1
|
Zhang J, Kaur G, Cai E, Gutierrez OT, Liu X, Baboo S, Diedrich JK, Zhu JF, Myers BR, Yates JR, Ge X. Proximity based proteomics reveals Git1 as a regulator of Smoothened signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.06.631593. [PMID: 39829937 PMCID: PMC11741329 DOI: 10.1101/2025.01.06.631593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
The GPCR-like protein Smoothened (Smo) plays a pivotal role in the Hedgehog (Hh) pathway. To initiate Hh signaling, active Smo binds to and inhibits the catalytic subunit of PKA in the primary cilium, a process facilitated by G protein-coupled receptor kinase 2 (Grk2). However, the precise regulatory mechanisms underlying this process, as well as the events preceding and following Smo activation, remain poorly understood. To address this question, we leveraged the proximity labeling tool TurboID and conducted a time-resolved proteomic study of Smo-associated proteins over the course of Hh signaling activation. Our results not only confirmed previously reported Smo interactors but also uncovered new Smo-associated proteins. We characterized one of these new Smo interactors, Grk-interacting protein 1 (Git1), previously known to modulate GPCR signaling. We found that Git1 localizes to the base of the primary cilium, where it controls the cilium transport of Grk2, an early event in Hh signaling. Loss of Git1 impairs Smo phosphorylation by Grk2, a critical step for Smo-PKA interaction, leading to attenuated Hh signaling and reduced cell proliferation in granule neuron precursors. These results revealed a critical regulatory mechanism of Grk2 phosphorylation on Smo in the primary cilium. Our Smo-TurboID proteomic dataset provides a unique resource for investigating Smo regulations across different stages of Hh pathway activation.
Collapse
Affiliation(s)
- Jingyi Zhang
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California Merced, Merced, California, USA
| | - Gurleen Kaur
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California Merced, Merced, California, USA
| | - Eva Cai
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California Merced, Merced, California, USA
| | - Oscar Torres Gutierrez
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California Merced, Merced, California, USA
| | - Xiaoliang Liu
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California Merced, Merced, California, USA
| | - Sabyasachi Baboo
- Departments of Molecular Medicine and Neurobiology, the Scripps Research Institute, San Diego, California, USA
| | - Jolene K Diedrich
- Departments of Molecular Medicine and Neurobiology, the Scripps Research Institute, San Diego, California, USA
| | - Ju-Fen Zhu
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah, USA
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, Utah, USA
- Department of Bioengineering, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Benjamin R. Myers
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah, USA
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, Utah, USA
- Department of Bioengineering, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - John R Yates
- Departments of Molecular Medicine and Neurobiology, the Scripps Research Institute, San Diego, California, USA
| | - Xuecai Ge
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California Merced, Merced, California, USA
| |
Collapse
|
2
|
Fass DM, Lewis MC, Ahmad R, Szucs MJ, Zhang Q, Fleishman M, Wang D, Kim MJ, Biag J, Carr SA, Scolnick EM, Premont RT, Haggarty SJ. Brain-specific deletion of GIT1 impairs cognition and alters phosphorylation of synaptic protein networks implicated in schizophrenia susceptibility. Mol Psychiatry 2022; 27:3272-3285. [PMID: 35505090 PMCID: PMC9630168 DOI: 10.1038/s41380-022-01557-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 03/18/2022] [Accepted: 03/29/2022] [Indexed: 11/09/2022]
Abstract
Despite tremendous effort, the molecular and cellular basis of cognitive deficits in schizophrenia remain poorly understood. Recent progress in elucidating the genetic architecture of schizophrenia has highlighted the association of multiple loci and rare variants that may impact susceptibility. One key example, given their potential etiopathogenic and therapeutic relevance, is a set of genes that encode proteins that regulate excitatory glutamatergic synapses in brain. A critical next step is to delineate specifically how such genetic variation impacts synaptic plasticity and to determine if and how the encoded proteins interact biochemically with one another to control cognitive function in a convergent manner. Towards this goal, here we study the roles of GPCR-kinase interacting protein 1 (GIT1), a synaptic scaffolding and signaling protein with damaging coding variants found in schizophrenia patients, as well as copy number variants found in patients with neurodevelopmental disorders. We generated conditional neural-selective GIT1 knockout mice and found that these mice have deficits in fear conditioning memory recall and spatial memory, as well as reduced cortical neuron dendritic spine density. Using global quantitative phospho-proteomics, we revealed that GIT1 deletion in brain perturbs specific networks of GIT1-interacting synaptic proteins. Importantly, several schizophrenia and neurodevelopmental disorder risk genes are present within these networks. We propose that GIT1 regulates the phosphorylation of a network of synaptic proteins and other critical regulators of neuroplasticity, and that perturbation of these networks may contribute specifically to cognitive deficits observed in schizophrenia and neurodevelopmental disorders.
Collapse
Affiliation(s)
- Daniel M. Fass
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, 75 Ames Street, Cambridge, Massachusetts 02142, USA,Chemical Neurobiology Laboratory, Center for Genomic Medicine, Departments of Neurology & Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Michael C. Lewis
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, 75 Ames Street, Cambridge, Massachusetts 02142, USA,Sage Therapeutics, Cambridge, MA, USA
| | - Rushdy Ahmad
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA, 02142, USA,Wyss Institute at Harvard University, Boston, MA, USA
| | - Matthew J. Szucs
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA, 02142, USA,Department of Biochemistry and Molecular Genetics, University of Colorado Denver School of Medicine, Aurora, Colorado, USA
| | - Qiangge Zhang
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Morgan Fleishman
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, 75 Ames Street, Cambridge, Massachusetts 02142, USA,McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Dongqing Wang
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Myung Jong Kim
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, 75 Ames Street, Cambridge, Massachusetts 02142, USA,Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Jonathan Biag
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, 75 Ames Street, Cambridge, Massachusetts 02142, USA,Novartis Pharmaceuticals, Cambridge, MA, USA
| | - Steven A. Carr
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA, 02142, USA
| | - Edward M. Scolnick
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, 75 Ames Street, Cambridge, Massachusetts 02142, USA,Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA, 02142, USA
| | - Richard T. Premont
- Harrington Discovery Institute, Cleveland, OH, 44106, USA; Institute for Transformative Molecular Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Stephen J. Haggarty
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, 75 Ames Street, Cambridge, Massachusetts 02142, USA,Chemical Neurobiology Laboratory, Center for Genomic Medicine, Departments of Neurology & Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, USA
| |
Collapse
|
3
|
Abstract
Immunity could be viewed as the common factor in neurodevelopmental disorders and cancer. The immune and nervous systems coevolve as the embryo develops. Immunity can release cytokines that activate MAPK signaling in neural cells. In specific embryonic brain cell types, dysregulated signaling that results from germline or embryonic mutations can promote changes in chromatin organization and gene accessibility, and thus expression levels of essential genes in neurodevelopment. In cancer, dysregulated signaling can emerge from sporadic somatic mutations during human life. Neurodevelopmental disorders and cancer share similarities. In neurodevelopmental disorders, immunity, and cancer, there appears an almost invariable involvement of small GTPases (e.g., Ras, RhoA, and Rac) and their pathways. TLRs, IL-1, GIT1, and FGFR signaling pathways, all can be dysregulated in neurodevelopmental disorders and cancer. Although there are signaling similarities, decisive differentiating factors are timing windows, and cell type specific perturbation levels, pointing to chromatin reorganization. Finally, we discuss drug discovery.
Collapse
Affiliation(s)
- Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD 21702, USA
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
- Corresponding author
| | - Chung-Jung Tsai
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD 21702, USA
| | - Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD 21702, USA
| |
Collapse
|
4
|
González-Mancha N, Rodríguez-Rodríguez C, Alcover A, Merida I. Sorting Nexin 27 Enables MTOC and Secretory Machinery Translocation to the Immune Synapse. Front Immunol 2022; 12:814570. [PMID: 35095913 PMCID: PMC8790036 DOI: 10.3389/fimmu.2021.814570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 12/20/2021] [Indexed: 11/24/2022] Open
Abstract
Sorting nexin 27 (SNX27) association to the retromer complex mediates intracellular trafficking of cargoes containing PSD95/Dlg1/ZO-1 (PDZ)-binding C-terminal sequences from endosomes to the cell surface, preventing their lysosomal degradation. Antigen recognition by T lymphocyte leads to the formation of a highly organized structure named the immune synapse (IS), which ensures cell-cell communication and sustained T cell activation. At the neuronal synapse, SNX27 recycles PDZ-binding receptors and its defective expression is associated with synaptic dysfunction and cognitive impairment. In T lymphocytes, SNX27 was found localized at recycling endosomal compartments that polarized to the IS, suggesting a function in polarized traffic to this structure. Proteomic analysis of PDZ-SNX27 interactors during IS formation identify proteins with known functions in cytoskeletal reorganization and lipid regulation, such as diacylglycerol (DAG) kinase (DGK) ζ, as well as components of the retromer and WASH complex. In this study, we investigated the consequences of SNX27 deficiency in cytoskeletal reorganization during IS formation. Our analyses demonstrate that SNX27 controls the polarization towards the cell-cell interface of the PDZ-interacting cargoes DGKζ and the retromer subunit vacuolar protein sorting protein 26, among others. SNX27 silencing abolishes the formation of a DAG gradient at the IS and prevents re-localization of the dynactin complex component dynactin-1/p150Glued, two events that correlate with impaired microtubule organizing center translocation (MTOC). SNX27 silenced cells show marked alteration in cytoskeleton organization including a failure in the organization of the microtubule network and defects in actin clearance at the IS. Reduced SNX27 expression was also found to hinder the arrangement of signaling microclusters at the IS, as well as the polarization of the secretory machinery towards the antigen presenting cells. Our results broaden the knowledge of SNX27 function in T lymphocytes by showing a function in modulating IS organization through regulated trafficking of cargoes.
