1
|
Altas B, Tuffy LP, Patrizi A, Dimova K, Soykan T, Brandenburg C, Romanowski AJ, Whitten JR, Robertson CD, Khim SN, Crutcher GW, Ambrozkiewicz MC, Yagensky O, Krueger-Burg D, Hammer M, Hsiao HH, Laskowski PR, Dyck L, Puche AC, Sassoè-Pognetto M, Chua JJE, Urlaub H, Jahn O, Brose N, Poulopoulos A. Region-Specific Phosphorylation Determines Neuroligin-3 Localization to Excitatory Versus Inhibitory Synapses. Biol Psychiatry 2024; 96:815-828. [PMID: 38154503 PMCID: PMC11209832 DOI: 10.1016/j.biopsych.2023.12.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 12/19/2023] [Accepted: 12/19/2023] [Indexed: 12/30/2023]
Abstract
BACKGROUND Neuroligin-3 is a postsynaptic adhesion molecule involved in synapse development and function. It is implicated in rare, monogenic forms of autism, and its shedding is critical to the tumor microenvironment of gliomas. While other members of the neuroligin family exhibit synapse-type specificity in localization and function through distinct interactions with postsynaptic scaffold proteins, the specificity of neuroligin-3 synaptic localization remains largely unknown. METHODS We investigated the synaptic localization of neuroligin-3 across regions in mouse and human brain samples after validating antibody specificity in knockout animals. We raised a phospho-specific neuroligin antibody and used phosphoproteomics, cell-based assays, and in utero CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/Cas9) knockout and gene replacement to identify mechanisms that regulate neuroligin-3 localization to distinct synapse types. RESULTS Neuroligin-3 exhibits region-dependent synapse specificity, largely localizing to excitatory synapses in cortical regions and inhibitory synapses in subcortical regions of the brain in both mice and humans. We identified specific phosphorylation of cortical neuroligin-3 at a key binding site for recruitment to inhibitory synapses, while subcortical neuroligin-3 remained unphosphorylated. In vitro, phosphomimetic mutation of that site disrupted neuroligin-3 association with the inhibitory postsynaptic scaffolding protein gephyrin. In vivo, phosphomimetic mutants of neuroligin-3 localized to excitatory postsynapses, while phospho-null mutants localized to inhibitory postsynapses. CONCLUSIONS These data reveal an unexpected region-specific pattern of neuroligin-3 synapse specificity, as well as a phosphorylation-dependent mechanism that regulates its recruitment to either excitatory or inhibitory synapses. These findings add to our understanding of how neuroligin-3 is involved in conditions that may affect the balance of excitation and inhibition.
Collapse
Affiliation(s)
- Bekir Altas
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Liam P Tuffy
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Annarita Patrizi
- Department of Neuroscience Rita Levi Montalcini, University of Turin, Turin, Italy
| | - Kalina Dimova
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany; Neuroproteomics Group, Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Tolga Soykan
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Cheryl Brandenburg
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Andrea J Romanowski
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Julia R Whitten
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Colin D Robertson
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Saovleak N Khim
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Garrett W Crutcher
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Mateusz C Ambrozkiewicz
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Oleksandr Yagensky
- Research Group Protein Trafficking in Synaptic Development and Function, Laboratory of Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Dilja Krueger-Burg
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Matthieu Hammer
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - He-Hsuan Hsiao
- Bioanalytical Mass Spectrometry, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Pawel R Laskowski
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Lydia Dyck
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Adam C Puche
- Department of Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland
| | | | - John J E Chua
- Research Group Protein Trafficking in Synaptic Development and Function, Laboratory of Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Henning Urlaub
- Bioanalytical Mass Spectrometry, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany; Bioanalytics Group, Institute of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Olaf Jahn
- Neuroproteomics Group, Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany; Translational Neuroproteomics Group, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
| | - Nils Brose
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Alexandros Poulopoulos
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland; Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.
| |
Collapse
|
2
|
Boyd R, Jaqaman K, Wang W. Weaker neuroligin 2 - neurexin 1β interaction tethers membranes and signal synaptogenesis through clustering. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.16.618688. [PMID: 39464163 PMCID: PMC11507839 DOI: 10.1101/2024.10.16.618688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Single-pass transmembrane proteins neuroligin (NL) and neurexin (NRX) constitute a pair of synaptic adhesion molecules (SAMs) that are essential for the formation of functional synapses. Binding affinities vary by ∼ 1000 folds between arrays of NL and NRX subtypes, which contribute to chemical and spatial specificities. Current structures are obtained with truncated extracellular domains of NL and NRX and are limited to the higher-affinity NL1/4-NRX complexes. How NL-NRX interaction leads to functional synapses remains unknown. Here we report structures of full-length NL2 alone, and in complex with NRX1β in several conformations, which has the lowest affinity among major NL-NRX subtypes. We show how conformational flexibilities may help in adapting local membrane geometry, and reveal mechanisms underlying variations in NL-NRX affinities modulation. We further show that, despite lower affinity, NL2-NRX1β interaction alone is capable of tethering different lipid membranes in total reconstitution, and that NL2 and NRX1β cluster at inter-cellular junctions without the need of other synaptic components. In addition, NL2 combines with the master post-synaptic scaffolding protein gephyrin and clusters neurotransmitter receptors at cellular membrane. These findings suggest dual roles of NL2 - NRX1β interaction - both as mechanical tether, and as signaling receptors, to ensure correct spatial and chemical coordination between two cells to generate function synapses.
Collapse
|
3
|
Abstract
Herein, I intend to capture highlights shared with my academic and research colleagues over the 60 years I devoted initially to my graduate and postdoctoral training and then to academic endeavors starting as an assistant professor in a new medical school at the University of California, San Diego (UCSD). During this period, the Department of Pharmacology emerged from a division within the Department of Medicine to become the first basic science department, solely within the School of Medicine at UCSD in 1979. As part of the school's plans to reorganize and to retain me at UCSD, I was appointed as founding chair. Some years later in 2002, faculty, led largely within the Department of Pharmacology and by practicing pharmacists within UCSD Healthcare, started the independent Skaggs School of Pharmacy and Pharmaceutical Sciences with a doctor of pharmacy (PharmD) program, where I served as the founding dean. My career pathway, from working at my family-owned pharmacy to chairing a department in a school of medicine and then becoming the dean of a school of pharmacy at a research-intensive, student-centered institution, involved some risky decisions. But the academic, curricular, and accreditation challenges posed were met by a cadre of creative faculty colleagues. I offer my experiences to individuals confronted with a multiplicity of real or imagined opportunities in academic health sciences, the related pharmaceutical industry, and government oversight agencies.
Collapse
Affiliation(s)
- Palmer Taylor
- Department of Pharmacology, Skaggs School of Pharmacy and Pharmaceutical Sciences, and School of Medicine, University of California, San Diego, La Jolla, California 92093, USA
| |
Collapse
|
4
|
Comoletti D, Trobiani L, Chatonnet A, Bourne Y, Marchot P. Comparative mapping of selected structural determinants on the extracellular domains of cholinesterase-like cell-adhesion molecules. Neuropharmacology 2020; 184:108381. [PMID: 33166544 DOI: 10.1016/j.neuropharm.2020.108381] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/10/2020] [Accepted: 10/29/2020] [Indexed: 11/18/2022]
Abstract
Cell adhesion generally involves formation of homophilic or heterophilic protein complexes between two cells to form transcellular junctions. Neural cell-adhesion members of the α/β-hydrolase fold superfamily of proteins use their extracellular or soluble cholinesterase-like domain to bind cognate partners across cell membranes, as illustrated by the neuroligins. These cell-adhesion molecules currently comprise the synaptic organizers neuroligins found in all animal phyla, along with three proteins found only in invertebrates: the guidance molecule neurotactin, the glia-specific gliotactin, and the basement membrane protein glutactin. Although these proteins share a cholinesterase-like fold, they lack one or more residues composing the catalytic triad responsible for the enzymatic activity of the cholinesterases. Conversely, they are found in various subcellular localisations and display specific disulfide bonding and N-glycosylation patterns, along with individual surface determinants possibly associated with recognition and binding of protein partners. Formation of non-covalent dimers typical of the cholinesterases is documented for mammalian neuroligins, yet whether invertebrate neuroligins and their neurotactin, gliotactin and glutactin relatives also form dimers in physiological conditions is unknown. Here we provide a brief overview of the localization, function, evolution, and conserved versus individual structural determinants of these cholinesterase-like cell-adhesion proteins. This article is part of the special issue entitled 'Acetylcholinesterase Inhibitors: From Bench to Bedside to Battlefield'.
Collapse
Affiliation(s)
- Davide Comoletti
- School of Biological Sciences, Victoria University of Wellington, Wellington, 6012, New Zealand; Child Health Institute of New Jersey, New Brunswick, NJ 08901, USA; Department of Neuroscience and Cell Biology Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA.
| | - Laura Trobiani
- School of Biological Sciences, Victoria University of Wellington, Wellington, 6012, New Zealand
| | - Arnaud Chatonnet
- Lab 'Dynamique Musculaire et Métabolisme', Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE) / Université Montpellier, Montpellier, France
| | - Yves Bourne
- Lab 'Architecture et Fonction des Macromolécules Biologiques (AFMB)', Centre National de la Recherche Scientifique (CNRS)/Aix-Marseille Univ, Faculté des Sciences - Campus Luminy, Marseille, France
| | - Pascale Marchot
- Lab 'Architecture et Fonction des Macromolécules Biologiques (AFMB)', Centre National de la Recherche Scientifique (CNRS)/Aix-Marseille Univ, Faculté des Sciences - Campus Luminy, Marseille, France.
| |
Collapse
|
5
|
The soluble neurexin-1β ectodomain causes calcium influx and augments dendritic outgrowth and synaptic transmission. Sci Rep 2020; 10:18041. [PMID: 33093500 PMCID: PMC7582164 DOI: 10.1038/s41598-020-75047-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/08/2020] [Indexed: 12/15/2022] Open
Abstract
Classically, neurexins are thought to mediate synaptic connections through trans interactions with a number of different postsynaptic partners. Neurexins are cleaved by metalloproteases in an activity-dependent manner, releasing the soluble extracellular domain. Here, we report that in both immature (before synaptogenesis) and mature (after synaptogenesis) hippocampal neurons, the soluble neurexin-1β ectodomain triggers acute Ca2+-influx at the dendritic/postsynaptic side. In both cases, neuroligin-1 expression was required. In immature neurons, calcium influx required N-type calcium channels and stimulated dendritic outgrowth and neuronal survival. In mature glutamatergic neurons the neurexin-1β ectodomain stimulated calcium influx through NMDA-receptors, which increased presynaptic release probability. In contrast, prolonged exposure to the ectodomain led to inhibition of synaptic transmission. This secondary inhibition was activity- and neuroligin-1 dependent and caused by a reduction in the readily-releasable pool of vesicles. A synthetic peptide modeled after the neurexin-1β:neuroligin-1 interaction site reproduced the cellular effects of the neurexin-1β ectodomain. Collectively, our findings demonstrate that the soluble neurexin ectodomain stimulates growth of neurons and exerts acute and chronic effects on trans-synaptic signaling involved in setting synaptic strength.
