laboratorio de Neurofarmacología, Subdirección de Investigaciones Clínicas, Mexico
| | - C Trueta
- Departamento de Neurofisiología, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz (INPRFM), Calzada México-Xochimilco No. 101, Col. San Lorenzo-Huipulco, C.P. 14370, Tlalpan, Distrito Federal, Mexico
| | - T Galván-Arrieta
- Laboratorio de Neurofarmacología, Subdirección de Investigaciones Clínicas, Mexico
| | - J Argueta
- Laboratorio de Neurofarmacología, Subdirección de Investigaciones Clínicas, Mexico
| | - S Alarcón
- Laboratorio de Neurofarmacología, Subdirección de Investigaciones Clínicas, Mexico
| | - A Lora-Castellanos
- Laboratorio de Neurofarmacología, Subdirección de Investigaciones Clínicas, Mexico
| | - H Solís-Chagoyán
- Laboratorio de Neurofarmacología, Subdirección de Investigaciones Clínicas, Mexico
| |
Collapse
|
14
|
Whalley HC, Dimitrova R, Sprooten E, Dauvermann MR, Romaniuk L, Duff B, Watson AR, Moorhead B, Bastin M, Semple SI, Giles S, Hall J, Thomson P, Roberts N, Hughes ZA, Brandon NJ, Dunlop J, Whitcher B, Blackwood DHR, McIntosh AM, Lawrie SM. Effects of a Balanced Translocation between Chromosomes 1 and 11 Disrupting the DISC1 Locus on White Matter Integrity. PLoS One 2015; 10:e0130900. [PMID: 26102360 PMCID: PMC4477898 DOI: 10.1371/journal.pone.0130900] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 05/25/2015] [Indexed: 11/18/2022] Open
Abstract
Objective Individuals carrying rare, but biologically informative genetic variants provide a unique opportunity to model major mental illness and inform understanding of disease mechanisms. The rarity of such variations means that their study involves small group numbers, however they are amongst the strongest known genetic risk factors for major mental illness and are likely to have large neural effects. DISC1 (Disrupted in Schizophrenia 1) is a gene containing one such risk variant, identified in a single Scottish family through its disruption by a balanced translocation of chromosomes 1 and 11; t(1;11) (q42.1;q14.3). Method Within the original pedigree, we examined the effects of the t(1;11) translocation on white matter integrity, measured by fractional anisotropy (FA). This included family members with (n = 7) and without (n = 13) the translocation, along with a clinical control sample of patients with psychosis (n = 34), and a group of healthy controls (n = 33). Results We report decreased white matter integrity in five clusters in the genu of the corpus callosum, the right inferior fronto-occipital fasciculus, acoustic radiation and fornix. Analysis of the mixed psychosis group also demonstrated decreased white matter integrity in the above regions. FA values within the corpus callosum correlated significantly with positive psychotic symptom severity. Conclusions We demonstrate that the t(1;11) translocation is associated with reduced white matter integrity in frontal commissural and association fibre tracts. These findings overlap with those shown in affected patients with psychosis and in DISC1 animal models and highlight the value of rare but biologically informative mutations in modeling psychosis.