Collapse
Affiliation(s)
- Natalia González-Mancha
- Department of Immunology and Oncology, Centro Nacional de Biotecnología-Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Cristina Rodríguez-Rodríguez
- Department of Immunology and Oncology, Centro Nacional de Biotecnología-Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Andrés Alcover
- Institut Pasteur, Université de Paris, Unité Biologie Cellulaire des Lymphocytes, INSERM U1224, Ligue Nationale Contre le Cancer, Équipe Labellisée Ligue-2018, Paris, France
| | - Isabel Merida
- Department of Immunology and Oncology, Centro Nacional de Biotecnología-Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| |
Collapse
|
5
|
Conde-Dusman MJ, Dey PN, Elía-Zudaire Ó, Rabaneda LG, García-Lira C, Grand T, Briz V, Velasco ER, Andero R, Niñerola S, Barco A, Paoletti P, Wesseling JF, Gardoni F, Tavalin SJ, Perez-Otaño I. Control of protein synthesis and memory by GluN3A-NMDA receptors through inhibition of GIT1/mTORC1 assembly. eLife 2021; 10:e71575. [PMID: 34787081 PMCID: PMC8598234 DOI: 10.7554/elife.71575] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 10/13/2021] [Indexed: 12/03/2022] Open
Abstract
De novo protein synthesis is required for synapse modifications underlying stable memory encoding. Yet neurons are highly compartmentalized cells and how protein synthesis can be regulated at the synapse level is unknown. Here, we characterize neuronal signaling complexes formed by the postsynaptic scaffold GIT1, the mechanistic target of rapamycin (mTOR) kinase, and Raptor that couple synaptic stimuli to mTOR-dependent protein synthesis; and identify NMDA receptors containing GluN3A subunits as key negative regulators of GIT1 binding to mTOR. Disruption of GIT1/mTOR complexes by enhancing GluN3A expression or silencing GIT1 inhibits synaptic mTOR activation and restricts the mTOR-dependent translation of specific activity-regulated mRNAs. Conversely, GluN3A removal enables complex formation, potentiates mTOR-dependent protein synthesis, and facilitates the consolidation of associative and spatial memories in mice. The memory enhancement becomes evident with light or spaced training, can be achieved by selectively deleting GluN3A from excitatory neurons during adulthood, and does not compromise other aspects of cognition such as memory flexibility or extinction. Our findings provide mechanistic insight into synaptic translational control and reveal a potentially selective target for cognitive enhancement.
Collapse
Affiliation(s)
- María J Conde-Dusman
- lnstituto de Neurociencias (UMH-CSIC)AlicanteSpain
- Centro de Investigación Médica Aplicada (CIMA), University of NavarraPamplonaSpain
- Centre for Developmental Neurobiology, Institute of Psychiatry, King’s College LondonLondonUnited Kingdom
| | - Partha N Dey
- Centro de Investigación Médica Aplicada (CIMA), University of NavarraPamplonaSpain
- National Eye Institute, National Institutes of HealthBethesdaUnited States
| | | | - Luis G Rabaneda
- lnstituto de Neurociencias (UMH-CSIC)AlicanteSpain
- Centro de Investigación Médica Aplicada (CIMA), University of NavarraPamplonaSpain
- Institute of Science and Technology AustriaKlosterneuburgAustria
| | | | - Teddy Grand
- Institut de Biologie de l’Ecole Normale Supérieure/CNRS/INSERMParisFrance
| | - Victor Briz
- Centro de Biología Molecular Severo Ochoa (UAM-CSIC)MadridSpain
| | - Eric R Velasco
- Institut de Neurociències, Universitat Autònoma de BarcelonaBellaterraSpain
| | - Raül Andero
- Institut de Neurociències, Departament de Psicobiologia i de Metodologia de les Ciències de la Salut, Unitat de Neurociència Traslacional, Parc Taulí Hospital Universitari, Institut d’Investigació i Innovació Parc Taulí (I3PT), Universitat Autònoma de BarcelonaBellaterraSpain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos IIIMadridSpain
- ICREABarcelonaSpain
| | | | - Angel Barco
- lnstituto de Neurociencias (UMH-CSIC)AlicanteSpain
| | - Pierre Paoletti
- Institut de Biologie de l’Ecole Normale Supérieure/CNRS/INSERMParisFrance
| | | | - Fabrizio Gardoni
- Department of Pharmacological and Biomolecular Sciences, University of MilanMilanItaly
| | - Steven J Tavalin
- Department of Pharmacology, Addiction Science, and Toxicology, University of Tennessee Health Science CenterMemphisUnited States
| | - Isabel Perez-Otaño
- lnstituto de Neurociencias (UMH-CSIC)AlicanteSpain
- Centro de Investigación Médica Aplicada (CIMA), University of NavarraPamplonaSpain
| |
Collapse
|
6
|
Yuval O, Iosilevskii Y, Meledin A, Podbilewicz B, Shemesh T. Neuron tracing and quantitative analyses of dendritic architecture reveal symmetrical three-way-junctions and phenotypes of git-1 in C. elegans. PLoS Comput Biol 2021; 17:e1009185. [PMID: 34280180 PMCID: PMC8321406 DOI: 10.1371/journal.pcbi.1009185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 07/29/2021] [Accepted: 06/15/2021] [Indexed: 11/18/2022] Open
Abstract
Complex dendritic trees are a distinctive feature of neurons. Alterations to dendritic morphology are associated with developmental, behavioral and neurodegenerative changes. The highly-arborized PVD neuron of C. elegans serves as a model to study dendritic patterning; however, quantitative, objective and automated analyses of PVD morphology are missing. Here, we present a method for neuronal feature extraction, based on deep-learning and fitting algorithms. The extracted neuronal architecture is represented by a database of structural elements for abstracted analysis. We obtain excellent automatic tracing of PVD trees and uncover that dendritic junctions are unevenly distributed. Surprisingly, these junctions are three-way-symmetrical on average, while dendritic processes are arranged orthogonally. We quantify the effect of mutation in git-1, a regulator of dendritic spine formation, on PVD morphology and discover a localized reduction in junctions. Our findings shed new light on PVD architecture, demonstrating the effectiveness of our objective analyses of dendritic morphology and suggest molecular control mechanisms.
Collapse
Affiliation(s)
- Omer Yuval
- Faculty of Biology, Technion–Israel Institute of Technology, Haifa, Israel
- School of Computing, Faculty of Engineering and Physical Sciences, University of Leeds, Leeds, United Kingdom
| | - Yael Iosilevskii
- Faculty of Biology, Technion–Israel Institute of Technology, Haifa, Israel
| | - Anna Meledin
- Faculty of Biology, Technion–Israel Institute of Technology, Haifa, Israel
| | | | - Tom Shemesh
- Faculty of Biology, Technion–Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
7
|
Reynolds N, Aceves NM, Liu JL, Compton JR, Leary DH, Freitas BT, Pegan SD, Doctor KZ, Wu FY, Hu X, Legler PM. The SARS-CoV-2 SSHHPS Recognized by the Papain-like Protease. ACS Infect Dis 2021; 7:1483-1502. [PMID: 34019767 PMCID: PMC8171221 DOI: 10.1021/acsinfecdis.0c00866] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Indexed: 12/16/2022]
Abstract
Viral proteases are highly specific and recognize conserved cleavage site sequences of ∼6-8 amino acids. Short stretches of homologous host-pathogen sequences (SSHHPS) can be found spanning the viral protease cleavage sites. We hypothesized that these sequences corresponded to specific host protein targets since >40 host proteins have been shown to be cleaved by Group IV viral proteases and one Group VI viral protease. Using PHI-BLAST and the viral protease cleavage site sequences, we searched the human proteome for host targets and analyzed the hit results. Although the polyprotein and host proteins related to the suppression of the innate immune responses may be the primary targets of these viral proteases, we identified other cleavable host proteins. These proteins appear to be related to the virus-induced phenotype associated with Group IV viruses, suggesting that information about viral pathogenesis may be extractable directly from the viral genome sequence. Here we identify sequences cleaved by the SARS-CoV-2 papain-like protease (PLpro) in vitro within human MYH7 and MYH6 (two cardiac myosins linked to several cardiomyopathies), FOXP3 (an X-linked Treg cell transcription factor), ErbB4 (HER4), and vitamin-K-dependent plasma protein S (PROS1), an anticoagulation protein that prevents blood clots. Zinc inhibited the cleavage of these host sequences in vitro. Other patterns emerged from multispecies sequence alignments of the cleavage sites, which may have implications for the selection of animal models and zoonosis. SSHHPS/nsP is an example of a sequence-specific post-translational silencing mechanism.