Collapse
|
6
|
Trobiani L, Meringolo M, Diamanti T, Bourne Y, Marchot P, Martella G, Dini L, Pisani A, De Jaco A, Bonsi P. The neuroligins and the synaptic pathway in Autism Spectrum Disorder. Neurosci Biobehav Rev 2020; 119:37-51. [PMID: 32991906 DOI: 10.1016/j.neubiorev.2020.09.017] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/11/2020] [Accepted: 09/19/2020] [Indexed: 12/13/2022]
Abstract
The genetics underlying autism spectrum disorder (ASD) is complex and heterogeneous, and de novo variants are found in genes converging in functional biological processes. Neuronal communication, including trans-synaptic signaling involving two families of cell-adhesion proteins, the presynaptic neurexins and the postsynaptic neuroligins, is one of the most recurrently affected pathways in ASD. Given the role of these proteins in determining synaptic function, abnormal synaptic plasticity and failure to establish proper synaptic contacts might represent mechanisms underlying risk of ASD. More than 30 mutations have been found in the neuroligin genes. Most of the resulting residue substitutions map in the extracellular, cholinesterase-like domain of the protein, and impair protein folding and trafficking. Conversely, the stalk and intracellular domains are less affected. Accordingly, several genetic animal models of ASD have been generated, showing behavioral and synaptic alterations. The aim of this review is to discuss the current knowledge on ASD-linked mutations in the neuroligin proteins and their effect on synaptic function, in various brain areas and circuits.
Collapse
Affiliation(s)
- Laura Trobiani
- Dept. Biology and Biotechnology, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy
| | - Maria Meringolo
- Lab. Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 64, 00143 Rome, Italy; Dept. Systems Medicine, University Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Tamara Diamanti
- Dept. Biology and Biotechnology, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy
| | - Yves Bourne
- Lab. "Architecture et Fonction des Macromolécules Biologiques", CNRS/Aix Marseille Univ, Faculté des Sciences - Campus Luminy, 163 Avenue de Luminy, 13288 Marseille cedex 09, France
| | - Pascale Marchot
- Lab. "Architecture et Fonction des Macromolécules Biologiques", CNRS/Aix Marseille Univ, Faculté des Sciences - Campus Luminy, 163 Avenue de Luminy, 13288 Marseille cedex 09, France
| | - Giuseppina Martella
- Lab. Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 64, 00143 Rome, Italy; Dept. Systems Medicine, University Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Luciana Dini
- Dept. Biology and Biotechnology, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy
| | - Antonio Pisani
- Lab. Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 64, 00143 Rome, Italy; Dept. Systems Medicine, University Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Antonella De Jaco
- Dept. Biology and Biotechnology, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy.
| | - Paola Bonsi
- Lab. Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 64, 00143 Rome, Italy.
| |
Collapse
|
7
|
Hu Z, Xiao X, Zhang Z, Li M. Genetic insights and neurobiological implications from NRXN1 in neuropsychiatric disorders. Mol Psychiatry 2019; 24:1400-1414. [PMID: 31138894 DOI: 10.1038/s41380-019-0438-9] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 03/31/2019] [Accepted: 04/29/2019] [Indexed: 02/08/2023]
Abstract
Many neuropsychiatric and neurodevelopmental disorders commonly share genetic risk factors. To date, the mechanisms driving the pathogenesis of these disorders, particularly how genetic variations affect the function of risk genes and contribute to disease symptoms, remain largely unknown. Neurexins are a family of synaptic adhesion molecules, which play important roles in the formation and establishment of synaptic structure, as well as maintenance of synaptic function. Accumulating genomic findings reveal that genetic variations within genes encoding neurexins are associated with a variety of psychiatric conditions such as schizophrenia, autism spectrum disorder, and some developmental abnormalities. In this review, we focus on NRXN1, one of the most compelling psychiatric risk genes of the neurexin family. We performed a comprehensive survey and analysis of current genetic and molecular data including both common and rare alleles within NRXN1 associated with psychiatric illnesses, thus providing insights into the genetic risk conferred by NRXN1. We also summarized the neurobiological evidences, supporting the function of NRXN1 and its protein products in synaptic formation, organization, transmission and plasticity, as well as disease-relevant behaviors, and assessed the mechanistic link between the mutations of NRXN1 and synaptic and behavioral pathology in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Zhonghua Hu
- Institute of Molecular Precision Medicine and Hunan Key Laboratory of Molecular Precision Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China. .,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China. .,Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China. .,Hunan Key Laboratory of Animal Models for Human Diseases, School of Life Sciences, Central South University, Changsha, Hunan, China. .,Department of Psychiatry, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China. .,National Clinical Research Center on Mental Disorders, Changsha, Hunan, China.
| | - Xiao Xiao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Zhuohua Zhang
- Institute of Molecular Precision Medicine and Hunan Key Laboratory of Molecular Precision Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Animal Models for Human Diseases, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Ming Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China. .,CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
8
|
Wu X, Morishita WK, Riley AM, Hale WD, Südhof TC, Malenka RC. Neuroligin-1 Signaling Controls LTP and NMDA Receptors by Distinct Molecular Pathways. Neuron 2019; 102:621-635.e3. [PMID: 30871858 PMCID: PMC6509009 DOI: 10.1016/j.neuron.2019.02.013] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 12/17/2018] [Accepted: 02/07/2019] [Indexed: 12/27/2022]
Abstract
Neuroligins, postsynaptic cell adhesion molecules that are linked to neuropsychiatric disorders, are extensively studied, but fundamental questions about their functions remain. Using in vivo replacement strategies in quadruple conditional knockout mice of all neuroligins to avoid heterodimerization artifacts, we show, in hippocampal CA1 pyramidal neurons, that neuroligin-1 performs two key functions in excitatory synapses by distinct molecular mechanisms. N-methyl-D-aspartate (NMDA) receptor-dependent LTP requires trans-synaptic binding of postsynaptic neuroligin-1 to presynaptic β-neurexins but not the cytoplasmic sequences of neuroligins. In contrast, postsynaptic NMDA receptor (NMDAR)-mediated responses involve a neurexin-independent mechanism that requires the neuroligin-1 cytoplasmic sequences. Strikingly, deletion of neuroligins blocked the spine expansion associated with LTP, as monitored by two-photon imaging; this block involved a mechanism identical to that of the role of neuroligin-1 in NMDAR-dependent LTP. Our data suggest that neuroligin-1 performs two mechanistically distinct signaling functions and that neurolign-1-mediated trans-synaptic cell adhesion signaling critically regulates LTP.
Collapse
Affiliation(s)
- Xiaoting Wu
- Nancy Pritzker Laboratory, Dept. of Psychiatry & Behavioral Sciences, Stanford University School of Medicine, Stanford, CA. 94305,Department of Molecular and Cellular Physiology, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA. 94305
| | - Wade K. Morishita
- Nancy Pritzker Laboratory, Dept. of Psychiatry & Behavioral Sciences, Stanford University School of Medicine, Stanford, CA. 94305
| | - Ashley M. Riley
- Nancy Pritzker Laboratory, Dept. of Psychiatry & Behavioral Sciences, Stanford University School of Medicine, Stanford, CA. 94305,Department of Molecular and Cellular Physiology, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA. 94305
| | - William D. Hale
- Department of Molecular and Cellular Physiology, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA. 94305
| | - Thomas C. Südhof
- Department of Molecular and Cellular Physiology, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA. 94305,Co-corresponding authors: R. Malenka; T. Südhof, Stanford University School of Medicine, 265 Campus Drive, Room G1021, G1022, Stanford, CA 94305-5453, 650-724-2730; ;
| | - Robert C. Malenka
- Nancy Pritzker Laboratory, Dept. of Psychiatry & Behavioral Sciences, Stanford University School of Medicine, Stanford, CA. 94305,Co-corresponding authors: R. Malenka; T. Südhof, Stanford University School of Medicine, 265 Campus Drive, Room G1021, G1022, Stanford, CA 94305-5453, 650-724-2730; ;
| |
Collapse
|
9
|
Liu R, Qin XP, Zhuang Y, Zhang Y, Liao HB, Tang JC, Pan MX, Zeng FF, Lei Y, Lei RX, Wang S, Liu AC, Chen J, Zhang ZF, Zhao D, Wu SL, Liu RZ, Wang ZF, Wan Q. Glioblastoma recurrence correlates with NLGN3 levels. Cancer Med 2018; 7:2848-2859. [PMID: 29777576 PMCID: PMC6051187 DOI: 10.1002/cam4.1538] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 04/05/2018] [Accepted: 04/11/2018] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma (GBM) is the most aggressive glioma in the brain. Recurrence of GBM is almost inevitable within a short term after tumor resection. In a retrospective study of 386 cases of GBM collected between 2013 and 2016, we found that recurrence of GBM mainly occurs in the deep brain regions, including the basal ganglia, thalamus, and corpus callosum. But the mechanism underlying this phenomenon is not clear. Previous studies suggest that neuroligin‐3 (NLGN3) is necessary for GBM growth. Our results show that the levels of NLGN3 in the cortex are higher than those in the deep regions in a normal human brain, and similar patterns are also found in a normal mouse brain. In contrast, NLGN3 levels in the deep brain regions of GBM patients are high. We also show that an increase in NLGN3 concentration promotes the growth of U251 cells and U87‐MG cells. Respective use of the cortex neuron culture medium (C‐NCM) and basal ganglia neuron culture medium (BG‐NCM) with DMEM to cultivate U251, U87‐MG and GBM cells isolated from patients, we found that these cells grew faster after treatment with C‐NCM and BG‐NCM in which the cells treated with C‐NCM grew faster than the ones treated with BG‐NCM group. Inhibition of NLGN3 release by ADAM10i prevents NCM‐induced cell growth. Together, this study suggests that increased levels of NLGN3 in the deep brain region under the GBM pathological circumstances may contribute to GBM recurrence in the basal ganglia, thalamus, and corpus callosum.