Collapse
MESH Headings
- Adolescent
- Adult
- Bipolar Disorder/genetics
- Bipolar Disorder/pathology
- Chromosomes, Human, Pair 1/genetics
- Chromosomes, Human, Pair 1/ultrastructure
- Chromosomes, Human, Pair 11/genetics
- Chromosomes, Human, Pair 11/ultrastructure
- Corpus Callosum/pathology
- Cyclothymic Disorder/genetics
- Cyclothymic Disorder/pathology
- Depressive Disorder, Major/genetics
- Depressive Disorder, Major/pathology
- Diffusion Tensor Imaging
- Exons/genetics
- Female
- Humans
- Male
- Middle Aged
- Nerve Tissue Proteins/deficiency
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/physiology
- Schizophrenia/genetics
- Schizophrenia/pathology
- Severity of Illness Index
- Translocation, Genetic
- White Matter/pathology
- Young Adult
Collapse
Affiliation(s)
- Heather C. Whalley
- Division of Psychiatry, University of Edinburgh, Edinburgh, United Kingdom
- * E-mail:
| | - Rali Dimitrova
- Division of Psychiatry, University of Edinburgh, Edinburgh, United Kingdom
- Centre for the Developing Brain, St Thomas’ Hospital, King’s College London, London, United Kingdom
| | - Emma Sprooten
- Department of Psychiatry, Yale University, New Haven, CT, United States of America
| | - Maria R. Dauvermann
- Division of Psychiatry, University of Edinburgh, Edinburgh, United Kingdom
- McGovern Institute for Brain Research, Cambridge, MA, United States of America
| | - Liana Romaniuk
- Division of Psychiatry, University of Edinburgh, Edinburgh, United Kingdom
| | - Barbara Duff
- Division of Psychiatry, University of Edinburgh, Edinburgh, United Kingdom
| | - Andrew R. Watson
- Division of Psychiatry, University of Edinburgh, Edinburgh, United Kingdom
| | - Bill Moorhead
- Division of Psychiatry, University of Edinburgh, Edinburgh, United Kingdom
| | - Mark Bastin
- Centre for Clinical Brain Sciences, Western General Hospital, University of Edinburgh, Edinburgh, United Kingdom
| | - Scott I. Semple
- Clinical Research Imaging Centre, University of Edinburgh, Edinburgh, United Kingdom
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Stephen Giles
- Division of Psychiatry, University of Edinburgh, Edinburgh, United Kingdom
| | - Jeremy Hall
- Division of Psychiatry, University of Edinburgh, Edinburgh, United Kingdom
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Pippa Thomson
- Department of Medical Genetics, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Neil Roberts
- Clinical Research Imaging Centre, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Zoe A. Hughes
- Neuroscience Research Unit, Pfizer Inc, Cambridge, MA, United States of America
| | - Nick J. Brandon
- Neuroscience Research Unit, Pfizer Inc, Cambridge, MA, United States of America
- Current affiliation: AstraZeneca Neuroscience IMED, Cambridge, MA, United States of America
| | - John Dunlop
- Neuroscience Research Unit, Pfizer Inc, Cambridge, MA, United States of America
- Current affiliation: AstraZeneca Neuroscience IMED, Cambridge, MA, United States of America
| | - Brandon Whitcher
- Clinical and Translational Imaging, Pfizer Inc, Cambridge, MA, United States of America
| | | | - Andrew M. McIntosh
- Division of Psychiatry, University of Edinburgh, Edinburgh, United Kingdom
| | - Stephen M. Lawrie
- Division of Psychiatry, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
15
|
Zhao Z, Xu J, Chen J, Kim S, Reimers M, Bacanu SA, Yu H, Liu C, Sun J, Wang Q, Jia P, Xu F, Zhang Y, Kendler KS, Peng Z, Chen X. Transcriptome sequencing and genome-wide association analyses reveal lysosomal function and actin cytoskeleton remodeling in schizophrenia and bipolar disorder. Mol Psychiatry 2015; 20:563-572. [PMID: 25113377 PMCID: PMC4326626 DOI: 10.1038/mp.2014.82] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Revised: 05/23/2014] [Accepted: 06/17/2014] [Indexed: 12/20/2022]
Abstract
Schizophrenia (SCZ) and bipolar disorder (BPD) are severe mental disorders with high heritability. Clinicians have long noticed the similarities of clinic symptoms between these disorders. In recent years, accumulating evidence indicates some shared genetic liabilities. However, what is shared remains elusive. In this study, we conducted whole transcriptome analysis of post-mortem brain tissues (cingulate cortex) from SCZ, BPD and control subjects, and identified differentially expressed genes in these disorders. We found 105 and 153 genes differentially expressed in SCZ and BPD, respectively. By comparing the t-test scores, we found that many of the genes differentially expressed in SCZ and BPD are concordant in their expression level (q⩽0.01, 53 genes; q⩽0.05, 213 genes; q⩽0.1, 885 genes). Using genome-wide association data from the Psychiatric Genomics Consortium, we found that these differentially and concordantly expressed genes were enriched in association signals for both SCZ (P<10(-7)) and BPD (P=0.029). To our knowledge, this is the first time that a substantially large number of genes show concordant expression and association for both SCZ and BPD. Pathway analyses of these genes indicated that they are involved in the lysosome, Fc gamma receptor-mediated phagocytosis, regulation of actin cytoskeleton pathways, along with several cancer pathways. Functional analyses of these genes revealed an interconnected pathway network centered on lysosomal function and the regulation of actin cytoskeleton. These pathways and their interacting network were principally confirmed by an independent transcriptome sequencing data set of the hippocampus. Dysregulation of lysosomal function and cytoskeleton remodeling has direct impacts on endocytosis, phagocytosis, exocytosis, vesicle trafficking, neuronal maturation and migration, neurite outgrowth and synaptic density and plasticity, and different aspects of these processes have been implicated in SCZ and BPD.