Collapse
Affiliation(s)
- Nathanael
D. Reynolds
- Center
for Bio/molecular Science and Engineering (CBMSE), U.S. Naval Research Laboratory, 4555 Overlook Avenue, Washington, DC 20375, United States
| | | | - Jinny L. Liu
- Center
for Bio/molecular Science and Engineering (CBMSE), U.S. Naval Research Laboratory, 4555 Overlook Avenue, Washington, DC 20375, United States
| | - Jaimee R. Compton
- Center
for Bio/molecular Science and Engineering (CBMSE), U.S. Naval Research Laboratory, 4555 Overlook Avenue, Washington, DC 20375, United States
| | - Dagmar H. Leary
- Center
for Bio/molecular Science and Engineering (CBMSE), U.S. Naval Research Laboratory, 4555 Overlook Avenue, Washington, DC 20375, United States
| | - Brendan T. Freitas
- Center
for Drug Discovery, College of Pharmacy, University of Georgia, Athens, Georgia 30602, United States
| | - Scott D. Pegan
- Center
for Drug Discovery, College of Pharmacy, University of Georgia, Athens, Georgia 30602, United States
| | - Katarina Z. Doctor
- Navy
Center for Applied Research in AI (NCARAI) Information Technology
Division, U.S. Naval Research Laboratory, 4555 Overlook Ave., Washington, DC 20375, United States
| | - Fred Y. Wu
- Indiana
University Health Systems, Indiana University
School of Medicine, Bloomington, Indiana 47401, United States
| | - Xin Hu
- National
Center for Advancing Translational Sciences, National Institutes of
Health, Rockville, Maryland 20850, United
States
| | - Patricia M. Legler
- Center
for Bio/molecular Science and Engineering (CBMSE), U.S. Naval Research Laboratory, 4555 Overlook Avenue, Washington, DC 20375, United States
| |
Collapse
|
8
|
Ezan J, Moreau MM, Mamo TM, Shimbo M, Decroo M, Richter M, Peyroutou R, Rachel R, Tissir F, de Anda FC, Sans N, Montcouquiol M. Early loss of Scribble affects cortical development, interhemispheric connectivity and psychomotor activity. Sci Rep 2021; 11:9106. [PMID: 33907211 PMCID: PMC8079449 DOI: 10.1038/s41598-021-88147-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 04/01/2021] [Indexed: 12/03/2022] Open
Abstract
Neurodevelopmental disorders arise from combined defects in processes including cell proliferation, differentiation, migration and commissure formation. The evolutionarily conserved tumor-suppressor protein Scribble (Scrib) serves as a nexus to transduce signals for the establishment of apicobasal and planar cell polarity during these processes. Human SCRIB gene mutations are associated with neural tube defects and this gene is located in the minimal critical region deleted in the rare Verheij syndrome. In this study, we generated brain-specific conditional cKO mouse mutants and assessed the impact of the Scrib deletion on brain morphogenesis and behavior. We showed that embryonic deletion of Scrib in the telencephalon leads to cortical thickness reduction (microcephaly) and partial corpus callosum and hippocampal commissure agenesis. We correlated these phenotypes with a disruption in various developmental mechanisms of corticogenesis including neurogenesis, neuronal migration and axonal connectivity. Finally, we show that Scrib cKO mice have psychomotor deficits such as locomotor activity impairment and memory alterations. Altogether, our results show that Scrib is essential for early brain development due to its role in several developmental cellular mechanisms that could underlie some of the deficits observed in complex neurodevelopmental pathologies.
Collapse
Affiliation(s)
- Jerome Ezan
- Université de Bordeaux, INSERM, Neurocentre Magendie, U1215, 33077, Bordeaux, France.
| | - Maité M Moreau
- Université de Bordeaux, INSERM, Neurocentre Magendie, U1215, 33077, Bordeaux, France
| | - Tamrat M Mamo
- Université de Bordeaux, INSERM, Neurocentre Magendie, U1215, 33077, Bordeaux, France
| | - Miki Shimbo
- Université de Bordeaux, INSERM, Neurocentre Magendie, U1215, 33077, Bordeaux, France
| | - Maureen Decroo
- Université de Bordeaux, INSERM, Neurocentre Magendie, U1215, 33077, Bordeaux, France
| | - Melanie Richter
- Germany Center for Molecular Neurobiology Hamburg (ZMNH), Research Group Neuronal Development, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ronan Peyroutou
- Université de Bordeaux, INSERM, Neurocentre Magendie, U1215, 33077, Bordeaux, France
| | - Rivka Rachel
- Neurobiology-Neurodegeneration and Repair Laboratory, National Eye Institute, NIH, Bethesda, MD, 20892, USA
| | - Fadel Tissir
- Developmental Neurobiology Group, Institute of Neuroscience, University of Louvain, Avenue Mounier 73, Box B1.73.16, 1200, Brussels, Belgium
| | - Froylan Calderon de Anda
- Germany Center for Molecular Neurobiology Hamburg (ZMNH), Research Group Neuronal Development, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nathalie Sans
- Université de Bordeaux, INSERM, Neurocentre Magendie, U1215, 33077, Bordeaux, France
| | - Mireille Montcouquiol
- Université de Bordeaux, INSERM, Neurocentre Magendie, U1215, 33077, Bordeaux, France.
| |
Collapse
|
9
|
Xie X, Liang M, Yu C, Wei Z. Liprin-α-Mediated Assemblies and Their Roles in Synapse Formation. Front Cell Dev Biol 2021; 9:653381. [PMID: 33869211 PMCID: PMC8044993 DOI: 10.3389/fcell.2021.653381] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 02/25/2021] [Indexed: 01/20/2023] Open
Abstract
Brain's functions, such as memory and learning, rely on synapses that are highly specialized cellular junctions connecting neurons. Functional synapses orchestrate the assembly of ion channels, receptors, enzymes, and scaffold proteins in both pre- and post-synapse. Liprin-α proteins are master scaffolds in synapses and coordinate various synaptic proteins to assemble large protein complexes. The functions of liprin-αs in synapse formation have been largely uncovered by genetic studies in diverse model systems. Recently, emerging structural and biochemical studies on liprin-α proteins and their binding partners begin to unveil the molecular basis of the synaptic assembly. This review summarizes the recent structural findings on liprin-αs, proposes the assembly mechanism of liprin-α-mediated complexes, and discusses the liprin-α-organized assemblies in the regulation of synapse formation and function.
Collapse
Affiliation(s)
- Xingqiao Xie
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
- Academy for Advanced Interdisciplinary Studies, Southern University of Science and Technology, Shenzhen, China
| | - Mingfu Liang
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Cong Yu
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, China
| | - Zhiyi Wei
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
- Brain Research Center, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
10
|
Geisler SM, Benedetti A, Schöpf CL, Schwarzer C, Stefanova N, Schwartz A, Obermair GJ. Phenotypic Characterization and Brain Structure Analysis of Calcium Channel Subunit α 2δ-2 Mutant (Ducky) and α 2δ Double Knockout Mice. Front Synaptic Neurosci 2021; 13:634412. [PMID: 33679366 PMCID: PMC7933509 DOI: 10.3389/fnsyn.2021.634412] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 01/11/2021] [Indexed: 01/19/2023] Open
Abstract
Auxiliary α2δ subunits of voltage-gated calcium channels modulate channel trafficking, current properties, and synapse formation. Three of the four isoforms (α2δ-1, α2δ-2, and α2δ-3) are abundantly expressed in the brain; however, of the available knockout models, only α2δ-2 knockout or mutant mice display an obvious abnormal neurological phenotype. Thus, we hypothesize that the neuronal α2δ isoforms may have partially specific as well as redundant functions. To address this, we generated three distinct α2δ double knockout mouse models by crossbreeding single knockout (α2δ-1 and -3) or mutant (α2δ-2/ducky) mice. Here, we provide a first phenotypic description and brain structure analysis. We found that genotypic distribution of neonatal litters in distinct α2δ-1/-2, α2δ-1/-3, and α2δ-2/-3 breeding combinations did not conform to Mendel's law, suggesting premature lethality of single and double knockout mice. Notably, high occurrences of infant mortality correlated with the absence of specific α2δ isoforms (α2Δ-2 > α2δ-1 > α2δ-3), and was particularly observed in cages with behaviorally abnormal parenting animals of α2δ-2/-3 cross-breedings. Juvenile α2δ-1/-2 and α2δ-2/-3 double knockout mice displayed a waddling gate similar to ducky mice. However, in contrast to ducky and α2δ-1/-3 double knockout animals, α2δ-1/-2 and α2δ-2/-3 double knockout mice showed a more severe disease progression and highly impaired development. The observed phenotypes within the individual mouse lines may be linked to differences in the volume of specific brain regions. Reduced cortical volume in ducky mice, for example, was associated with a progressively decreased space between neurons, suggesting a reduction of total synaptic connections. Taken together, our findings show that α2δ subunits differentially regulate premature survival, postnatal growth, brain development, and behavior, suggesting specific neuronal functions in health and disease.