Collapse
Affiliation(s)
- Rui Liu
- Department of Physiology, Collaborative Innovation Center for Brain Science, School of Basic Medical Sciences, School of Medicine, Wuhan University, Wuhan, China
| | - Xing-Ping Qin
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yang Zhuang
- Department of Physiology, Collaborative Innovation Center for Brain Science, School of Basic Medical Sciences, School of Medicine, Wuhan University, Wuhan, China
| | - Ya Zhang
- Department of Physiology, Collaborative Innovation Center for Brain Science, School of Basic Medical Sciences, School of Medicine, Wuhan University, Wuhan, China
| | - Hua-Bao Liao
- Department of Physiology, Collaborative Innovation Center for Brain Science, School of Basic Medical Sciences, School of Medicine, Wuhan University, Wuhan, China
| | - Jun-Chun Tang
- Department of Physiology, Collaborative Innovation Center for Brain Science, School of Basic Medical Sciences, School of Medicine, Wuhan University, Wuhan, China
| | - Meng-Xian Pan
- Department of Physiology, Collaborative Innovation Center for Brain Science, School of Basic Medical Sciences, School of Medicine, Wuhan University, Wuhan, China
| | - Fei-Fei Zeng
- Department of Radiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yang Lei
- Department of Physiology, Collaborative Innovation Center for Brain Science, School of Basic Medical Sciences, School of Medicine, Wuhan University, Wuhan, China
| | - Rui-Xue Lei
- Department of Physiology, Collaborative Innovation Center for Brain Science, School of Basic Medical Sciences, School of Medicine, Wuhan University, Wuhan, China
| | - Shu Wang
- Department of Physiology, Collaborative Innovation Center for Brain Science, School of Basic Medical Sciences, School of Medicine, Wuhan University, Wuhan, China
| | - An-Chun Liu
- Department of Physiology, Collaborative Innovation Center for Brain Science, School of Basic Medical Sciences, School of Medicine, Wuhan University, Wuhan, China
| | - Juan Chen
- Department of Neurology, the Central Hospital of Wuhan, Tongji Medical College of Huazhong University of Science & Technology, Wuhan, China
| | - Zhi-Feng Zhang
- Department of Physiology, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China
| | - Dan Zhao
- Department of Physiology, Collaborative Innovation Center for Brain Science, School of Basic Medical Sciences, School of Medicine, Wuhan University, Wuhan, China.,Department of Physiology, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China
| | - Song-Lin Wu
- Department of Geriatrics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ren-Zhong Liu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ze-Fen Wang
- Department of Physiology, Collaborative Innovation Center for Brain Science, School of Basic Medical Sciences, School of Medicine, Wuhan University, Wuhan, China
| | - Qi Wan
- Department of Physiology, Collaborative Innovation Center for Brain Science, School of Basic Medical Sciences, School of Medicine, Wuhan University, Wuhan, China.,Institute of Neuroregeneration & Neurorehabilitation, Department of Neurosurgery of the Affiliated Hospital, Qingdao University, Qingdao, China
| |
Collapse
|
10
|
Bourne Y, Marchot P. Hot Spots for Protein Partnerships at the Surface of Cholinesterases and Related α/β Hydrolase Fold Proteins or Domains-A Structural Perspective. Molecules 2017; 23:molecules23010035. [PMID: 29295471 PMCID: PMC5943944 DOI: 10.3390/molecules23010035] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 12/21/2017] [Accepted: 12/21/2017] [Indexed: 12/12/2022] Open
Abstract
The hydrolytic enzymes acetyl- and butyryl-cholinesterase, the cell adhesion molecules neuroligins, and the hormonogenic macromolecule thyroglobulin are a few of the many members of the α/β hydrolase fold superfamily of proteins. Despite their distinctive functions, their canonical subunits, with a molecular surface area of ~20,000 Å2, they share binding patches and determinants for forming homodimers and for accommodating structural subunits or protein partners. Several of these surface regions of high functional relevance have been mapped through structural or mutational studies, while others have been proposed based on biochemical data or molecular docking studies. Here, we review these binding interfaces and emphasize their specificity versus potentially multifunctional character.
Collapse
Affiliation(s)
- Yves Bourne
- Centre National de la Recherche Scientifique, Aix-Marseille Université, "Architecture et Fonction des Macromolécules Biologiques" Laboratory, 13288 Marseille, France.
| | - Pascale Marchot
- Centre National de la Recherche Scientifique, Aix-Marseille Université, "Architecture et Fonction des Macromolécules Biologiques" Laboratory, 13288 Marseille, France.
| |
Collapse
|
11
|
Hybrid Applications of Solution Scattering to Aid Structural Biology. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1009:215-227. [PMID: 29218562 DOI: 10.1007/978-981-10-6038-0_13] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Biomolecular applications of solution X-ray and neutron scattering (SAXS and SANS, respectively) started in late 1960s - early 1970s but were relatively limited in their ability to provide a detailed structural picture and lagged behind what became the two primary methods of experimental structural biology - X-ray crystallography and NMR. However, improvements in both data analysis and instrumentation led to an explosive growth in the number of studies that used small-angle scattering (SAS) for investigation of macromolecular structure, often in combination with other biophysical techniques. Such hybrid applications are nowadays quickly becoming a norm whenever scattering data are used for two reasons. First, it is generally accepted that SAS data on their own cannot lead to a uniquely defined high-resolution structural model, creating a need for supplementing them with information from complementary techniques. Second, solution scattering data are frequently applied in situations when a method such NMR or X-ray crystallography cannot provide a satisfactory structural picture, which makes these additional restraints highly desirable. Maturation of the hybrid bio-SAS approaches brings to light new questions including completeness of the conformational space sampling, model validation, and data compatibility.
Collapse
|
12
|
Palamini M, Canciani A, Forneris F. Identifying and Visualizing Macromolecular Flexibility in Structural Biology. Front Mol Biosci 2016; 3:47. [PMID: 27668215 PMCID: PMC5016524 DOI: 10.3389/fmolb.2016.00047] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 08/22/2016] [Indexed: 12/29/2022] Open
Abstract
Structural biology comprises a variety of tools to obtain atomic resolution data for the investigation of macromolecules. Conventional structural methodologies including crystallography, NMR and electron microscopy often do not provide sufficient details concerning flexibility and dynamics, even though these aspects are critical for the physiological functions of the systems under investigation. However, the increasing complexity of the molecules studied by structural biology (including large macromolecular assemblies, integral membrane proteins, intrinsically disordered systems, and folding intermediates) continuously demands in-depth analyses of the roles of flexibility and conformational specificity involved in interactions with ligands and inhibitors. The intrinsic difficulties in capturing often subtle but critical molecular motions in biological systems have restrained the investigation of flexible molecules into a small niche of structural biology. Introduction of massive technological developments over the recent years, which include time-resolved studies, solution X-ray scattering, and new detectors for cryo-electron microscopy, have pushed the limits of structural investigation of flexible systems far beyond traditional approaches of NMR analysis. By integrating these modern methods with powerful biophysical and computational approaches such as generation of ensembles of molecular models and selective particle picking in electron microscopy, more feasible investigations of dynamic systems are now possible. Using some prominent examples from recent literature, we review how current structural biology methods can contribute useful data to accurately visualize flexibility in macromolecular structures and understand its important roles in regulation of biological processes.
Collapse
Affiliation(s)
| | | | - Federico Forneris
- The Armenise-Harvard Laboratory of Structural Biology, Department of Biology and Biotechnology, University of PaviaPavia, Italy
| |
Collapse
|
13
|
Rubio-Marrero EN, Vincelli G, Jeffries CM, Shaikh TR, Pakos IS, Ranaivoson FM, von Daake S, Demeler B, De Jaco A, Perkins G, Ellisman MH, Trewhella J, Comoletti D. Structural Characterization of the Extracellular Domain of CASPR2 and Insights into Its Association with the Novel Ligand Contactin1. J Biol Chem 2016; 291:5788-5802. [PMID: 26721881 PMCID: PMC4786715 DOI: 10.1074/jbc.m115.705681] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 12/28/2015] [Indexed: 01/06/2023] Open
Abstract
Contactin-associated protein-like 2 (CNTNAP2) encodes for CASPR2, a multidomain single transmembrane protein belonging to the neurexin superfamily that has been implicated in a broad range of human phenotypes including autism and language impairment. Using a combination of biophysical techniques, including small angle x-ray scattering, single particle electron microscopy, analytical ultracentrifugation, and bio-layer interferometry, we present novel structural and functional data that relate the architecture of the extracellular domain of CASPR2 to a previously unknown ligand, Contactin1 (CNTN1). Structurally, CASPR2 is highly glycosylated and has an overall compact architecture. Functionally, we show that CASPR2 associates with micromolar affinity with CNTN1 but, under the same conditions, it does not interact with any of the other members of the contactin family. Moreover, by using dissociated hippocampal neurons we show that microbeads loaded with CASPR2, but not with a deletion mutant, co-localize with transfected CNTN1, suggesting that CNTN1 is an endogenous ligand for CASPR2. These data provide novel insights into the structure and function of CASPR2, suggesting a complex role of CASPR2 in the nervous system.
Collapse
Affiliation(s)
- Eva N Rubio-Marrero
- From the Child Health Institute of New Jersey and Departments of Neuroscience and Cell Biology and
| | - Gabriele Vincelli
- From the Child Health Institute of New Jersey and Departments of Neuroscience and Cell Biology and
| | - Cy M Jeffries
- the School of Molecular Bioscience, University of Sydney, New South Wales 2006, Australia
| | - Tanvir R Shaikh
- the Structural Biology Programme, Central European Institute of Technology, Masaryk University, 62500 Brno, Czech Republic
| | - Irene S Pakos
- From the Child Health Institute of New Jersey and Departments of Neuroscience and Cell Biology and
| | - Fanomezana M Ranaivoson
- From the Child Health Institute of New Jersey and Departments of Neuroscience and Cell Biology and
| | - Sventja von Daake
- From the Child Health Institute of New Jersey and Departments of Neuroscience and Cell Biology and
| | - Borries Demeler
- the Department of Biochemistry, The University of Texas Health Science Center, San Antonio, Texas 78229
| | - Antonella De Jaco
- the Department of Biology and Biotechnologies "Charles Darwin" and Pasteur Institute-Cenci Bolognetti Foundation, Sapienza University of Rome, Rome, Italy 00185
| | - Guy Perkins
- the National Center for Microscopy and Imaging Research, University of California San Diego, La Jolla, California 92093, and
| | - Mark H Ellisman
- the National Center for Microscopy and Imaging Research, University of California San Diego, La Jolla, California 92093, and
| | - Jill Trewhella
- the School of Molecular Bioscience, University of Sydney, New South Wales 2006, Australia,; the Department of Chemistry, University of Utah, Salt Lake City, Utah 84112
| | - Davide Comoletti
- From the Child Health Institute of New Jersey and Departments of Neuroscience and Cell Biology and; Pediatrics, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey 08901,.
| |
Collapse
|
14
|
Abstract
A fundamental physical interaction exists across the synapse. It is mediated by synaptic adhesion molecules, and is among the earliest and most indispensable of molecular events occurring during synaptogenesis. The regulation of adhesion molecules and their interactions with other synaptic proteins likely affect not only on synapse formation but also on ongoing synaptic function. We review research on one major family of postsynaptic adhesion molecules, neuroligins, which bind to their presynaptic partner neurexin across the synaptic cleft. We move from a structural overview to the broad cellular and synaptic context of neuroligins, intermolecular interactions, and molecular modifications that occur within a synapse. Finally, we examine evidence concerning the physiological functions of neuroligin in a cell and highlight areas requiring further investigation.
Collapse
Affiliation(s)
- Michael A Bemben
- Receptor Biology Section, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health, Bethesda, MD 20892, USA; Department of Biology, The Johns Hopkins University, 3400 North Charles Street, Baltimore, MD, 21218, USA
| | - Seth L Shipman
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Roger A Nicoll
- Departments of Cellular and Molecular Pharmacology and Physiology, University of California, San Francisco, San Francisco, CA 94158, USA.
| | - Katherine W Roche
- Receptor Biology Section, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
15
|
Chaudhuri BN. Emerging applications of small angle solution scattering in structural biology. Protein Sci 2015; 24:267-76. [PMID: 25516491 PMCID: PMC4353354 DOI: 10.1002/pro.2624] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Accepted: 12/05/2014] [Indexed: 12/12/2022]
Abstract
Small angle solution X-ray and neutron scattering recently resurfaced as powerful tools to address an array of biological problems including folding, intrinsic disorder, conformational transitions, macromolecular crowding, and self or hetero-assembling of biomacromolecules. In addition, small angle solution scattering complements crystallography, nuclear magnetic resonance spectroscopy, and other structural methods to aid in the structure determinations of multidomain or multicomponent proteins or nucleoprotein assemblies. Neutron scattering with hydrogen/deuterium contrast variation, or X-ray scattering with sucrose contrast variation to a certain extent, is a convenient tool for characterizing the organizations of two-component systems such as a nucleoprotein or a lipid-protein assembly. Time-resolved small and wide-angle solution scattering to study biological processes in real time, and the use of localized heavy-atom labeling and anomalous solution scattering for applications as FRET-like molecular rulers, are amongst promising newer developments. Despite the challenges in data analysis and interpretation, these X-ray/neutron solution scattering based approaches hold great promise for understanding a wide variety of complex processes prevalent in the biological milieu.