Collapse
Affiliation(s)
- Zhongming Zhao
- Departments of Biomedical Informatics and Psychiatry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Jiabao Xu
- Beijing Genomics Institute (BGI), Shenzhen, Guangdong, 518083, China
| | - Jingchun Chen
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Sanghyeon Kim
- Stanley Laboratory of Brain Research, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Mark Reimers
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Silviu-Alin Bacanu
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Hui Yu
- Departments of Biomedical Informatics and Psychiatry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Chunyu Liu
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL 60637, USA
| | - Jingchun Sun
- Departments of Biomedical Informatics and Psychiatry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Quan Wang
- Departments of Biomedical Informatics and Psychiatry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Peilin Jia
- Departments of Biomedical Informatics and Psychiatry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Fengping Xu
- Beijing Genomics Institute (BGI), Shenzhen, Guangdong, 518083, China
| | - Yong Zhang
- Beijing Genomics Institute (BGI), Shenzhen, Guangdong, 518083, China
| | - Kenneth S. Kendler
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Zhiyu Peng
- Beijing Genomics Institute (BGI), Shenzhen, Guangdong, 518083, China
| | - Xiangning Chen
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA 23298, USA
| |
Collapse
|
16
|
Muñoz-Estrada J, Benítez-King G, Berlanga C, Meza I. Altered subcellular distribution of the 75-kDa DISC1 isoform, cAMP accumulation, and decreased neuronal migration in schizophrenia and bipolar disorder: implications for neurodevelopment. CNS Neurosci Ther 2015; 21:446-53. [PMID: 25620115 DOI: 10.1111/cns.12377] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 12/09/2014] [Accepted: 12/09/2014] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND DISC1 (Disrupted-In-Schizophrenia-1) is considered a genetic risk factor for schizophrenia (SZ) and bipolar disorder (BD). DISC1 regulates microtubule stability, migration, and cAMP signaling in mammalian cell lines and mouse brain tissue. cAMP is a regulator of microtubule organization and migration in neurons. Aberrant microtubule organization has been observed in olfactory neuronal precursors (ONP) derived from patients with SZ and BD, which suggests involvement of DISC1 and cAMP. However, the biology of DISC1 in the physiopathology of psychiatric conditions remains elusive. AIMS Herein, utilizing ONP obtained from SZ, BD patients and healthy subjects, we have studied DISC1 expression, protein levels, and subcellular distribution by qRT-PCR, immunoblotting, subcellular fractionation, and confocal microscopy. Cell migration and cAMP accumulation were assessed by Transwell and PKA competition assays. RESULTS We found increased levels of the 75-kDa DISC1 isoform in total cell extracts of ONP from patients with SZ and BD compared with controls. Subcellular distribution showed a significant decrease of cytoplasmic DISC1 concomitant with its augmented levels in transcription sites. Moreover, significant cAMP accumulation and diminished migration were also observed in patients' cells. CONCLUSION Alterations of DISC1 levels and its cellular distribution, which negatively modify cAMP homeostasis, microtubule organization, and cell migration, in ONP from patients with SZ and BD, suggest that their presence in early stages of brain development may impact brain maturation and function.