Collapse
Affiliation(s)
- Stefanie M. Geisler
- Institute of Physiology, Medical University Innsbruck, Innsbruck, Austria
- Department of Pharmacology and Toxicology, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria
| | - Ariane Benedetti
- Institute of Physiology, Medical University Innsbruck, Innsbruck, Austria
| | - Clemens L. Schöpf
- Institute of Physiology, Medical University Innsbruck, Innsbruck, Austria
| | - Christoph Schwarzer
- Department of Pharmacology, Medical University Innsbruck, Innsbruck, Austria
| | - Nadia Stefanova
- Division of Neurobiology, Department of Neurology, Medical University Innsbruck, Innsbruck, Austria
| | - Arnold Schwartz
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - Gerald J. Obermair
- Institute of Physiology, Medical University Innsbruck, Innsbruck, Austria
- Division Physiology, Karl Landsteiner University of Health Sciences, Krems an der Donau, Austria
| |
Collapse
|
11
|
Badea A, Schmalzigaug R, Kim W, Bonner P, Ahmed U, Johnson GA, Cofer G, Foster M, Anderson RJ, Badea C, Premont RT. Microcephaly with altered cortical layering in GIT1 deficiency revealed by quantitative neuroimaging. Magn Reson Imaging 2021; 76:26-38. [PMID: 33010377 PMCID: PMC7802083 DOI: 10.1016/j.mri.2020.09.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/25/2020] [Accepted: 09/25/2020] [Indexed: 01/06/2023]
Abstract
G Protein-Coupled Receptor Kinase-Interacting Protein-1 (GIT1) regulates neuronal functions, including cell and axon migration and synapse formation and maintenance, and GIT1 knockout (KO) mice exhibit learning and memory deficits. We noted that male and female GIT1-KO mice exhibit neuroimaging phenotypes including microcephaly, and altered cortical layering, with a decrease in neuron density in cortical layer V. Micro-CT and magnetic resonance microscopy (MRM) were used to identify morphometric phenotypes for the skulls and throughout the GIT1-KO brains. High field MRM of actively-stained mouse brains from GIT1-KO and wild type (WT) controls (n = 6 per group) allowed segmenting 37 regions, based on co-registration to the Waxholm Space atlas. Overall brain size in GIT1-KO mice was ~32% smaller compared to WT controls. After correcting for brain size, several regions were significantly different in GIT1-KO mice relative to WT, including the gray matter of the ventral thalamic nuclei and the rest of the thalamus, the inferior colliculus, and pontine nuclei. GIT1-KO mice had reduced volume of white matter tracts, most notably in the anterior commissure (~26% smaller), but also in the cerebral peduncle, fornix, and spinal trigeminal tract. On the other hand, the basal ganglia appeared enlarged in GIT1-KO mice, including the globus pallidus, caudate putamen, and particularly the accumbens - supporting a possible vulnerability to addiction. Volume based morphometry based on high-resolution MRM (21.5 μm isotropic voxels) was effective in detecting overall, and local differences in brain volumes in GIT1-KO mice, including in white matter tracts. The reduced relative volume of specific brain regions suggests a critical, but not uniform, role for GIT1 in brain development, conducive to brain microcephaly, and aberrant connectivity.
Collapse
Affiliation(s)
- Alexandra Badea
- Department of Radiology, Duke University Medical Center, Durham, NC 27710, United States of America; Department of Neurology, Duke University Medical Center, Durham, NC 27710, United States of America; Departments of Biomedical Engineering, Duke University Medical Center, Durham, NC 27710, United States of America; Brain Imaging and Analysis Center, Duke University Medical Center, Durham, NC 27710, United States of America.
| | - Robert Schmalzigaug
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States of America
| | - Woojoo Kim
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States of America
| | - Pamela Bonner
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States of America
| | - Umer Ahmed
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States of America
| | - G Allan Johnson
- Department of Radiology, Duke University Medical Center, Durham, NC 27710, United States of America; Departments of Biomedical Engineering, Duke University Medical Center, Durham, NC 27710, United States of America
| | - Gary Cofer
- Department of Radiology, Duke University Medical Center, Durham, NC 27710, United States of America
| | - Mark Foster
- Department of Radiology, Duke University Medical Center, Durham, NC 27710, United States of America
| | - Robert J Anderson
- Department of Radiology, Duke University Medical Center, Durham, NC 27710, United States of America
| | - Cristian Badea
- Department of Radiology, Duke University Medical Center, Durham, NC 27710, United States of America; Departments of Biomedical Engineering, Duke University Medical Center, Durham, NC 27710, United States of America
| | - Richard T Premont
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States of America.
| |
Collapse
|
12
|
Shao X, Liu L, Wei F, Liu Y, Wang F, Yi J, Sun L, Huang Y, Song Z, Yin W, Zhao H, Li Y. Fas and GIT1 signalling in the prefrontal cortex mediate behavioural sensitization to methamphetamine in mice. Brain Res Bull 2020; 164:361-371. [PMID: 32777257 DOI: 10.1016/j.brainresbull.2020.07.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 06/28/2020] [Accepted: 07/03/2020] [Indexed: 12/24/2022]
Abstract
PURPOSE Repeated methamphetamine (METH) administration in mice readily produces behavioural sensitization, but the underlying mechanisms remain elusive. The present research aimed to identify new targets affecting METH-induced behavioural sensitization. METHODS We first established a mouse model of METH-induced behavioural sensitization. To characterize the animal model, we performed behavioural tests at different stages of behavioural sensitization and simultaneously detected changes in several neurotransmitters in the prefrontal cortex (PFC). Next, we perfromed RNA sequencing (RNA-seq) to screen new targets, which were subsequently and verified by RT-PCR and western blot. Finally, we confirmed the roles of the new targets in METH-induced behavioural sensitization by injection of overexpressed lentiviruses and further detected related protein levels by western blot and histological changes by haematoxylin and eosin (HE) staining. RESULTS We successfully established a mouse model of METH-induced behavioural sensitization. The locomotor activities of the mice changed at different stages of sensitization, accompanied by changes in the levels of DA, 5-HT, GABA and glutamate. For RNA-seq analysis, we chose Fas as target, meanwhile, we chose GIT1 as target through literature. The detection of gene expression by RT-PCR indicated that METH-sensitized mice exhibited decreased levels of Fas, MEK1 and CREB and increased levels of Erk1/2 in the PFC. Western blot analysis revealed decreased Fas, GIT1, MEK1 and phosphorylated CREB levels and increased phosphorylated Erk1/2 levels in METH-sensitized mice. Injection of Fas and GIT1 injection showed that overexpression of Fas and GIT1 inhibited the induction of METH sensitization and reversed the changes in neurotransmitter levels and related protein levels, including MEK1, phosphorylated CREB and phosphorylated Erk1/2, in METH-sensitized mice. Overexpression of Fas and GIT1 also reduced histological lesions induced by METH. CONCLUSION The findings indicated that the development of behavioural sensitization to METH may be mediated by Fas and GIT1 through the MEK1-Erk1/2-CREB pathway.
Collapse
Affiliation(s)
- Xiaotong Shao
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, 130024, China
| | - Lei Liu
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, 130024, China
| | - Fuyao Wei
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, 130024, China
| | - Yucui Liu
- Research Center of Agriculture and Medicine Gene Engineering of Ministry of Education, Northeast Normal University, Changchun, 130024, China
| | - Fei Wang
- Research Center of Agriculture and Medicine Gene Engineering of Ministry of Education, Northeast Normal University, Changchun, 130024, China
| | - Jingwen Yi
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, 130024, China
| | - Luguo Sun
- Research Center of Agriculture and Medicine Gene Engineering of Ministry of Education, Northeast Normal University, Changchun, 130024, China
| | - Yanxin Huang
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, 130024, China
| | - Zhenbo Song
- Research Center of Agriculture and Medicine Gene Engineering of Ministry of Education, Northeast Normal University, Changchun, 130024, China
| | - Wu Yin
- Research Center of Agriculture and Medicine Gene Engineering of Ministry of Education, Northeast Normal University, Changchun, 130024, China.