Collapse
Affiliation(s)
- Barnali N Chaudhuri
- Faculty of Life Sciences and Biotechnology, South Asian UniversityAkbar Bhawan, Chanakyapuri, New Delhi, India
| |
Collapse
|
16
|
The cancer glycocalyx mechanically primes integrin-mediated growth and survival. Nature 2014; 511:319-25. [PMID: 25030168 DOI: 10.1038/nature13535] [Citation(s) in RCA: 522] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2013] [Accepted: 05/23/2014] [Indexed: 12/27/2022]
Abstract
Malignancy is associated with altered expression of glycans and glycoproteins that contribute to the cellular glycocalyx. We constructed a glycoprotein expression signature, which revealed that metastatic tumours upregulate expression of bulky glycoproteins. A computational model predicted that these glycoproteins would influence transmembrane receptor spatial organization and function. We tested this prediction by investigating whether bulky glycoproteins in the glycocalyx promote a tumour phenotype in human cells by increasing integrin adhesion and signalling. Our data revealed that a bulky glycocalyx facilitates integrin clustering by funnelling active integrins into adhesions and altering integrin state by applying tension to matrix-bound integrins, independent of actomyosin contractility. Expression of large tumour-associated glycoproteins in non-transformed mammary cells promoted focal adhesion assembly and facilitated integrin-dependent growth factor signalling to support cell growth and survival. Clinical studies revealed that large glycoproteins are abundantly expressed on circulating tumour cells from patients with advanced disease. Thus, a bulky glycocalyx is a feature of tumour cells that could foster metastasis by mechanically enhancing cell-surface receptor function.
Collapse
|
17
|
O'Sullivan ML, Martini F, von Daake S, Comoletti D, Ghosh A. LPHN3, a presynaptic adhesion-GPCR implicated in ADHD, regulates the strength of neocortical layer 2/3 synaptic input to layer 5. Neural Dev 2014; 9:7. [PMID: 24739570 PMCID: PMC3996519 DOI: 10.1186/1749-8104-9-7] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 03/31/2014] [Indexed: 11/18/2022] Open
Abstract
Background Latrophilins (LPHNs) are a small family of neuronal adhesion-GPCRs originally discovered as receptors for the black widow spider toxin α-latrotoxin. Mutations in LPHN3 have recently been identified as risk factors for attention deficit hyperactivity disorder (ADHD) in humans, but their physiological function has remained elusive. In this study, we tested two hypotheses regarding LPHN3 function: (1) LPHN3 regulates synaptic transmission by modulating probability of release; and (2) LPHN3 controls synapse development and the abundance of synapses. Results We manipulated LPHN3 expression in mouse layer 2/3 (L2/3) pyramidal neurons and examined the consequences on the L2/3 to L5 cortical microcircuit. Employing an optogenetic strategy combined with shRNA knockdown of LPHN3, we found that LPHN3 did not influence probability of release at synapses formed by L2/3 neurons onto L5 pyramidal cells. The strength of L2/3 afferent input to L5, however, was weakened by loss of LPHN3. Using Synaptophysin-GFP as an anatomical marker of presynaptic terminals, we found that the density of synapses formed by L2/3 axons in L5 was reduced when LPHN3 was lost. Finally, we investigated the structural organization of the extracellular domain of LPHN3. We used single particle negative stain electron microscopy to image the extracellular domain of LPHN3 and showed that the Olfactomedin and Lectin domains form a globular domain on an elongated stalk. Cell-based binding experiments with mutant proteins revealed that the Olfactomedin domain was required for binding to FLRT3, whereas both the Olfactomedin and Lectin domains were involved in binding to Teneurin 1. Mutant LPHN3 lacking the Olfactomedin domain was not capable of rescuing the deficit in presynaptic density following knockdown of endogenous LPHN3. Conclusions We find that LPHN3 regulates the number of synapses formed by L2/3 neurons in L5 and the strength of synaptic drive from the L2/3-L5 pathway. The Olfactomedin domain of LPHN3 is required for this effect on synapse number and binding to its postsynaptic ligand FLRT3. We propose that LPHN3 functions in synaptic development and is important in determining the connectivity rates between principal neurons in the cortex.
Collapse
Affiliation(s)
| | | | | | | | - Anirvan Ghosh
- Neurobiology Section, Division of Biology, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
18
|
The neuroligins and their ligands: from structure to function at the synapse. J Mol Neurosci 2014; 53:387-96. [PMID: 24497299 DOI: 10.1007/s12031-014-0234-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 01/10/2014] [Indexed: 10/25/2022]
Abstract
The neuroligins are cell adhesion proteins whose extracellular domain belongs to the α/β-hydrolase fold family of proteins, mainly containing enzymes and exemplified by acetylcholinesterase. The ectodomain of postsynaptic neuroligins interacts through a calcium ion with the ectodomain of presynaptic neurexins to form flexible trans-synaptic associations characterized by selectivity for neuroligin or neurexin subtypes. This heterophilic interaction, essential for synaptic differentiation, maturation, and maintenance, is regulated by gene selection, alternative mRNA splicing, and posttranslational modifications. Mutations leading to deficiencies in the expression, folding, maturation, and binding properties of either partner are associated with autism spectrum disorders. The currently available structural and functional data illustrate how these two families of cell adhesion molecules bridge the synaptic cleft to participate in synapse plasticity and support its dynamic nature. Neuroligin partners distinct from the neurexins, and which may undergo either trans or cis interaction, have also been described, and tridimensional structures of some of them are available. Our study emphasizes the partnership versatility of the neuroligin ectodomain associated with molecular flexibility and alternative binding sites, proposes homology models of the structurally non-characterized neuroligin partners, and exemplifies the large structural variability at the surface of the α/β-hydrolase fold subunit. This study also provides new insights into possible surface binding sites associated with non-catalytic properties of the acetylcholinesterase subunit.
Collapse
|
19
|
Ankner JF, Heller WT, Herwig KW, Meilleur F, Myles DAA. Neutron scattering techniques and applications in structural biology. ACTA ACUST UNITED AC 2013; Chapter 17:Unit17.16. [PMID: 23546619 DOI: 10.1002/0471140864.ps1716s72] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neutron scattering is exquisitely sensitive to the position, concentration, and dynamics of hydrogen atoms in materials and is a powerful tool for the characterization of structure-function and interfacial relationships in biological systems. Modern neutron scattering facilities offer access to a sophisticated, nondestructive suite of instruments for biophysical characterization that provides spatial and dynamic information spanning from Ångstroms to microns and from picoseconds to microseconds, respectively. Applications in structural biology range from the atomic-resolution analysis of individual hydrogen atoms in enzymes through to meso- and macro-scale analysis of complex biological structures, membranes, and assemblies. The large difference in neutron scattering length between hydrogen and deuterium allows contrast variation experiments to be performed and enables H/D isotopic labeling to be used for selective and systematic analysis of the local structure, dynamics, and interactions of multi-component systems. This overview describes the available techniques and summarizes their practical application to the study of biomolecular systems.
Collapse
Affiliation(s)
- John F Ankner
- Neutron Sciences Directorate, Oak Ridge National Laboratory, Oak Ridge, Tennessee, USA
| | | | | | | | | |
Collapse
|
20
|
Specific trans-synaptic interaction with inhibitory interneuronal neurexin underlies differential ability of neuroligins to induce functional inhibitory synapses. J Neurosci 2013; 33:3612-23. [PMID: 23426688 DOI: 10.1523/jneurosci.1811-12.2013] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Synaptic transmission depends on the matching and alignment of presynaptically released transmitters and postsynaptic neurotransmitter receptors. Neuroligin (NL) and Neurexin (Nrxn) proteins are trans-synaptic adhesion molecules that are important in validation and maturation of specific synapses. NL isoforms NL1 and NL2 have specific functional roles in excitatory and inhibitory synapses, respectively, but the molecular basis behind this distinction is still unclear. We show here that the extracellular domain of NL2 confers its unique ability to enhance inhibitory synaptic function when overexpressed in rat hippocampal pyramidal neurons, whereas NL1 normally only promotes excitatory synapses. This specificity is conferred by presynaptic Nrxn isoforms, as NL1 can also induce functional inhibitory synapse connections when the presynaptic interneurons ectopically express an Nrxn isoform that binds to NL1. Our results indicate that trans-synaptic interaction with differentially expressed presynaptic Nrxns underlies the distinct functions of NL1 and NL2, and is sufficient to induce functional inhibitory synapse formation.
Collapse
|
21
|
Bang ML, Owczarek S. A Matter of Balance: Role of Neurexin and Neuroligin at the Synapse. Neurochem Res 2013; 38:1174-89. [DOI: 10.1007/s11064-013-1029-9] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Revised: 02/01/2013] [Accepted: 03/26/2013] [Indexed: 11/29/2022]
|
22
|
Dimerization of postsynaptic neuroligin drives synaptic assembly via transsynaptic clustering of neurexin. Proc Natl Acad Sci U S A 2012; 109:19432-7. [PMID: 23129658 DOI: 10.1073/pnas.1217633109] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The transsynaptic complex of neuroligin (NLGN) and neurexin forms a physical connection between pre- and postsynaptic neurons that occurs early in the course of new synapse assembly. Both neuroligin and neurexin have, indeed, been proposed to exhibit active, instructive roles in the formation of synapses. However, the process by which these instructive roles play out during synaptogenesis is not well understood. Here, we examine one aspect of postsynaptic neuroligin with regard to its synaptogenic properties: its basal state as a constitutive dimer. We show that dimerization is required for the synaptogenic properties of neuroligin and likely serves to induce presynaptic differentiation via a transsynaptic clustering of neurexin. Further, we introduce chemically inducible, exogenous dimerization domains to the neuroligin molecule, effectively bestowing chemical control of neuroligin dimerization. This allows us to identify the acute requirements of neuroligin dimerization by chemically manipulating the monomeric-to-dimeric conversion of neuroligin. Based on the results of the inducible dimerization experiments, we propose a model in which dimerized neuroligin induces the mechanical clustering of presynaptic molecules as part of a requisite step in the coordinated assembly of a chemical synapse.
Collapse
|
23
|
The role of neurexins and neuroligins in the formation, maturation, and function of vertebrate synapses. Curr Opin Neurobiol 2012; 22:412-22. [PMID: 22424845 DOI: 10.1016/j.conb.2012.02.012] [Citation(s) in RCA: 184] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2011] [Accepted: 02/23/2012] [Indexed: 11/20/2022]
Abstract
Neurexins (NXs) and neuroligins (NLs) are transsynaptically interacting cell adhesion proteins that play a key role in the formation, maturation, activity-dependent validation, and maintenance of synapses. As complex alternative splicing processes in nerve cells generate a large number of NX and NLs variants, it has been proposed that a combinatorial interaction code generated by these variants may determine synapse identity and network connectivity during brain development. The functional importance of NXs and NLs is exemplified by the fact that mutations in NX and NL genes are associated with several neuropsychiatric disorders, most notably with autism. Accordingly, major research efforts have focused on the molecular mechanisms by which NXs and NLs operate at synapses. In this review, we summarize recent progress in this field and discuss emerging topics, such as the role of alternative interaction partners of NXs and NLs in synapse formation and function, and their relevance for synaptic plasticity in the mature brain. The novel findings highlight the fundamental importance of NX-NL interactions in a wide range of synaptic functions.