Collapse
Affiliation(s)
- Jesús Muñoz-Estrada
- Department of Molecular Biomedicine, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico, Mexico; Laboratory of Neuropharmacology, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico, Mexico
| | | | | | | |
Collapse
|
17
|
Steinecke A, Gampe C, Nitzsche F, Bolz J. DISC1 knockdown impairs the tangential migration of cortical interneurons by affecting the actin cytoskeleton. Front Cell Neurosci 2014; 8:190. [PMID: 25071449 PMCID: PMC4086047 DOI: 10.3389/fncel.2014.00190] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 06/20/2014] [Indexed: 12/29/2022] Open
Abstract
Disrupted-in-Schizophrenia 1 (DISC1) is a risk gene for a spectrum of major mental disorders. It has been shown to regulate radial migration as well as dendritic arborization during neurodevelopment and corticogenesis. In a previous study we demonstrated through in vitro experiments that DISC1 also controls the tangential migration of cortical interneurons originating from the medial ganglionic eminence (MGE). Here we first show that DISC1 is necessary for the proper tangential migration of cortical interneurons in the intact brain. Expression of EGFP under the Lhx6 promotor allowed us to analyze exclusively interneurons transfected in the MGE after in utero electroporation. After 3 days in utero, DISC1 deficient interneurons displayed prolonged leading processes and, compared to control, fewer neurons reached the cortex. Time-lapse video microscopy of cortical feeder-layers revealed a decreased migration velocity due to a reduction of soma translocations. Immunostainings indicated that DISC1 is co-localized with F-actin in the growth cone-like structure of the leading process. DISC1 knockdown reduced F-actin levels whereas the overall actin level was not altered. Moreover, DISC1 knockdown also decreased levels of phosphorylated Girdin, which cross-links F-actin, as well as the Girdin-activator pAkt. In contrast, using time-lapse video microscopy of fluorescence-tagged tubulin and EB3 in fibroblasts, we found no effects on microtubule polymerization when DISC1 was reduced. However, DISC1 affected the acetylation of microtubules in the leading processes of MGE-derived cortical interneurons. Together, our results provide a mechanism how DISC1 might contribute to interneuron migration thereby explaining the reduced number of specific classes of cortical interneurons in some DISC1 mouse models.
Collapse
Affiliation(s)
- André Steinecke
- Universität Jena, Institut für Allgemeine Zoologie und Tierphysiologie Jena, Germany
| | - Christin Gampe
- Universität Jena, Institut für Allgemeine Zoologie und Tierphysiologie Jena, Germany
| | - Falk Nitzsche
- Universität Jena, Institut für Allgemeine Zoologie und Tierphysiologie Jena, Germany
| | - Jürgen Bolz
- Universität Jena, Institut für Allgemeine Zoologie und Tierphysiologie Jena, Germany
| |
Collapse
|
18
|
Ratta-Apha W, Hishimoto A, Mouri K, Shiroiwa K, Sasada T, Yoshida M, Supriyanto I, Ueno Y, Asano M, Shirakawa O, Togashi H, Takai Y, Sora I. Association analysis of the DISC1 gene with schizophrenia in the Japanese population and DISC1 immunoreactivity in the postmortem brain. Neurosci Res 2013; 77:222-7. [PMID: 24013095 DOI: 10.1016/j.neures.2013.08.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Revised: 08/24/2013] [Accepted: 08/26/2013] [Indexed: 12/11/2022]
Abstract
The Disrupted-in-Schizophrenia 1 (DISC1) gene plays a role in the regulation of neural development. Previous evidence from genetic association and biological studies implicates the DISC1 gene as having a role in the pathophysiology of schizophrenia. In the present study, we explored the association between DISC1 missense mutation rs821616 (Ser704Cys) single nucleotide polymorphism (SNP) and four other SNPs (rs1772702, rs1754603, rs821621, rs821624) in the related haplotype block and schizophrenia in the Japanese population. We could not find a significant association of selected SNPs with schizophrenia after correction for multiple testing. We performed a meta-analysis of the Ser704Cys variant in schizophrenia using data from the present study and five previous Japanese population studies, and found no association with schizophrenia. We also examined DISC1 immunoreactivity in postmortem prefrontal cortex specimens of schizophrenia patients compared to control samples. The immunoreactivity revealed a significant decrease of DISC1 protein expression in the schizophrenia samples after ruling out potential confounding factors. However, the Ser704Cys variant did not show effects on DISC1 immunoreactivity. These results provide evidence that this functional genetic variation of DISC1 do not underlie the pathophysiology of schizophrenia in the Japanese population.