| | - Huiying Zhao
- Department of Geriatrics, The First Hospital of Jilin University, Changchun, 130021, China
| | - Yunxin Li
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, 130024, China
| |
Collapse
|
13
|
Blanco-Luquin I, Acha B, Urdánoz-Casado A, Sánchez-Ruiz De Gordoa J, Vicuña-Urriza J, Roldán M, Labarga A, Zelaya MV, Cabello C, Méndez-López I, Mendioroz M. Early epigenetic changes of Alzheimer's disease in the human hippocampus. Epigenetics 2020; 15:1083-1092. [PMID: 32233750 DOI: 10.1080/15592294.2020.1748917] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The discovery of new biomarkers would be very valuable to improve the detection of early Alzheimer's disease (AD). DNA methylation marks may serve as epigenetic biomarkers of early AD. Here we identified epigenetic marks that are present in the human hippocampus from the earliest stages of AD. A previous methylome dataset of the human AD hippocampus was used to select a set of eight differentially methylated positions (DMPs) since early AD stages. Next, bisulphite pyrosequencing was performed in an expanded homogeneous cohort of 18 pure controls and 35 hippocampal samples with neuropathological changes of pure AD. Correlation between DNA methylation levels in DMPs and phospho-tau protein burden assessed by immunohistochemistry in the hippocampus was also determined. We found four DMPs showing higher levels of DNA methylation at early AD stages compared to controls, involving ELOVL2, GIT1/TP53I13 and the histone gene locus at chromosome 6. DNA methylation levels assessed by bisulphite pyrosequencing correlated with phospho-tau protein burden for ELOVL2 and HIST1H3E/HIST1H3 F genes. In this discovery study, a set of four epigenetic marks of early AD stages have been identified in the human hippocampus. It would be worth studying in-depth the specific pathways related to these epigenetic marks. These early alterations in DNA methylation in the AD hippocampus could be regarded as candidate biomarkers to be explored in future translational studies. ABBREVIATIONS AD: Alzheimer's disease; DMPs: Differentially methylated positions; CSF: Cerebrospinal fluid; βA42: β-amyloid 42; PET: positron emission tomography; 5mC: 5-methyl cytosine; CpG: cytosine-guanine dinucleotides; ANK1: ankyrin-1; BIN1: amphiphysin II; p-tau: hyperphosphorylated tau; CERAD: Consortium to Establish A Registry for Alzheimer's Disease; SD: standard deviation; ANOVA: one-way analysis of variance; VLCFAs: very long-chain fatty acids; DHA: docosahexaenoic acid; mTOR: mechanistic target of rapamycin.
Collapse
Affiliation(s)
- Idoia Blanco-Luquin
- Neuroepigenetics Laboratory-Navarrabiomed, Complejo Hospitalario De Navarra, Universidad Pública De Navarra (UPNA), IdiSNA (Navarra Institute for Health Research) , Pamplona, Spain
| | - Blanca Acha
- Neuroepigenetics Laboratory-Navarrabiomed, Complejo Hospitalario De Navarra, Universidad Pública De Navarra (UPNA), IdiSNA (Navarra Institute for Health Research) , Pamplona, Spain
| | - Amaya Urdánoz-Casado
- Neuroepigenetics Laboratory-Navarrabiomed, Complejo Hospitalario De Navarra, Universidad Pública De Navarra (UPNA), IdiSNA (Navarra Institute for Health Research) , Pamplona, Spain
| | - Javier Sánchez-Ruiz De Gordoa
- Neuroepigenetics Laboratory-Navarrabiomed, Complejo Hospitalario De Navarra, Universidad Pública De Navarra (UPNA), IdiSNA (Navarra Institute for Health Research) , Pamplona, Spain.,Department of Neurology, Complejo Hospitalario De Navarra- IdiSNA (Navarra Institute for Health Research) , Pamplona, Spain
| | - Janire Vicuña-Urriza
- Neuroepigenetics Laboratory-Navarrabiomed, Complejo Hospitalario De Navarra, Universidad Pública De Navarra (UPNA), IdiSNA (Navarra Institute for Health Research) , Pamplona, Spain
| | - Miren Roldán
- Neuroepigenetics Laboratory-Navarrabiomed, Complejo Hospitalario De Navarra, Universidad Pública De Navarra (UPNA), IdiSNA (Navarra Institute for Health Research) , Pamplona, Spain
| | - Alberto Labarga
- Bioinformatics Unit, Navarrabiomed, Public University of Navarre (UPNA), IdiSNA (Navarra Institute for Health Research) , Pamplona, Spain
| | - María Victoria Zelaya
- Department of Pathology, Complejo Hospitalario De Navarra- IdiSNA (Navarra Institute for Health Research) , Pamplona, Spain
| | - Carolina Cabello
- Department of Neurology, Complejo Hospitalario De Navarra- IdiSNA (Navarra Institute for Health Research) , Pamplona, Spain
| | - Iván Méndez-López
- Neuroepigenetics Laboratory-Navarrabiomed, Complejo Hospitalario De Navarra, Universidad Pública De Navarra (UPNA), IdiSNA (Navarra Institute for Health Research) , Pamplona, Spain.,Department of Internal Medicine, Hospital García-Orcoyen , Estella, Spain
| | - Maite Mendioroz
- Neuroepigenetics Laboratory-Navarrabiomed, Complejo Hospitalario De Navarra, Universidad Pública De Navarra (UPNA), IdiSNA (Navarra Institute for Health Research) , Pamplona, Spain.,Department of Neurology, Complejo Hospitalario De Navarra- IdiSNA (Navarra Institute for Health Research) , Pamplona, Spain
| |
Collapse
|
14
|
Kwon Y, Lee SJ, Lee E, Kim D, Park D. βPix heterozygous mice have defects in neuronal morphology and social interaction. Biochem Biophys Res Commun 2019; 516:1204-1210. [PMID: 31296385 DOI: 10.1016/j.bbrc.2019.07.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 07/01/2019] [Indexed: 11/20/2022]
Abstract
βPix activates Rho family small GTPases, Rac1 and Cdc42 as a guanine nucleotide exchange factor. Although overexpression of βPix in cultured neurons indicates that βPix is involved in spine morphogenesis and synapse formation in vitro, the in vivo role of βPix in the neuron is not well understood. Recently, we generated βPix knockout mice that showed lethality at embryonic day 9.5. Here, we investigate the neuronal role of βPix using βPix heterozygous mice that are viable and fertile. βPix heterozygous mice show decreased expression levels of βPix proteins in various tissues including the brain. Cultured hippocampal neurons from βPix heterozygous mice show a decrease in neurite length and complexity as well as synaptic density. Both excitatory and inhibitory synapse densities are decreased in these neurons. Golgi-staining of hippocampal tissues from the brain of these mice show reduced dendritic complexity and spine density in the hippocampal neurons. Expression levels of NMDA- and AMPA-receptor subunits and Git1 protein in hippocampal tissues are also decreased in these mice. Behaviorally, βPix heterozygous mice exhibit impaired social interaction. Altogether, these results indicate that βPix is required for neurite morphogenesis and synapse formation, and the reduced expression of βPix proteins results in a defect in social behavior.
Collapse
Affiliation(s)
- Younghee Kwon
- School of Biological Sciences, Seoul National University, Seoul, 08826, Republic of Korea.
| | - Seung Joon Lee
- Computational Biology & Genomics, Biogen, Cambridge, MA, 02142, USA.
| | - Eunee Lee
- Department of Anatomy, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea.
| | - Daesoo Kim
- Department of Biological Sciences, KAIST, Daejeon, 34141, Republic of Korea.
| | - Dongeun Park
- School of Biological Sciences, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
15
|
Trivino-Paredes JS, Nahirney PC, Pinar C, Grandes P, Christie BR. Acute slice preparation for electrophysiology increases spine numbers equivalently in the male and female juvenile hippocampus: a DiI labeling study. J Neurophysiol 2019; 122:958-969. [PMID: 31268808 DOI: 10.1152/jn.00332.2019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Hippocampal slices are widely used for in vitro electrophysiological experiments to study underlying mechanisms for synaptic transmission and plasticity, and there is a growing appreciation for sex differences in synaptic plasticity. To date, several studies have shown that the process of making slices from male animals can induce synaptogenesis in cornu ammonis area 1 (CA1) pyramidal cells, but there is a paucity of data for females and other brain regions. In the current study we use microcrystals of the lipophilic carbocyanine dye DiI (1,1'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate) to stain individual neurons in the CA1 and dentate gyrus (DG) hippocampal subfields of postnatal day 21 male and female rats. We show that the preparation of sections for electrophysiology produces significant increases in spines in sections obtained from females, similar to that observed in males. We also show that the procedures used for in vitro electrophysiology also result in significant spine increases in the DG and CA1 subfields. These results demonstrate the utility of this refined DiI procedure for staining neuronal dendrites and spines. They also show, for the first time, that in vitro electrophysiology slice preparations enhance spine numbers on hippocampal cells equivalently in both juvenile females and males.NEW & NOTEWORTHY This study introduces a new DiI technique that elucidates differences in spine numbers in juvenile female and male hippocampus, and shows that slice preparations for hippocampal electrophysiology in vitro may mask these differences.