Collapse
|
24
|
Bottos A, Rissone A, Bussolino F, Arese M. Neurexins and neuroligins: synapses look out of the nervous system. Cell Mol Life Sci 2011; 68:2655-66. [PMID: 21394644 PMCID: PMC11115133 DOI: 10.1007/s00018-011-0664-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2010] [Revised: 02/16/2011] [Accepted: 02/22/2011] [Indexed: 12/18/2022]
Abstract
The scientific interest in the family of the so-called nervous vascular parallels has been growing steadily for the past 15 years, either by addition of new members to the group or, lately, by deepening the analysis of established concepts and mediators. Proteins governing both neurons and vascular cells are known to be involved in events such as cell fate determination and migration/guidance but not in the last and apparently most complex step of nervous system development, the formation and maturation of synapses. Hence, the recent addition to this family of the specific synaptic proteins, Neurexin and Neuroligin, is a double innovation. The two proteins, which were thought to be "simple" adhesive links between the pre- and post-synaptic sides of chemical synapses, are in fact extremely complex and modulate the most subtle synaptic activities. We will discuss the relevant data and the intriguing challenge of transferring synaptic activities to vascular functions.
Collapse
Affiliation(s)
- Alessia Bottos
- Department of Oncological Sciences, University of Torino, IRCC, Institute for Cancer Research and Treatment at Candiolo, Strada prov 142, km 3, 95, 10060 Candiolo (TO), Italy
| | - Alberto Rissone
- Department of Oncological Sciences, University of Torino, IRCC, Institute for Cancer Research and Treatment at Candiolo, Strada prov 142, km 3, 95, 10060 Candiolo (TO), Italy
| | - Federico Bussolino
- Department of Oncological Sciences, University of Torino, IRCC, Institute for Cancer Research and Treatment at Candiolo, Strada prov 142, km 3, 95, 10060 Candiolo (TO), Italy
| | - Marco Arese
- Department of Oncological Sciences, University of Torino, IRCC, Institute for Cancer Research and Treatment at Candiolo, Strada prov 142, km 3, 95, 10060 Candiolo (TO), Italy
| |
Collapse
|
25
|
Miller MT, Mileni M, Comoletti D, Stevens RC, Harel M, Taylor P. The crystal structure of the α-neurexin-1 extracellular region reveals a hinge point for mediating synaptic adhesion and function. Structure 2011; 19:767-78. [PMID: 21620717 PMCID: PMC3279696 DOI: 10.1016/j.str.2011.03.011] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Revised: 03/09/2011] [Accepted: 03/13/2011] [Indexed: 10/18/2022]
Abstract
α- and β-neurexins (NRXNs) are transmembrane cell adhesion proteins that localize to presynaptic membranes in neurons and interact with the postsynaptic neuroligins (NLGNs). Their gene mutations are associated with the autism spectrum disorders. The extracellular region of α-NRXNs, containing nine independently folded domains, has structural complexity and unique functional characteristics, distinguishing it from the smaller β-NRXNs. We have solved the X-ray crystal structure of seven contiguous domains of the α-NRXN-1 extracellular region at 3.0 Å resolution. The structure reveals an arrangement where the N-terminal five domains adopt a more rigid linear conformation and the two C-terminal domains form a separate arm connected by a flexible hinge. In an extended conformation the molecule is suitably configured to accommodate a bound NLGN molecule, as supported by structural comparison and surface plasmon resonance. These studies provide the structural basis for a multifunctional synaptic adhesion complex mediated by α-NRXN-1.
Collapse
Affiliation(s)
- Meghan T. Miller
- Department of Pharmacology, Skaggs School of Pharmacy and Pharmaceutical Sciences, La Jolla, CA 92093, USA
| | - Mauro Mileni
- Department of Molecular Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Davide Comoletti
- Department of Pharmacology, Skaggs School of Pharmacy and Pharmaceutical Sciences, La Jolla, CA 92093, USA
| | - Raymond C. Stevens
- Department of Molecular Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Michal Harel
- Department of Pharmacology, Skaggs School of Pharmacy and Pharmaceutical Sciences, La Jolla, CA 92093, USA
- Department of Structural Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Palmer Taylor
- Department of Pharmacology, Skaggs School of Pharmacy and Pharmaceutical Sciences, La Jolla, CA 92093, USA
| |
Collapse
|
26
|
Varley ZK, Pizzarelli R, Antonelli R, Stancheva SH, Kneussel M, Cherubini E, Zacchi P. Gephyrin regulates GABAergic and glutamatergic synaptic transmission in hippocampal cell cultures. J Biol Chem 2011; 286:20942-51. [PMID: 21507951 DOI: 10.1074/jbc.m111.234641] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Gephyrin is a scaffold protein essential for stabilizing glycine and GABA(A) receptors at inhibitory synapses. Here, recombinant intrabodies against gephyrin (scFv-gephyrin) were used to assess whether this protein exerts a transynaptic action on GABA and glutamate release. Pair recordings from interconnected hippocampal cells in culture revealed a reduced probability of GABA release in scFv-gephyrin-transfected neurons compared with controls. This effect was associated with a significant decrease in VGAT, the vesicular GABA transporter, and in neuroligin 2 (NLG2), a protein that, interacting with neurexins, ensures the cross-talk between the post- and presynaptic sites. Interestingly, hampering gephyrin function also produced a significant reduction in VGLUT, the vesicular glutamate transporter, an effect accompanied by a significant decrease in frequency of miniature excitatory postsynaptic currents. Overexpressing NLG2 in gephyrin-deprived neurons rescued GABAergic but not glutamatergic innervation, suggesting that the observed changes in the latter were not due to a homeostatic compensatory mechanism. Pulldown experiments demonstrated that gephyrin interacts not only with NLG2 but also with NLG1, the isoform enriched at excitatory synapses. These results suggest a key role of gephyrin in regulating transynaptic signaling at both inhibitory and excitatory synapses.
Collapse
Affiliation(s)
- Zeynep Kasap Varley
- Neurobiology Department and Italian Institute of Technology Unit, International School for Advanced Studies (SISSA), Via Bonomea 265, 34136 Trieste, Italy
| | | | | | | | | | | | | |
Collapse
|
27
|
Taylor P. From Split to Sibenik: the tortuous pathway in the cholinesterase field. Chem Biol Interact 2010; 187:3-9. [PMID: 20493179 PMCID: PMC3000743 DOI: 10.1016/j.cbi.2010.05.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2010] [Revised: 05/03/2010] [Accepted: 05/07/2010] [Indexed: 11/22/2022]
Abstract
The interim between the first and tenth International Cholinesterase Meetings has seen remarkable advances associated with the applications of structural biology and recombinant DNA methodology to our field. The cloning of the cholinesterase genes led to the identification of a new super family of proteins, termed the alpha,beta-hydrolase fold; members of this family possess a four helix bundle capable of linking structural subunits to the functioning globular protein. Sequence comparisons and three-dimensional structural studies revealed unexpected cousins possessing this fold that, in turn, revealed three distinct functions for the alpha,beta-hydrolase proteins. These encompass: (1) a capacity for hydrolytic cleavage of a great variety of substrates, (2) a heterophilic adhesion function that results in trans-synaptic associations in linked neurons, (3) a chaperone function leading to stabilization of nascent protein and its trafficking to an extracellular or secretory storage location. The analysis and modification of structure may go beyond understanding mechanism, since it may be possible to convert the cholinesterases to efficient detoxifying agents of organophosphatases assisted by added oximes. Also, the study of the relationship between the alpha,beta-hydrolase fold proteins and their biosynthesis may yield means by which aberrant trafficking may be corrected, enhancing expression of mutant proteins. Those engaged in cholinesterase research should take great pride in our accomplishments punctuated by the series of ten meetings. The momentum established and initial studies with related proteins all hold great promise for the future.
Collapse
Affiliation(s)
- Palmer Taylor
- Department of Pharmacology 0636, Skaggs School of Pharmacy & Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093-0650, USA.
| |
Collapse
|
28
|
Structure–function relationships of the α/β-hydrolase fold domain of neuroligin: A comparison with acetylcholinesterase. Chem Biol Interact 2010; 187:49-55. [DOI: 10.1016/j.cbi.2010.01.030] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2009] [Revised: 01/15/2010] [Accepted: 01/18/2010] [Indexed: 11/18/2022]
|
29
|
Lee H, Dean C, Isacoff E. Alternative splicing of neuroligin regulates the rate of presynaptic differentiation. J Neurosci 2010; 30:11435-46. [PMID: 20739565 PMCID: PMC3241922 DOI: 10.1523/jneurosci.2946-10.2010] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2010] [Revised: 06/30/2010] [Accepted: 07/02/2010] [Indexed: 11/21/2022] Open
Abstract
Neuroligins (NLGs) and Neurexins (NRXs) are important adhesion molecules that promote synapse formation. Multiple splice variants of NLG and NRX exist, but their specific functions are unclear. Here we report that a surrogate postsynaptic cell expressing full-length NLG-1 triggers slow presynaptic differentiation in a contacting axon. In contrast, a version of NLG-1, which lacks insert B (NLG-1DeltaB), induces rapid presynaptic differentiation, reaching the rate seen at native neuronal synapses. We show that this acceleration is attributed to the removal of the N-linked glycosylation site within insert B. NLG-1DeltaB also increases synaptic density at neuro-neuronal synapses more than does full-length NLG-1. Other postsynaptic adhesion proteins, such as N-cadherin, EphB2, and SynCAM-1, alone or in combination with full-length NLG-1, do not trigger fast differentiation, suggesting that rapid presynaptic differentiation depends on a unique interaction of NLG-1DeltaB with axonal proteins. Indeed, we find that NLG-1DeltaB recruits more axonal alpha-NRX. Our results suggest that the engagement of alpha-NRX is a key to rapid induction of synapses at new sites of axo-dendritic contact.
Collapse
Affiliation(s)
- Hanson Lee
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California 94720, and
| | - Camin Dean
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California 94720, and
| | - Ehud Isacoff
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California 94720, and
- Physical Bioscience Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720
| |
Collapse
|
30
|
Comoletti D, Miller MT, Jeffries CM, Wilson J, Demeler B, Taylor P, Trewhella J, Nakagawa T. The macromolecular architecture of extracellular domain of alphaNRXN1: domain organization, flexibility, and insights into trans-synaptic disposition. Structure 2010; 18:1044-53. [PMID: 20696403 PMCID: PMC2948785 DOI: 10.1016/j.str.2010.06.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2010] [Revised: 06/14/2010] [Accepted: 06/17/2010] [Indexed: 02/05/2023]
Abstract
Neurexins are multidomain synaptic cell-adhesion proteins that associate with multiple partnering proteins. Genetic evidence indicates that neurexins may contribute to autism, schizophrenia, and nicotine dependence. Using analytical ultracentrifugation, single-particle electron microscopy, and solution X-ray scattering, we obtained a three-dimensional structural model of the entire extracellular domain of neurexin-1alpha. This protein adopts a dimensionally asymmetric conformation that is monomeric in solution, with a maximum dimension of approximately 170 A. The extracellular domain of alpha-neurexin maintains a characteristic "Y" shape, whereby LNS domains 1-4 form an extended base of the "Y" and LNS5-6 the shorter arms. Moreover, two major regions of flexibility are present: one between EGF1 and LNS2, corresponding to splice site 1, another between LNS5 and 6. We thus provide the first structural insights into the architecture of the extracellular region of neurexin-1alpha, show how the protein may fit in the synaptic cleft, and how partnering proteins could bind simultaneously.