Collapse
Affiliation(s)
- Woraphat Ratta-Apha
- Department of Psychiatry, Kobe University Graduate School of Medicine, Kobe, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Goudarzi S, Smith LJM, Schütz S, Hafizi S. Interaction of DISC1 with the PTB domain of Tensin2. Cell Mol Life Sci 2013; 70:1663-72. [PMID: 23233134 PMCID: PMC11113815 DOI: 10.1007/s00018-012-1228-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Revised: 11/08/2012] [Accepted: 11/29/2012] [Indexed: 12/27/2022]
Abstract
The gene for Disrupted-in-Schizophrenia 1 (DISC1) is amongst the most significant risk genes for schizophrenia. The DISC1 protein is an intracellular scaffolding molecule thought to act an important hub for protein interactions involved in signalling for neural cell differentiation and function. Tensin2 is an intracellular actin-binding protein that bridges the intracellular portion of transmembrane receptors to the cytoskeleton, thereby regulating signalling for cell shape and motility. In this study, we probed in molecular detail a novel interaction between DISC1 and Tensin2. Western blot and confocal microscopic analyses revealed widespread expression of both DISC1 and Tensin2 proteins throughout the mouse brain. Furthermore, we have developed novel anti-DISC1 antibodies that verified the predominant expression of a 105-kDa isoform of DISC1 in the rodent brain as well as in human cells. In the mouse brain, both proteins showed region-specific expression patterns, including strong expression in the pyramidal cell layer of the hippocampus and dentate gyrus. DISC1-Tensin2 colocalisation was most clearly observed in the Purkinje cells of the mouse cerebellum. Biochemical coimmunoprecipitation experiments revealed an interaction between endogenous DISC1 and Tensin2 proteins in the mouse brain. Further pulldown studies in human cells using Myc-tagged Tensin2 constructs revealed that DISC1 specifically interacts with the C-terminal PTB domain of Tensin2 in a phosphorylation-independent manner. This new knowledge on the DISC1-Tensin2 interaction, as part of the wider DISC1 interactome, should further elucidate the signalling pathways that are perturbed in schizophrenia and other mental disorders.
Collapse
Affiliation(s)
- Salman Goudarzi
- Institute of Biomedical and Biomolecular Science (IBBS), University of Portsmouth, Portsmouth, PO1 2DT UK
| | - Luke J. M. Smith
- Institute of Biomedical and Biomolecular Science (IBBS), University of Portsmouth, Portsmouth, PO1 2DT UK
| | - Steffen Schütz
- Institute of Biomedical and Biomolecular Science (IBBS), University of Portsmouth, Portsmouth, PO1 2DT UK
| | - Sassan Hafizi
- Institute of Biomedical and Biomolecular Science (IBBS), University of Portsmouth, Portsmouth, PO1 2DT UK
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, St. Michael’s Building, White Swan Road, Portsmouth, PO1 2DT UK
| |
Collapse
|
20
|
Lepagnol-Bestel AM, Kvajo M, Karayiorgou M, Simonneau M, Gogos JA. A Disc1 mutation differentially affects neurites and spines in hippocampal and cortical neurons. Mol Cell Neurosci 2013; 54:84-92. [PMID: 23396153 DOI: 10.1016/j.mcn.2013.01.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Revised: 01/21/2013] [Accepted: 01/24/2013] [Indexed: 02/05/2023] Open
Abstract
A balanced chromosomal translocation segregating with schizophrenia and affective disorders in a large Scottish family disrupting DISC1 implicated this gene as a susceptibility gene for major mental illness. Here we study neurons derived from a genetically engineered mouse strain with a truncating lesion disrupting the endogenous Disc1 ortholog. We provide a detailed account of the consequences of this mutation on axonal and dendritic morphogenesis as well as dendritic spine development in cultured hippocampal and cortical neurons. We show that the mutation has distinct effects on these two types of neurons, supporting a cell-type specific role of Disc1 in establishing structural connections among neurons. Moreover, using a validated antibody we provide evidence indicating that Disc1 localizes primarily to Golgi apparatus-related vesicles. Our results support the notion that in vitro cultures derived from Disc1(Tm1Kara) mice provide a valuable model for future mechanistic analysis of the cellular and biochemical effects of this mutation, and can thus serve as a platform for drug discovery efforts.