Collapse
Affiliation(s)
- J S Trivino-Paredes
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - P C Nahirney
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada.,Island Medical Program, University of British Columbia, Victoria, British Columbia, Canada.,Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - C Pinar
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - P Grandes
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada.,Department of Neurosciences, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, Vizcaya, Spain.,Achucarro Basque Center for Neuroscience, Science Park of the UPV/EHU, Leioa, Vizcaya, Spain
| | - B R Christie
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada.,Island Medical Program, University of British Columbia, Victoria, British Columbia, Canada.,Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
16
|
Liang M, Xie X, Pan J, Jin G, Yu C, Wei Z. Structural basis of the target-binding mode of the G protein-coupled receptor kinase-interacting protein in the regulation of focal adhesion dynamics. J Biol Chem 2019; 294:5827-5839. [PMID: 30737283 DOI: 10.1074/jbc.ra118.006915] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 02/04/2019] [Indexed: 12/27/2022] Open
Abstract
Focal adhesions (FAs) are specialized sites where intracellular cytoskeleton elements connect to the extracellular matrix and thereby control cell motility. FA assembly depends on various scaffold proteins, including the G protein-coupled receptor kinase-interacting protein 1 (GIT1), paxillin, and liprin-α. Although liprin-α and paxillin are known to competitively interact with GIT1, the molecular basis governing these interactions remains elusive. To uncover the underlying mechanisms of how GIT1 is involved in FA assembly by alternatively binding to liprin-α and paxillin, here we solved the crystal structures of GIT1 in complex with liprin-α and paxillin at 1.8 and 2.6 Å resolutions, respectively. These structures revealed that the paxillin-binding domain (PBD) of GIT1 employs distinct binding modes to recognize a single α-helix of liprin-α and the LD4 motif of paxillin. Structure-based design of protein variants produced two binding-deficient GIT1 variants; specifically, these variants lost the ability to interact with liprin-α only or with both liprin-α and paxillin. Expressing the GIT1 variants in COS7 cells, we discovered that the two PBD-meditated interactions play different roles in either recruiting GIT1 to FA or facilitating FA assembly. Additionally, we demonstrate that, unlike for the known binding mode of the FAT domain to LD motifs, the PBD of GIT1 uses different surface patches to achieve high selectivity in LD motif recognition. In summary, our results have uncovered the mechanisms by which GIT1's PBD recognizes cognate paxillin and liprin-α structures, information we anticipate will be useful for future investigations of GIT1-protein interactions in cells.
Collapse
Affiliation(s)
- Mingfu Liang
- From the Department of Biology, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xingqiao Xie
- From the Department of Biology, Southern University of Science and Technology, Shenzhen 518055, China; Academy for Advanced Interdisciplinary Studies, Southern University of Science and Technology, Shenzhen 518055, China
| | - Jian Pan
- From the Department of Biology, Southern University of Science and Technology, Shenzhen 518055, China
| | - Gaowei Jin
- From the Department of Biology, Southern University of Science and Technology, Shenzhen 518055, China
| | - Cong Yu
- From the Department of Biology, Southern University of Science and Technology, Shenzhen 518055, China; the Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen 518055, China; the Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen 518055, China.
| | - Zhiyi Wei
- From the Department of Biology, Southern University of Science and Technology, Shenzhen 518055, China.
| |
Collapse
|
17
|
Chen J, Wang Q, Zhou W, Zhou Z, Tang PY, Xu T, Liu W, Li LW, Cheng L, Zhou ZM, Fan J, Yin GY. GPCR kinase 2-interacting protein-1 protects against ischemia-reperfusion injury of the spinal cord by modulating ASK1/JNK/p38 signaling. FASEB J 2018; 32:fj201800548. [PMID: 29912587 DOI: 10.1096/fj.201800548] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
GPCR kinase 2-interacting protein-1 (GIT1) is a scaffold protein that plays an important role in cell adaptation, proliferation, migration, and differentiation; however, the role of GIT1 in the regulation of neuronal death after spinal cord injury remains obscure. Here, we demonstrate that GIT1 deficiency remarkably increased neuronal apoptosis and enhanced JNK/p38 signaling, which resulted in stronger motor deficits by ischemia-reperfusion in vivo, consistent with the finding of oxygen-glucose deprivation/reoxygenation-induced neuronal injury in vitro. After treatment with JNK and p38 inhibitors, abnormally necroptotic cell death caused by GIT1 knockdown could be partially rescued, with the recovery of neuronal viability, which was still poorer than that in control neurons. Meanwhile, overactivation of JNK/p38 after GIT1 depletion was concomitant with excessive activity of apoptosis signal-regulating kinase-1 (ASK1) that could be abolished by ASK1 silencing in HEK293T cells. Finally, GIT1 could disrupt the oligomerization of ASK1 via interaction between the synaptic localization domain that contains the coiled-coil (CC)-2 domain of GIT1 and the C-terminal CC domain of ASK1. It suppressed the autophosphorylation of ASK1 and led to decreasing activity of the ASK1/JNK/p38 pathway. These data reveal a protective role for GIT1 in neuronal damage by modulating ASK1/JNK/p38 signaling.-Chen, J., Wang, Q., Zhou, W., Zhou, Z., Tang, P.-Y., Xu, T., Liu, W., Li, L.-W., Cheng, L., Zhou, Z.-M., Fan, J., Yin, G.-Y. GPCR kinase 2-interacting protein-1 protects against ischemia-reperfusion injury of the spinal cord by modulating ASK1/JNK/p38 signaling.
Collapse
Affiliation(s)
- Jian Chen
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qian Wang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Zhou
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zheng Zhou
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Peng-Yu Tang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Tao Xu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Liu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lin-Wei Li
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lin Cheng
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhi-Min Zhou
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jin Fan
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Guo-Yong Yin
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
18
|
Edogawa S, Peters SA, Jenkins GD, Gurunathan SV, Sundt WJ, Johnson S, Lennon RJ, Dyer RB, Camilleri M, Kashyap PC, Farrugia G, Chen J, Singh RJ, Grover M. Sex differences in NSAID-induced perturbation of human intestinal barrier function and microbiota. FASEB J 2018; 32:fj201800560R. [PMID: 29897814 PMCID: PMC6219825 DOI: 10.1096/fj.201800560r] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 05/21/2018] [Indexed: 12/12/2022]
Abstract
Intestinal barrier function and microbiota are integrally related and play critical roles in maintenance of host physiology. Sex is a key biologic variable for several disorders. Our aim was to determine sex-based differences in response to perturbation and subsequent recovery of intestinal barrier function and microbiota in healthy humans. Twenty-three volunteers underwent duodenal biopsies, mucosal impedance, and in vivo permeability measurement. Permeability testing was repeated after administration of indomethacin, then 4 to 6 wk after its discontinuation. Duodenal and fecal microbiota composition was determined using 16S rRNA amplicon sequencing. Healthy women had lower intestinal permeability and higher duodenal and fecal microbial diversity than healthy men. Intestinal permeability increases after indomethacin administration in both sexes. However, only women demonstrated decreased fecal microbial diversity, including an increase in Prevotella abundance, after indomethacin administration. Duodenal microbiota composition did not show sex-specific changes. The increase in permeability and microbiota changes normalized after discontinuation of indomethacin. In summary, women have lower intestinal permeability and higher microbial diversity. Intestinal permeability is sensitive to perturbation but recovers to baseline. Gut microbiota in women is sensitive to perturbation but appears to be more stable in men. Sex-based differences in intestinal barrier function and microbiome should be considered in future studies.-Edogawa, S., Peters, S. A., Jenkins, G. D., Gurunathan, S. V., Sundt, W. J., Johnson, S., Lennon, R. J., Dyer, R. B., Camilleri, M., Kashyap, P. C., Farrugia, G., Chen, J., Singh, R. J., Grover, M. Sex differences in NSAID-induced perturbation of human intestinal barrier function and microbiota.
Collapse
Affiliation(s)
- Shoko Edogawa
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Stephanie A. Peters
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Gregory D. Jenkins
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, USA
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Wendy J. Sundt
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Stephen Johnson
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, USA
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Ryan J. Lennon
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, USA
| | - Roy B. Dyer
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Michael Camilleri
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Purna C. Kashyap
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Gianrico Farrugia
- Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, Florida, USA
| | - Jun Chen
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, USA
| | - Ravinder J. Singh
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Madhusudan Grover
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
19
|
van Gastel J, Boddaert J, Jushaj A, Premont RT, Luttrell LM, Janssens J, Martin B, Maudsley S. GIT2-A keystone in ageing and age-related disease. Ageing Res Rev 2018; 43:46-63. [PMID: 29452267 DOI: 10.1016/j.arr.2018.02.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 02/06/2018] [Accepted: 02/08/2018] [Indexed: 12/15/2022]
Abstract
Since its discovery, G protein-coupled receptor kinase-interacting protein 2, GIT2, and its family member, GIT1, have received considerable interest concerning their potential key roles in regulating multiple inter-connected physiological and pathophysiological processes. GIT2 was first identified as a multifunctional protein that is recruited to G protein-coupled receptors (GPCRs) during the process of receptor internalization. Recent findings have demonstrated that perhaps one of the most important effects of GIT2 in physiology concerns its role in controlling multiple aspects of the complex ageing process. Ageing can be considered the most prevalent pathophysiological condition in humans, affecting all tissue systems and acting as a driving force for many common and intractable disorders. The ageing process involves a complex interplay among various deleterious activities that profoundly disrupt the body's ability to cope with damage, thus increasing susceptibility to pathophysiologies such as neurodegeneration, central obesity, osteoporosis, type 2 diabetes mellitus and atherosclerosis. The biological systems that control ageing appear to function as a series of interconnected complex networks. The inter-communication among multiple lower-complexity signaling systems within the global ageing networks is likely coordinated internally by keystones or hubs, which regulate responses to dynamic molecular events through protein-protein interactions with multiple distinct partners. Multiple lines of research have suggested that GIT2 may act as one of these network coordinators in the ageing process. Identifying and targeting keystones, such as GIT2, is thus an important approach in our understanding of, and eventual ability to, medically ameliorate or interdict age-related progressive cellular and tissue damage.