Collapse
Affiliation(s)
- Davide Comoletti
- Department of Pharmacology, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Leone P, Comoletti D, Ferracci G, Conrod S, Garcia SU, Taylor P, Bourne Y, Marchot P. Structural insights into the exquisite selectivity of neurexin/neuroligin synaptic interactions. EMBO J 2010; 29:2461-71. [PMID: 20543817 PMCID: PMC2910273 DOI: 10.1038/emboj.2010.123] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2010] [Accepted: 05/17/2010] [Indexed: 11/08/2022] Open
Abstract
The extracellular domains of neuroligins and neurexins interact through Ca(2+) to form flexible trans-synaptic associations characterized by selectivity for neuroligin or neurexin subtypes. This heterophilic interaction, essential for synaptic maturation and differentiation, is regulated by gene selection, alternative mRNA splicing and post-translational modifications. A new, 2.6 A-resolution crystal structure of a soluble neurexin-1beta-neuroligin-4 (Nrx1beta-NL4) complex permits a detailed description of the Ca(2+)-coordinated interface and unveils concerted positional rearrangements of several residues of NL4, not observed in neuroligin-1, associated with Nrx1beta binding. Surface plasmon resonance analysis of the binding of structure-guided Nrx1beta mutants towards NL4 and neuroligin-1 shows that flexibility of the Nrx1beta-binding site in NL4 is reflected in a greater dissociation constant of the complex and higher sensitivity to ionic strength and pH variations. Analysis of neuroligin mutants points to critical functions for two respective residues in neuroligin-1 and neuroligin-2 in governing the affinity of the complexes. Although neuroligin-1 and neuroligin-2 have pre-determined conformations that respectively promote and prevent Nrx1beta association, unique conformational reshaping of the NL4 surface is required to permit Nrx1beta association.
Collapse
Affiliation(s)
- Philippe Leone
- Architecture et Fonction des Macromolécules Biologiques (AFMB), CNRS/Université d'Aix-Marseille, Campus Luminy, Marseille, France
| | - Davide Comoletti
- Department of Pharmacology, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California at San Diego, La Jolla, CA, USA
| | - Géraldine Ferracci
- Centre d'Analyse Protéomique de Marseille (CAPM), Institut Fédératif de Recherche Jean Roche, Faculté de Médecine—Secteur Nord, Marseille, France
| | - Sandrine Conrod
- Centre de Recherche en Neurobiologie-Neurophysiologie de Marseille (CRN2M), CNRS/Université d'Aix-Marseille, Institut Fédératif de Recherche Jean Roche, Faculté de Médecine—Secteur Nord, Marseille, France
| | - Simon U Garcia
- Department of Pharmacology, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California at San Diego, La Jolla, CA, USA
| | - Palmer Taylor
- Department of Pharmacology, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California at San Diego, La Jolla, CA, USA
| | - Yves Bourne
- Architecture et Fonction des Macromolécules Biologiques (AFMB), CNRS/Université d'Aix-Marseille, Campus Luminy, Marseille, France
| | - Pascale Marchot
- Centre de Recherche en Neurobiologie-Neurophysiologie de Marseille (CRN2M), CNRS/Université d'Aix-Marseille, Institut Fédératif de Recherche Jean Roche, Faculté de Médecine—Secteur Nord, Marseille, France
| |
Collapse
|
32
|
Ching MSL, Shen Y, Tan WH, Jeste SS, Morrow EM, Chen X, Mukaddes NM, Yoo SY, Hanson E, Hundley R, Austin C, Becker RE, Berry GT, Driscoll K, Engle EC, Friedman S, Gusella JF, Hisama FM, Irons MB, Lafiosca T, LeClair E, Miller DT, Neessen M, Picker JD, Rappaport L, Rooney CM, Sarco DP, Stoler JM, Walsh CA, Wolff RR, Zhang T, Nasir RH, Wu BL, on behalf of the Children's Hospital Boston Genotype Phenotype Study Group. Deletions of NRXN1 (neurexin-1) predispose to a wide spectrum of developmental disorders. Am J Med Genet B Neuropsychiatr Genet 2010; 153B:937-47. [PMID: 20468056 PMCID: PMC3001124 DOI: 10.1002/ajmg.b.31063] [Citation(s) in RCA: 160] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Collaborators] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2009] [Accepted: 12/15/2009] [Indexed: 01/22/2023]
Abstract
Research has implicated mutations in the gene for neurexin-1 (NRXN1) in a variety of conditions including autism, schizophrenia, and nicotine dependence. To our knowledge, there have been no published reports describing the breadth of the phenotype associated with mutations in NRXN1. We present a medical record review of subjects with deletions involving exonic sequences of NRXN1. We ascertained cases from 3,540 individuals referred clinically for comparative genomic hybridization testing from March 2007 to January 2009. Twelve subjects were identified with exonic deletions. The phenotype of individuals with NRXN1 deletion is variable and includes autism spectrum disorders, mental retardation, language delays, and hypotonia. There was a statistically significant increase in NRXN1 deletion in our clinical sample compared to control populations described in the literature (P = 8.9 x 10(-7)). Three additional subjects with NRXN1 deletions and autism were identified through the Homozygosity Mapping Collaborative for Autism, and this deletion segregated with the phenotype. Our study indicates that deletions of NRXN1 predispose to a wide spectrum of developmental disorders.
Collapse
Affiliation(s)
- Michael SL Ching
- Division of Developmental Medicine, Children's Hospital BostonBoston, Massachusetts
- Harvard Medical SchoolBoston, Massachusetts
| | - Yiping Shen
- Harvard Medical SchoolBoston, Massachusetts
- Center for Human Genetic Research, Massachusetts General HospitalBoston, Massachusetts
- Department of Laboratory Medicine, Children's Hospital BostonBoston, Massachusetts
| | - Wen-Hann Tan
- Harvard Medical SchoolBoston, Massachusetts
- Division of Genetics, Children's Hospital BostonBoston, Massachusetts
| | - Shafali S Jeste
- Harvard Medical SchoolBoston, Massachusetts
- Department of Neurology, Children's Hospital BostonBoston, Massachusetts
| | - Eric M Morrow
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown UniversityProvidence, Rhode Island
| | - Xiaoli Chen
- Department of Laboratory Medicine, Children's Hospital BostonBoston, Massachusetts
- Department of Molecular Immunology, Capital Institute of PediatricsBeijing, China
| | - Nahit M Mukaddes
- Istanbul Faculty of Medicine, Department of Child Psychiatry, Istanbul UniversityIstanbul, Turkey
| | - Seung-Yun Yoo
- Division of Genetics, Children's Hospital BostonBoston, Massachusetts
| | - Ellen Hanson
- Division of Developmental Medicine, Children's Hospital BostonBoston, Massachusetts
- Harvard Medical SchoolBoston, Massachusetts
| | - Rachel Hundley
- Division of Developmental Medicine, Children's Hospital BostonBoston, Massachusetts
- Harvard Medical SchoolBoston, Massachusetts
| | - Christina Austin
- Division of Genetics, Children's Hospital BostonBoston, Massachusetts
| | - Ronald E Becker
- Division of Developmental Medicine, Children's Hospital BostonBoston, Massachusetts
- Harvard Medical SchoolBoston, Massachusetts
| | - Gerard T Berry
- Harvard Medical SchoolBoston, Massachusetts
- Division of Genetics, Children's Hospital BostonBoston, Massachusetts
| | - Katherine Driscoll
- Division of Developmental Medicine, Children's Hospital BostonBoston, Massachusetts
- Harvard Medical SchoolBoston, Massachusetts
| | - Elizabeth C Engle
- Harvard Medical SchoolBoston, Massachusetts
- Department of Neurology, Children's Hospital BostonBoston, Massachusetts
- Children's Hospital Boston, Howard Hughes Medical InstituteBoston, Massachusetts
- Manton Center for Orphan Disease Research, Children's Hospital BostonBoston, Massachusetts
- Department of Ophthalmology, Children's Hospital BostonBoston, Massachusetts
| | - Sandra Friedman
- Division of Developmental Medicine, Children's Hospital BostonBoston, Massachusetts
- Harvard Medical SchoolBoston, Massachusetts
| | - James F Gusella
- Harvard Medical SchoolBoston, Massachusetts
- Center for Human Genetic Research, Massachusetts General HospitalBoston, Massachusetts
- Department of Genetics, Harvard Medical SchoolBoston, Massachusetts
| | - Fuki M Hisama
- Harvard Medical SchoolBoston, Massachusetts
- Division of Genetics, Children's Hospital BostonBoston, Massachusetts
| | - Mira B Irons
- Harvard Medical SchoolBoston, Massachusetts
- Division of Genetics, Children's Hospital BostonBoston, Massachusetts
| | - Tina Lafiosca
- Division of Developmental Medicine, Children's Hospital BostonBoston, Massachusetts
- Harvard Medical SchoolBoston, Massachusetts
| | - Elaine LeClair
- Division of Developmental Medicine, Children's Hospital BostonBoston, Massachusetts
- Harvard Medical SchoolBoston, Massachusetts
| | - David T Miller
- Harvard Medical SchoolBoston, Massachusetts
- Division of Genetics, Children's Hospital BostonBoston, Massachusetts
- Department of Laboratory Medicine, Children's Hospital BostonBoston, Massachusetts
| | - Michael Neessen
- Division of Developmental Medicine, Children's Hospital BostonBoston, Massachusetts
- Harvard Medical SchoolBoston, Massachusetts
| | - Jonathan D Picker
- Harvard Medical SchoolBoston, Massachusetts
- Division of Genetics, Children's Hospital BostonBoston, Massachusetts
| | - Leonard Rappaport
- Division of Developmental Medicine, Children's Hospital BostonBoston, Massachusetts
- Harvard Medical SchoolBoston, Massachusetts
| | - Cynthia M Rooney
- Harvard Medical SchoolBoston, Massachusetts
- Department of Neurology, Children's Hospital BostonBoston, Massachusetts
| | - Dean P Sarco
- Harvard Medical SchoolBoston, Massachusetts
- Department of Neurology, Children's Hospital BostonBoston, Massachusetts
| | - Joan M Stoler
- Harvard Medical SchoolBoston, Massachusetts
- Division of Genetics, Children's Hospital BostonBoston, Massachusetts
| | - Christopher A Walsh
- Harvard Medical SchoolBoston, Massachusetts
- Division of Genetics, Children's Hospital BostonBoston, Massachusetts
- Manton Center for Orphan Disease Research, Children's Hospital BostonBoston, Massachusetts
- Howard Hughes Medical Institute, Beth Israel Deaconess Medical CenterBoston, Massachusetts
| | - Robert R Wolff
- Harvard Medical SchoolBoston, Massachusetts
- Department of Neurology, Children's Hospital BostonBoston, Massachusetts
| | - Ting Zhang
- Department of Molecular Immunology, Capital Institute of PediatricsBeijing, China
| | - Ramzi H Nasir
- Division of Developmental Medicine, Children's Hospital BostonBoston, Massachusetts
- Harvard Medical SchoolBoston, Massachusetts
| | - Bai-Lin Wu
- Harvard Medical SchoolBoston, Massachusetts
- Department of Laboratory Medicine, Children's Hospital BostonBoston, Massachusetts
- Children's Hospital and Institutes of Biomedical Science, Fudan UniversityShanghai, China
| | - on behalf of the Children's Hospital Boston Genotype Phenotype Study Group
- Division of Developmental Medicine, Children's Hospital BostonBoston, Massachusetts
- Harvard Medical SchoolBoston, Massachusetts
- Center for Human Genetic Research, Massachusetts General HospitalBoston, Massachusetts
- Division of Genetics, Children's Hospital BostonBoston, Massachusetts
- Department of Neurology, Children's Hospital BostonBoston, Massachusetts
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown UniversityProvidence, Rhode Island
- Department of Laboratory Medicine, Children's Hospital BostonBoston, Massachusetts
- Department of Molecular Immunology, Capital Institute of PediatricsBeijing, China
- Istanbul Faculty of Medicine, Department of Child Psychiatry, Istanbul UniversityIstanbul, Turkey
- Children's Hospital Boston, Howard Hughes Medical InstituteBoston, Massachusetts
- Manton Center for Orphan Disease Research, Children's Hospital BostonBoston, Massachusetts
- Department of Ophthalmology, Children's Hospital BostonBoston, Massachusetts
- Department of Genetics, Harvard Medical SchoolBoston, Massachusetts
- Howard Hughes Medical Institute, Beth Israel Deaconess Medical CenterBoston, Massachusetts
- Children's Hospital and Institutes of Biomedical Science, Fudan UniversityShanghai, China
| |
Collapse
Collaborators
Omar S Khwaja, Annapurna Poduri, Mustafa Sahin, Magdi Sobeih,
Collapse
|
33
|
Jacques DA, Trewhella J. Small-angle scattering for structural biology--expanding the frontier while avoiding the pitfalls. Protein Sci 2010; 19:642-57. [PMID: 20120026 PMCID: PMC2867006 DOI: 10.1002/pro.351] [Citation(s) in RCA: 315] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2009] [Revised: 01/17/2010] [Accepted: 01/19/2010] [Indexed: 12/18/2022]
Abstract
The last decade has seen a dramatic increase in the use of small-angle scattering for the study of biological macromolecules in solution. The drive for more complete structural characterization of proteins and their interactions, coupled with the increasing availability of instrumentation and easy-to-use software for data analysis and interpretation, is expanding the utility of the technique beyond the domain of the biophysicist and into the realm of the protein scientist. However, the absence of publication standards and the ease with which 3D models can be calculated against the inherently 1D scattering data means that an understanding of sample quality, data quality, and modeling assumptions is essential to have confidence in the results. This review is intended to provide a road map through the small-angle scattering experiment, while also providing a set of guidelines for the critical evaluation of scattering data. Examples of current best practice are given that also demonstrate the power of the technique to advance our understanding of protein structure and function.