Collapse
Affiliation(s)
- A M Lepagnol-Bestel
- Department of Physiology and Cellular Biophysics, Columbia University, 630 West, 168th Street, New York, NY 10032, USA
| | | | | | | | | |
Collapse
|
21
|
GABA through the ages: regulation of cortical function and plasticity by inhibitory interneurons. Neural Plast 2012; 2012:892784. [PMID: 22792496 PMCID: PMC3390141 DOI: 10.1155/2012/892784] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Revised: 03/30/2012] [Accepted: 03/30/2012] [Indexed: 11/17/2022] Open
Abstract
Inhibitory interneurons comprise only about 20% of cortical neurons and thus constitute a clear minority compared to the vast number of excitatory projection neurons. They are, however, an influential minority with important roles in cortical maturation, function, and plasticity. In this paper, we will highlight the functional importance of cortical inhibition throughout brain development, starting with the embryonal formation of the cortex, proceeding by the regulation of sensory cortical plasticity in adulthood, and finishing with the GABA involvement in sensory information processing in old age.
Collapse
|
22
|
Wei Q, Diao F, Kang Z, Gan Z, Han Z, Zheng L, Li L, Guo X, Shan B, Liu C, Zhao J, Zhang J. The effect of DISC1 on regional gray matter density of schizophrenia in Han Chinese population. Neurosci Lett 2012; 517:21-4. [PMID: 22516458 DOI: 10.1016/j.neulet.2012.03.098] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Revised: 03/30/2012] [Accepted: 03/31/2012] [Indexed: 12/23/2022]
Abstract
Schizophrenia is thought to arise in part from abnormal gray matter (GM), which are partly shared by the relatives of the probands. DISC1 is one of the most promising susceptibility genes of schizophrenia and a SNP rs821597 (A) in the gene was associated with schizophrenia in Han Chinese population. In this study, 61 healthy controls and 72 with schizophrenic patients were genotyped at rs821597, and underwent T1-weighted MRI for the density of GM. The results showed that the risk allele (A) carriers had higher GM density in regional left parahippocampal gyrus and right orbitofrontal cortex in schizophrenic patients, but had reduced GM density of these brain regions in healthy controls. The DISC1 variant rs821597 may confer risk for schizophrenia by its effects on the regional GM in left parahippocampal gyrus and right orbitofrontal cortex with other risk factors for schizophrenia.
Collapse
Affiliation(s)
- Qinling Wei
- Psychiatry Department, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, PR China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
McLinden KA, Trunova S, Giniger E. At the Fulcrum in Health and Disease: Cdk5 and the Balancing Acts of Neuronal Structure and Physiology. ACTA ACUST UNITED AC 2012; 2012:001. [PMID: 25364642 PMCID: PMC4212508 DOI: 10.4172/2168-975x.s1-001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cdk5 has been implicated in a multitude of processes in neuronal development, cell biology and physiology. These influence many neurological disorders, but the very breadth of Cdk5 effects has made it difficult to synthesize a coherent picture of the part played by this protein in health and disease. In this review, we focus on the roles of Cdk5 in neuronal function, particularly synaptic homeostasis, plasticity, neurotransmission, subcellular organization, and trafficking. We then discuss how disruption of these Cdk5 activities may initiate or exacerbate neural disorders. A recurring theme will be the sensitivity of Cdk5 sequelae to the precise biological context under consideration.
Collapse
Affiliation(s)
- Kristina A McLinden
- National Institute of Neurological Disorders and Stroke, USA ; National Human Genome Research Institute, USA
| | - Svetlana Trunova
- National Institute of Neurological Disorders and Stroke, USA ; National Human Genome Research Institute, USA
| | - Edward Giniger
- National Institute of Neurological Disorders and Stroke, USA ; National Human Genome Research Institute, USA
| |
Collapse
|
24
|
Brandon NJ, Sawa A. Linking neurodevelopmental and synaptic theories of mental illness through DISC1. Nat Rev Neurosci 2011; 12:707-22. [PMID: 22095064 DOI: 10.1038/nrn3120] [Citation(s) in RCA: 331] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Recent advances in our understanding of the underlying genetic architecture of psychiatric disorders has blown away the diagnostic boundaries that are defined by currently used diagnostic manuals. The disrupted in schizophrenia 1 (DISC1) gene was originally discovered at the breakpoint of an inherited chromosomal translocation, which segregates with major mental illnesses. In addition, many biological studies have indicated a role for DISC1 in early neurodevelopment and synaptic regulation. Given that DISC1 is thought to drive a range of endophenotypes that underlie major mental conditions, elucidating the biology of DISC1 may enable the construction of new diagnostic categories for mental illnesses with a more meaningful biological foundation.
Collapse
|