Collapse
|
20
|
Kim YS, Woo J, Lee CJ, Yoon BE. Decreased Glial GABA and Tonic Inhibition in Cerebellum of Mouse Model for Attention-Deficit/Hyperactivity Disorder (ADHD). Exp Neurobiol 2017; 26:206-212. [PMID: 28912643 PMCID: PMC5597551 DOI: 10.5607/en.2017.26.4.206] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 08/23/2017] [Accepted: 08/23/2017] [Indexed: 01/19/2023] Open
Abstract
About 5~12% of school-aged children suffer from the Attention-Deficit/Hyperactivity Disorder (ADHD). However, the core mechanism of ADHD remains unclear. G protein-coupled receptor kinase-interacting protein-1 (GIT1) has recently been reported to be associated with ADHD in human and the genetic deletion of GIT1 result in ADHD-like behaviors in mice. Mice lacking GIT1 shows a shift in neuronal excitation/inhibition (E/I) balance. However, the pricise mechanism for E/I imbalance and the role of neuron-glia interaction in GIT1 knockout (KO) mice have not been studied. Especially, a possible contribution of glial GABA and tonic inhibition mediated by astrocytic GABA release in the mouse model for ADHD remains unexplored. Therefore, we investigated the changes in the amount of GABA and degree of tonic inhibition in GIT1 KO mice. We observed a decreased glial GABA intensity in GIT1 KO mice compared to wild type (WT) mice and an attenuation of tonic current from cerebellar granule cells in GIT1 KO mice. Our study identifies the previously unknown mechanism of reduced astrocytic GABA and tonic inhibition in GIT1 lacking mice as a potential cause of hyperactivity disorder.
Collapse
Affiliation(s)
- Yoo Sung Kim
- Department of Molecular biology, Dankook University, Cheonan, Chungnam 31116, Korea
| | - Junsung Woo
- Center for Neuroscience and Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea.,Neuroscience Program, University of Science and Technology (UST), Daejeon 34113, Korea
| | - C Justin Lee
- Center for Neuroscience and Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea.,Neuroscience Program, University of Science and Technology (UST), Daejeon 34113, Korea.,KU-KIST Graduate School of Converging Sciences and Technologies, Korea University, Seoul 02841, Korea
| | - Bo-Eun Yoon
- Department of Molecular biology, Dankook University, Cheonan, Chungnam 31116, Korea
| |
Collapse
|
21
|
Zhou W, Li X, Premont RT. Expanding functions of GIT Arf GTPase-activating proteins, PIX Rho guanine nucleotide exchange factors and GIT-PIX complexes. J Cell Sci 2017; 129:1963-74. [PMID: 27182061 DOI: 10.1242/jcs.179465] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The GIT proteins, GIT1 and GIT2, are GTPase-activating proteins (inactivators) for the ADP-ribosylation factor (Arf) small GTP-binding proteins, and function to limit the activity of Arf proteins. The PIX proteins, α-PIX and β-PIX (also known as ARHGEF6 and ARHGEF7, respectively), are guanine nucleotide exchange factors (activators) for the Rho family small GTP-binding protein family members Rac1 and Cdc42. Through their multi-domain structures, GIT and PIX proteins can also function as signaling scaffolds by binding to numerous protein partners. Importantly, the constitutive association of GIT and PIX proteins into oligomeric GIT-PIX complexes allows these two proteins to function together as subunits of a larger structure that coordinates two distinct small GTP-binding protein pathways and serves as multivalent scaffold for the partners of both constituent subunits. Studies have revealed the involvement of GIT and PIX proteins, and of the GIT-PIX complex, in numerous fundamental cellular processes through a wide variety of mechanisms, pathways and signaling partners. In this Commentary, we discuss recent findings in key physiological systems that exemplify current understanding of the function of this important regulatory complex. Further, we draw attention to gaps in crucial information that remain to be filled to allow a better understanding of the many roles of the GIT-PIX complex in health and disease.
Collapse
Affiliation(s)
- Wu Zhou
- Department of Medicine, College of Medicine and Health, Lishui University, Lishui 323000, China
| | - Xiaobo Li
- Department of Computer Science and Technology, College of Engineering and Design, Lishui University, Lishui 323000, China
| | - Richard T Premont
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
22
|
Chen P, Gu WL, Gong MZ, Wang J, Li DQ. GIT1 gene deletion delays chondrocyte differentiation and healing of tibial plateau fracture through suppressing proliferation and apoptosis of chondrocyte. BMC Musculoskelet Disord 2017; 18:320. [PMID: 28754105 PMCID: PMC5534123 DOI: 10.1186/s12891-017-1653-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 06/30/2017] [Indexed: 12/30/2022] Open
Abstract
Background Although tibial plateau fracture is an uncommon injury, its regulation is challenging and there are some influencing factors, including the effects of severe bone displacement, depression and cancellous bone cartilage, and inevitable cartilage damage. And GIT1 plays an important role in bone mass and 78 osteoblast cell migration. Methods The study used 72 C57/BL6 mice. A tibial plateau fracture model was established by using mice with the same number of GIT1 gene deletions (the experimental group) and their wild-type littermates (the control group). Joint and bone callus recovery were evaluated by X-ray and CT thin layer scans. Micro CT assay and histomorphometry were conducted in order to evaluate the volume of newly formed blood vessels. Type II collagen expression in tibial tissues after tibial plateau fracture were detected by immunohistochemistry after 7, 14 and 21 days. The number of proliferating cell nuclear antigen (PCNA) positive cells after tibial plateau fracture was tested by immunohistochemistry after 14 and 21 days. The terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling (TUNEL) staining was conducted after 14 and 21 days in order to test chondrocyte apoptosis in tibial tissues after tibial plateau fracture. Results The GIT1 gene deletion group mice spent less time on the rotating rod than the control group mice (P < 0.05). Compared with the control group, postoperative recovery was retarded, because GIT1 gene deletion slowed down neovascularization after tibial plateau fracture (P < 0.05). Compared with the control group, mouse type II collagen expression significantly decreased in the GIT1 gene deletion group, and the proportion of PCNA positive cells significantly decreased (P < 0.05). The TUNEL results indicate that GIT1 gene deletion led to reduced chondrocyte apoptosis. Conclusion GIT1 gene deletion can inhibit chondrocyte proliferation and apoptosis during the recovery of tibial plateau fracture, so as to delay chondrocyte differentiation and tibial plateau fracture healing.
Collapse
Affiliation(s)
- Peng Chen
- Department of Trauma Orthopedics, The Second Hospital of Shandong University, Jinan, 250033, People's Republic of China
| | - Wan-Li Gu
- Department of Trauma Orthopedics, The Second Hospital of Shandong University, Jinan, 250033, People's Republic of China
| | - Ming-Zhi Gong
- Department of Trauma Orthopedics, The Second Hospital of Shandong University, Jinan, 250033, People's Republic of China
| | - Jun Wang
- Department of Trauma Orthopedics, The Second Hospital of Shandong University, Jinan, 250033, People's Republic of China
| | - Dong-Qing Li
- Department of Operating Theater, The Second Hospital of Shandong University, No. 247, Beiyuan Street, Jinan, 250033, Shandong Province, People's Republic of China.
| |
Collapse
|
23
|
Uddin M, Pellecchia G, Thiruvahindrapuram B, D'Abate L, Merico D, Chan A, Zarrei M, Tammimies K, Walker S, Gazzellone MJ, Nalpathamkalam T, Yuen RKC, Devriendt K, Mathonnet G, Lemyre E, Nizard S, Shago M, Joseph-George AM, Noor A, Carter MT, Yoon G, Kannu P, Tihy F, Thorland EC, Marshall CR, Buchanan JA, Speevak M, Stavropoulos DJ, Scherer SW. Indexing Effects of Copy Number Variation on Genes Involved in Developmental Delay. Sci Rep 2016; 6:28663. [PMID: 27363808 PMCID: PMC4929460 DOI: 10.1038/srep28663] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 06/06/2016] [Indexed: 01/03/2023] Open
Abstract
A challenge in clinical genomics is to predict whether copy number variation (CNV) affecting a gene or multiple genes will manifest as disease. Increasing recognition of gene dosage effects in neurodevelopmental disorders prompted us to develop a computational approach based on critical-exon (highly expressed in brain, highly conserved) examination for potential etiologic effects. Using a large CNV dataset, our updated analyses revealed significant (P < 1.64 × 10−15) enrichment of critical-exons within rare CNVs in cases compared to controls. Separately, we used a weighted gene co-expression network analysis (WGCNA) to construct an unbiased protein module from prenatal and adult tissues and found it significantly enriched for critical exons in prenatal (P < 1.15 × 10−50, OR = 2.11) and adult (P < 6.03 × 10−18, OR = 1.55) tissues. WGCNA yielded 1,206 proteins for which we prioritized the corresponding genes as likely to have a role in neurodevelopmental disorders. We compared the gene lists obtained from critical-exon and WGCNA analysis and found 438 candidate genes associated with CNVs annotated as pathogenic, or as variants of uncertain significance (VOUS), from among 10,619 developmental delay cases. We identified genes containing CNVs previously considered to be VOUS to be new candidate genes for neurodevelopmental disorders (GIT1, MVB12B and PPP1R9A) demonstrating the utility of this strategy to index the clinical effects of CNVs.