Collapse
Affiliation(s)
| | - Jill Trewhella
- School of Molecular and Microbial Biosciences, The University of SydneySydney, New South Wales 2006, Australia
| |
Collapse
|
34
|
Venselaar H, Joosten RP, Vroling B, Baakman CAB, Hekkelman ML, Krieger E, Vriend G. Homology modelling and spectroscopy, a never-ending love story. EUROPEAN BIOPHYSICS JOURNAL : EBJ 2010; 39:551-63. [PMID: 19718498 PMCID: PMC2841279 DOI: 10.1007/s00249-009-0531-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2009] [Revised: 07/29/2009] [Accepted: 08/04/2009] [Indexed: 01/29/2023]
Abstract
Homology modelling is normally the technique of choice when experimental structure data are not available but three-dimensional coordinates are needed, for example, to aid with detailed interpretation of results of spectroscopic studies. Herein, the state of the art of homology modelling will be described in the light of a series of recent developments, and an overview will be given of the problems and opportunities encountered in this field. The major topic, the accuracy and precision of homology models, will be discussed extensively due to its influence on the reliability of conclusions drawn from the combination of homology models and spectroscopic data. Three real-world examples will illustrate how both homology modelling and spectroscopy can be beneficial for (bio)medical research.
Collapse
Affiliation(s)
- Hanka Venselaar
- Centre for Molecular and Biomolecular Informatics, CMBI, NCMLS 260, Radboud University Medical Centre, 6500 HB Nijmegen, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
35
|
Pampanos A, Volaki K, Kanavakis E, Papandreou O, Youroukos S, Thomaidis L, Karkelis S, Tzetis M, Kitsiou-Tzeli S. A substitution involving the NLGN4 gene associated with autistic behavior in the Greek population. Genet Test Mol Biomarkers 2010; 13:611-5. [PMID: 19645625 DOI: 10.1089/gtmb.2009.0005] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Autism is a neurodevelopmental disorder characterized by clinical, etiologic, and genetic heterogeneity. During the last decade, predisposing genes and genetic loci were under investigation. Recently, mutations in two X-linked neuroligin genes, neuroligin 3 (NLGN3) and neuroligin 4 (NLGN4), have been implicated in the pathogenesis of autism. In our ongoing survey, we screened 169 patients with autism for mutations linked with autism. In the preliminary study of specific exons of NLGN3 and NLGN4 genes, we identified the p.K378R substitution (c.1597 A > G) in exon 5 of the NLGN4 gene in a patient who was found to have mild autism and normal IQ at 3 years of age. The same mutation has previously been found in a patient with autism. It is important that, for the first time, a specific mutation in neuroligins is confirmed in a molecular screen in another homogeneous ethnic population. This finding further contributes to consideration of neuroligins as probable candidate genes for future molecular genetic studies, suggesting that a defect of synaptogenesis may predispose to autism.
Collapse
Affiliation(s)
- Andreas Pampanos
- Department of Medical Genetics, Medical School, University of Athens, Aghia Sophia Children's Hospital, Athens, Greece
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Kolozsi E, Mackenzie R, Roullet F, Decatanzaro D, Foster J. Prenatal exposure to valproic acid leads to reduced expression of synaptic adhesion molecule neuroligin 3 in mice. Neuroscience 2009; 163:1201-10. [DOI: 10.1016/j.neuroscience.2009.07.021] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2009] [Revised: 07/04/2009] [Accepted: 07/09/2009] [Indexed: 01/25/2023]
|
37
|
Synapse formation regulated by protein tyrosine phosphatase receptor T through interaction with cell adhesion molecules and Fyn. EMBO J 2009; 28:3564-78. [PMID: 19816407 DOI: 10.1038/emboj.2009.289] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2009] [Accepted: 09/07/2009] [Indexed: 01/06/2023] Open
Abstract
The receptor-type protein tyrosine phosphatases (RPTPs) have been linked to signal transduction, cell adhesion, and neurite extension. PTPRT/RPTPrho is exclusively expressed in the central nervous system and regulates synapse formation by interacting with cell adhesion molecules and Fyn protein tyrosine kinase. Overexpression of PTPRT in cultured neurons increased the number of excitatory and inhibitory synapses by recruiting neuroligins that interact with PTPRT through their ecto-domains. In contrast, knockdown of PTPRT inhibited synapse formation and withered dendrites. Incubation of cultured neurons with recombinant proteins containing the extracellular region of PTPRT reduced the number of synapses by inhibiting the interaction between ecto-domains. Synapse formation by PTPRT was inhibited by phosphorylation of tyrosine 912 within the membrane-proximal catalytic domain of PTPRT by Fyn. This tyrosine phosphorylation reduced phosphatase activity of PTPRT and reinforced homophilic interactions of PTPRT, thereby preventing the heterophilic interaction between PTPRT and neuroligins. These results suggest that brain-specific PTPRT regulates synapse formation through interaction with cell adhesion molecules, and this function and the phosphatase activity are attenuated through tyrosine phosphorylation by the synaptic tyrosine kinase Fyn.
Collapse
|
38
|
Lakey JH. Neutrons for biologists: a beginner's guide, or why you should consider using neutrons. J R Soc Interface 2009; 6 Suppl 5:S567-73. [PMID: 19656821 DOI: 10.1098/rsif.2009.0156.focus] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
From the structures of isolated protein complexes to the molecular dynamics of whole cells, neutron methods can achieve a resolution in complex systems that is inaccessible to other techniques. Biology is fortunate in that it is rich in water and hydrogen, and this allows us to exploit the differential sensitivity of neutrons to this element and its major isotope, deuterium. Furthermore, neutrons exhibit wave properties that allow us to use them in similar ways to light, X-rays and electrons. This review aims to explain the basics of biological neutron science to encourage its greater use in solving difficult problems in the life sciences.
Collapse
Affiliation(s)
- Jeremy H Lakey
- Institute for Cell and Molecular Biosciences, University of Newcastle, Framlington Place, Newcastle-upon-Tyne NE2 4HH, UK.
| |
Collapse
|
39
|
Suckow AT, Comoletti D, Waldrop MA, Mosedale M, Egodage S, Taylor P, Chessler SD. Expression of neurexin, neuroligin, and their cytoplasmic binding partners in the pancreatic beta-cells and the involvement of neuroligin in insulin secretion. Endocrinology 2008; 149:6006-17. [PMID: 18755801 PMCID: PMC2613060 DOI: 10.1210/en.2008-0274] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The composition of the beta-cell exocytic machinery is very similar to that of neuronal synapses, and the developmental pathway of beta-cells and neurons substantially overlap. beta-Cells secrete gamma-aminobutyric acid and express proteins that, in the brain, are specific markers of inhibitory synapses. Recently, neuronal coculture experiments have identified three families of synaptic cell-surface molecules (neurexins, neuroligins, and SynCAM) that drive synapse formation in vitro and that control the differentiation of nascent synapses into either excitatory or inhibitory fully mature nerve terminals. The inhibitory synapse-like character of the beta-cells led us to hypothesize that members of these families of synapse-inducing adhesion molecules would be expressed in beta-cells and that the pattern of expression would resemble that associated with neuronal inhibitory synaptogenesis. Here, we describe beta-cell expression of the neuroligins, neurexins, and SynCAM, and show that neuroligin expression affects insulin secretion in INS-1 beta-cells and rat islet cells. Our findings demonstrate that neuroligins and neurexins are expressed outside the central nervous system and help confer an inhibitory synaptic-like phenotype onto the beta-cell surface. Analogous to their role in synaptic neurotransmission, neurexin-neuroligin interactions may play a role in the formation of the submembrane insulin secretory apparatus.
Collapse
Affiliation(s)
- Arthur T Suckow
- Department of Medicine, Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, California 92093, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
Biswas S, Russell RJ, Jackson CJ, Vidovic M, Ganeshina O, Oakeshott JG, Claudianos C. Bridging the synaptic gap: neuroligins and neurexin I in Apis mellifera. PLoS One 2008; 3:e3542. [PMID: 18974885 PMCID: PMC2570956 DOI: 10.1371/journal.pone.0003542] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2008] [Accepted: 09/16/2008] [Indexed: 01/07/2023] Open
Abstract
Vertebrate studies show neuroligins and neurexins are binding partners in a trans-synaptic cell adhesion complex, implicated in human autism and mental retardation disorders. Here we report a genetic analysis of homologous proteins in the honey bee. As in humans, the honeybee has five large (31–246 kb, up to 12 exons each) neuroligin genes, three of which are tightly clustered. RNA analysis of the neuroligin-3 gene reveals five alternatively spliced transcripts, generated through alternative use of exons encoding the cholinesterase-like domain. Whereas vertebrates have three neurexins the bee has just one gene named neurexin I (400 kb, 28 exons). However alternative isoforms of bee neurexin I are generated by differential use of 12 splice sites, mostly located in regions encoding LNS subdomains. Some of the splice variants of bee neurexin I resemble the vertebrate α- and β-neurexins, albeit in vertebrates these forms are generated by alternative promoters. Novel splicing variations in the 3′ region generate transcripts encoding alternative trans-membrane and PDZ domains. Another 3′ splicing variation predicts soluble neurexin I isoforms. Neurexin I and neuroligin expression was found in brain tissue, with expression present throughout development, and in most cases significantly up-regulated in adults. Transcripts of neurexin I and one neuroligin tested were abundant in mushroom bodies, a higher order processing centre in the bee brain. We show neuroligins and neurexins comprise a highly conserved molecular system with likely similar functional roles in insects as vertebrates, and with scope in the honeybee to generate substantial functional diversity through alternative splicing. Our study provides important prerequisite data for using the bee as a model for vertebrate synaptic development.