Collapse
Affiliation(s)
- Mohammed Uddin
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada.,Program in Genetics and Genome Biology (GGB), The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Giovanna Pellecchia
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada.,Program in Genetics and Genome Biology (GGB), The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Bhooma Thiruvahindrapuram
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada.,Program in Genetics and Genome Biology (GGB), The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Lia D'Abate
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada.,Program in Genetics and Genome Biology (GGB), The Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Daniele Merico
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada.,Program in Genetics and Genome Biology (GGB), The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Ada Chan
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada.,Program in Genetics and Genome Biology (GGB), The Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Mehdi Zarrei
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada.,Program in Genetics and Genome Biology (GGB), The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Kristiina Tammimies
- Center of Neurodevelopmental Disorders (KIND), Neuropsychiatric Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Susan Walker
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada.,Program in Genetics and Genome Biology (GGB), The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Matthew J Gazzellone
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada.,Program in Genetics and Genome Biology (GGB), The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Thomas Nalpathamkalam
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada.,Program in Genetics and Genome Biology (GGB), The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Ryan K C Yuen
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada.,Program in Genetics and Genome Biology (GGB), The Hospital for Sick Children, Toronto, Ontario, Canada
| | | | | | - Emmanuelle Lemyre
- CHU Sainte-Justine, University de Montreal, Montreal, Quebec, Canada
| | - Sonia Nizard
- CHU Sainte-Justine, University de Montreal, Montreal, Quebec, Canada
| | - Mary Shago
- Genome Diagnostics, Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Ann M Joseph-George
- Genome Diagnostics, Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Abdul Noor
- Department of Pathology and Laboratory Medicine, Division of Diagnostic Medical Genetics, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Melissa T Carter
- Department of Genetics, The Children's Hospital of Eastern Ontario, Ottawa, ON, Canada
| | - Grace Yoon
- Division of Clinical and Metabolic Genetics, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario M5G 2L3, Canada
| | - Peter Kannu
- Division of Clinical and Metabolic Genetics, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario M5G 2L3, Canada
| | - Frédérique Tihy
- CHU Sainte-Justine, University de Montreal, Montreal, Quebec, Canada
| | - Erik C Thorland
- Cytogenetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Christian R Marshall
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada.,Genome Diagnostics, Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Janet A Buchanan
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada.,Program in Genetics and Genome Biology (GGB), The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Marsha Speevak
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Dimitri J Stavropoulos
- Genome Diagnostics, Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Stephen W Scherer
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada.,Program in Genetics and Genome Biology (GGB), The Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada.,McLaughlin Centre, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
24
|
A novel de novo microdeletion at 17q11.2 adjacent to NF1 gene associated with developmental delay, short stature, microcephaly and dysmorphic features. Mol Cytogenet 2016; 9:41. [PMID: 27247625 PMCID: PMC4886423 DOI: 10.1186/s13039-016-0251-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 05/18/2016] [Indexed: 11/24/2022] Open
Abstract
Background Microdeletions at 17q11.2 often encompass NF1 gene, is the cause for NF1 microdeletion syndrome. Microdeletion at 17q11.2 without the involvement of NF1 gene is rarely reported. Case presentation Here we reported a patient carrying a novel de novo deletion at 17q11.2 adjacent to NF1 gene, who presented with developmental delay, short stature, postnatal microcephaly, underweight and dysmorphic features including flat facial profile, dolicocephaly, hypertelorism, short philtrum, flat nasal bridge and posteriorly rotated and low set ears. Chromosomal microarray analysis revealed a 1.69 Mb de novo deletion at 17q11.2 adjacent to NF1 gene, which involves 43 RefSeq genes. We compared this with four overlapping deletions at this interval. Conclusions A rare de novo microdeletion at 17q11.2 not involving NF1 gene is associated with developmental delay and dysmorphic features. Seven genes, TAOK1, PHF12, NUFIP2, SLC26A4, SEZ6, GIT1 and TRAF4 are possible candidates for the clinical features of our patient. The delineation of this rare deletion and description of associated clinical phenotypes will help to understand the genotype-phenotype correlation of genomic imbalances at this locus.
Collapse
|
25
|
Li YS, Qin LX, Liu J, Xia WL, Li JP, Shen HL, Gao WQ. GIT1 enhances neurite outgrowth by stimulating microtubule assembly. Neural Regen Res 2016; 11:427-34. [PMID: 27127481 PMCID: PMC4829007 DOI: 10.4103/1673-5374.179054] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
GIT1, a G-protein-coupled receptor kinase interacting protein, has been reported to be involved in neurite outgrowth. However, the neurobiological functions of the protein remain unclear. In this study, we found that GIT1 was highly expressed in the nervous system, and its expression was maintained throughout all stages of neuritogenesis in the brain. In primary cultured mouse hippocampal neurons from GIT1 knockout mice, there was a significant reduction in total neurite length per neuron, as well as in the average length of axon-like structures, which could not be prevented by nerve growth factor treatment. Overexpression of GIT1 significantly promoted axon growth and fully rescued the axon outgrowth defect in the primary hippocampal neuron cultures from GIT1 knockout mice. The GIT1 N terminal region, including the ADP ribosylation factor-GTPase activating protein domain, the ankyrin domains and the Spa2 homology domain, were sufficient to enhance axonal extension. Importantly, GIT1 bound to many tubulin proteins and microtubule-associated proteins, and it accelerated microtubule assembly in vitro. Collectively, our findings suggest that GIT1 promotes neurite outgrowth, at least partially by stimulating microtubule assembly. This study provides new insight into the cellular and molecular pathogenesis of GIT1-associated neurological diseases.
Collapse
Affiliation(s)
- Yi-Sheng Li
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Stem Cell Research Center, Ren Ji Hospital, School of Biomedical Engineering & Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Li-Xia Qin
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Stem Cell Research Center, Ren Ji Hospital, School of Biomedical Engineering & Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Jie Liu
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Stem Cell Research Center, Ren Ji Hospital, School of Biomedical Engineering & Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Wei-Liang Xia
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Stem Cell Research Center, Ren Ji Hospital, School of Biomedical Engineering & Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Jian-Ping Li
- Department of Neurology, Shanghai Renji Hospital, Shanghai, China
| | - Hai-Lian Shen
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Stem Cell Research Center, Ren Ji Hospital, School of Biomedical Engineering & Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Wei-Qiang Gao
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Stem Cell Research Center, Ren Ji Hospital, School of Biomedical Engineering & Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China; Collarative Innovation Center of Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
26
|
X-linked intellectual disability gene CASK regulates postnatal brain growth in a non-cell autonomous manner. Acta Neuropathol Commun 2016; 4:30. [PMID: 27036546 PMCID: PMC4818453 DOI: 10.1186/s40478-016-0295-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 03/07/2016] [Indexed: 01/07/2023] Open
Abstract
The phenotypic spectrum among girls with heterozygous mutations in the X-linked intellectual disability (XLID) gene CASK (calcium/calmodulin-dependent serine protein kinase) includes postnatal microcephaly, ponto-cerebellar hypoplasia, seizures, optic nerve hypoplasia, growth retardation and hypotonia. Although CASK knockout mice were previously reported to exhibit perinatal lethality and a 3-fold increased apoptotic rate in the brain, CASK deletion was not found to affect neuronal physiology and their electrical properties. The pathogenesis of CASK associated disorders and the potential function of CASK therefore remains unknown. Here, using Cre-LoxP mediated gene excision experiments; we demonstrate that deleting CASK specifically from mouse cerebellar neurons does not alter the cerebellar architecture or function. We demonstrate that the neuron-specific deletion of CASK in mice does not cause perinatal lethality but induces severe recurrent epileptic seizures and growth retardation before the onset of adulthood. Furthermore, we demonstrate that although neuron-specific haploinsufficiency of CASK is inconsequential, the CASK mutation associated human phenotypes are replicated with high fidelity in CASK heterozygous knockout female mice (CASK(+/-)). These data suggest that CASK-related phenotypes are not purely neuronal in origin. Surprisingly, the observed microcephaly in CASK(+/-) animals is not associated with a specific loss of CASK null brain cells indicating that CASK regulates postnatal brain growth in a non-cell autonomous manner. Using biochemical assay, we also demonstrate that CASK can interact with metabolic proteins. CASK knockdown in human cell lines cause reduced cellular respiration and CASK(+/-) mice display abnormalities in muscle and brain oxidative metabolism, suggesting a novel function of CASK in metabolism. Our data implies that some phenotypic components of CASK heterozygous deletion mutation associated disorders represent systemic manifestation of metabolic stress and therefore amenable to therapeutic intervention.
Collapse
|