Collapse
Affiliation(s)
- Sunita Biswas
- University of Queensland, Queensland Brain Institute, Brisbane, Queensland, Australia
- Australian National University, Research School of Biological Sciences, Canberra, Australian Capital Territory, Australia
- CSIRO Entomology, Black Mountain, Canberra, Australian Capital Territory, Australia
| | - Robyn J. Russell
- CSIRO Entomology, Black Mountain, Canberra, Australian Capital Territory, Australia
| | - Colin J. Jackson
- CSIRO Entomology, Black Mountain, Canberra, Australian Capital Territory, Australia
| | - Maria Vidovic
- Australian National University, Research School of Biological Sciences, Canberra, Australian Capital Territory, Australia
| | - Olga Ganeshina
- University of Queensland, Queensland Brain Institute, Brisbane, Queensland, Australia
| | - John G. Oakeshott
- CSIRO Entomology, Black Mountain, Canberra, Australian Capital Territory, Australia
| | - Charles Claudianos
- University of Queensland, Queensland Brain Institute, Brisbane, Queensland, Australia
- CSIRO Entomology, Black Mountain, Canberra, Australian Capital Territory, Australia
- * E-mail:
| |
Collapse
|
41
|
Comoletti D, Grishaev A, Whitten AE, Taylor P, Trewhella J. Characterization of the solution structure of a neuroligin/beta-neurexin complex. Chem Biol Interact 2008; 175:150-5. [PMID: 18550038 PMCID: PMC2587492 DOI: 10.1016/j.cbi.2008.04.040] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2007] [Revised: 04/09/2008] [Accepted: 04/09/2008] [Indexed: 11/20/2022]
Abstract
Neuroligins are post-synaptic cell adhesion molecules that promote synaptic maturation and stabilization upon binding with pre-synaptic partners, the alpha- and beta-neurexins. Using a combination of analytical ultracentrifugation, small angle X-ray, and neutron scattering, we have characterized the low-resolution three-dimensional structure of the extracellular domain of the neuroligins, free in solution, and in complex with beta-neurexin. The globular extracellular domain of the neuroligins forms stable homodimers through a four-helix bundle typical of the cholinesterases and other members of the alpha/beta-hydrolase fold family. The presence of the stalk region adds to the extracellular domain of neuroligin-1 an elongated structure, suggesting a rod-like nature of the stalk domain. Sedimentation equilibrium coupled with solution scattering data of the beta-neurexin/neuroligin-1 complex indicated a 2:2 stoichiometry where two beta-neurexin molecules bind to a neuroligin-1 dimer. Deuteration of neurexin allowed us to collect neutron scattering data that, in combination with other biochemical techniques, provide a basis for optimizing the positioning of each component in a detailed computational model of the neuroligin/neurexin complex. As several mutations of both neurexin and neuroligin genes have been linked to autism spectrum disorders and mental retardation, these new structures provide an important framework for the study of altered structure and function of these synaptic proteins.
Collapse
Affiliation(s)
- Davide Comoletti
- Department of Pharmacology, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California-San Diego, La Jolla, CA 92093-0650, USA.
| | | | | | | | | |
Collapse
|
42
|
Mutational analysis of the neurexin/neuroligin complex reveals essential and regulatory components. Proc Natl Acad Sci U S A 2008; 105:15124-9. [PMID: 18812509 DOI: 10.1073/pnas.0801639105] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Neurexins are cell-surface molecules that bind neuroligins to form a heterophilic, Ca(2+)-dependent complex at central synapses. This transsynaptic complex is required for efficient neurotransmission and is involved in the formation of synaptic contacts. In addition, both molecules have been identified as candidate genes for autism. Here we performed mutagenesis experiments to probe for essential components of the neurexin/neuroligin binding interface at the single-amino acid level. We found that in neurexins the contact area is sharply delineated and consists of hydrophobic residues of the LNS domain that surround a Ca(2+) binding pocket. Point mutations that changed electrostatic and shape properties leave Ca(2+) coordination intact but completely inhibit neuroligin binding, whereas alternative splicing in alpha- and beta-neurexins and in neuroligins has a weaker effect on complex formation. In neuroligins, the contact area appears less distinct because exchange of a more distant aspartate completely abolished binding to neurexin but many mutations of predicted interface residues had no strong effect on binding. Together with calculations of energy terms for presumed interface hot spots that complement and extend our mutagenesis and recent crystal structure data, this study presents a comprehensive structural basis for the complex formation of neurexins and neuroligins and their transsynaptic signaling between neurons.
Collapse
|
43
|
Old and new questions about cholinesterases. Chem Biol Interact 2008; 175:30-44. [DOI: 10.1016/j.cbi.2008.04.039] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2007] [Revised: 04/25/2008] [Accepted: 04/28/2008] [Indexed: 01/21/2023]
|
44
|
Abstract
PURPOSE OF REVIEW Autism is now recognized in one out of 150 children. This review highlights the topics within the growing autism literature that are shaping current thinking on autism and advancing research and clinical understanding of autism spectrum disorders. RECENT FINDINGS The role of single-stranded microdeletions and epigenetic influences on brain development has dramatically altered our understanding of the etiology of the autisms. Recent research has focused on the role of synapse structure and function as central to the development of autism and suggests possible targets of interventions. Brain underconnectivity has been a focus in recent imaging studies and has become a central theme in conceptualizing autism. Despite increased awareness of autism there is no 'epidemic' and no one cause for autism. Data from the sibling studies are identifying early markers of autism and defining the broader autism phenotype. SUMMARY Larger datasets in genetics, a focus on the early signs of autism, and increased recognition of the importance of defining subgroups of children with autism are leading to a greater understanding of the etiologies of autism. A growing interest in defining the molecular biology of social cognition, which is at the core of autism, will lead to expansion of our presently limited choices of mechanistically based interventions.
Collapse
|
45
|
Affiliation(s)
- Jill Trewhella
- School of Molecular and Microbial Biosciences, University of Sydney, NSW 2006, Australia.
| |
Collapse
|
46
|
Levinson JN, El-Husseini A. A crystal-clear interaction: relating neuroligin/neurexin complex structure to function at the synapse. Neuron 2008; 56:937-9. [PMID: 18093514 DOI: 10.1016/j.neuron.2007.12.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Neuronal circuits are maintained by homeostatic mechanisms controlling synapse maturation and signaling. Neuroligins (NLs) and neurexins (Nrxs) may regulate the fine balance between excitation and inhibition. In this issue of Neuron, Araç et al. and Fabrichny et al. define crystal structures of NLs bound to beta-Nrx, providing insights into their synaptic actions and clarifying structural defects associated with autism-linked mutations.
Collapse
Affiliation(s)
- Joshua N Levinson
- Department of Psychiatry, University of British Columbia, Brain Research Center, Vancouver, BC, Canada
| | | |
Collapse
|
47
|
Araç D, Boucard AA, Ozkan E, Strop P, Newell E, Südhof TC, Brunger AT. Structures of neuroligin-1 and the neuroligin-1/neurexin-1 beta complex reveal specific protein-protein and protein-Ca2+ interactions. Neuron 2008; 56:992-1003. [PMID: 18093522 DOI: 10.1016/j.neuron.2007.12.002] [Citation(s) in RCA: 167] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2007] [Revised: 11/30/2007] [Accepted: 12/03/2007] [Indexed: 01/11/2023]
Abstract
Neurexins and neuroligins provide trans-synaptic connectivity by the Ca2+-dependent interaction of their alternatively spliced extracellular domains. Neuroligins specify synapses in an activity-dependent manner, presumably by binding to neurexins. Here, we present the crystal structures of neuroligin-1 in isolation and in complex with neurexin-1 beta. Neuroligin-1 forms a constitutive dimer, and two neurexin-1 beta monomers bind to two identical surfaces on the opposite faces of the neuroligin-1 dimer to form a heterotetramer. The neuroligin-1/neurexin-1 beta complex exhibits a nanomolar affinity and includes a large binding interface that contains bound Ca2+. Alternatively spliced sites in neurexin-1 beta and in neuroligin-1 are positioned nearby the binding interface, explaining how they regulate the interaction. Structure-based mutations of neuroligin-1 at the interface disrupt binding to neurexin-1 beta, but not the folding of neuroligin-1 and confirm the validity of the binding interface of the neuroligin-1/neurexin-1 beta complex. Our results provide molecular insights for understanding the role of cell-adhesion proteins in synapse function.
Collapse
Affiliation(s)
- Demet Araç
- Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Koehnke J, Jin X, Trbovic N, Katsamba PS, Brasch J, Ahlsen G, Scheiffele P, Honig B, Palmer A, Shapiro L. Crystal structures of beta-neurexin 1 and beta-neurexin 2 ectodomains and dynamics of splice insertion sequence 4. Structure 2008; 16:410-21. [PMID: 18334216 PMCID: PMC2750865 DOI: 10.1016/j.str.2007.12.024] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2007] [Revised: 12/18/2007] [Accepted: 12/18/2007] [Indexed: 11/28/2022]
Abstract
Presynaptic neurexins (NRXs) bind to postsynaptic neuroligins (NLs) to form Ca(2+)-dependent complexes that bridge neural synapses. beta-NRXs bind NLs through their LNS domains, which contain a single site of alternative splicing (splice site 4) giving rise to two isoforms: +4 and Delta. We present crystal structures of the Delta isoforms of the LNS domains from beta-NRX1 and beta-NRX2, crystallized in the presence of Ca(2+) ions. The Ca(2+)-binding site is disordered in the beta-NRX2 structure, but the 1.7 A beta-NRX1 structure reveals a single Ca(2+) ion, approximately 12 A from the splice insertion site, with one coordinating ligand donated by a glutamic acid from an adjacent beta-NRX1 molecule. NMR studies of beta-NRX1+4 show that the insertion sequence is unstructured, and remains at least partially disordered in complex with NL. These results raise the possibility that beta-NRX insertion sequence 4 may function in roles independent of neuroligin binding.
Collapse
Affiliation(s)
- Jesko Koehnke
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032 USA
| | - Xiangshu Jin
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032 USA
| | - Nikola Trbovic
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032 USA
| | - Phinikoula S. Katsamba
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032 USA
- Howard Hughes Medical Institute
| | - Julia Brasch
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032 USA
| | - Goran Ahlsen
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032 USA
| | - Peter Scheiffele
- Department of Physiology and Cellular Biophysics and Department of Neuroscience, Columbia University, New York, NY 10032 USA
| | - Barry Honig
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032 USA
- Howard Hughes Medical Institute
| | - Arthur Palmer
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032 USA
| | - Lawrence Shapiro
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032 USA
- Edward S. Harkness Eye Institute, Columbia University, New York, NY 10032 USA
| |
Collapse
|
49
|
Minisymposium 2: Structural Refinement & Modeling Guided by Low-Resolution Experimental Poster. Biophys J 2008. [DOI: 10.1016/s0006-3495(08)79045-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
50
|
Budreck EC, Scheiffele P. Neuroligin-3 is a neuronal adhesion protein at GABAergic and glutamatergic synapses. Eur J Neurosci 2008; 26:1738-48. [PMID: 17897391 DOI: 10.1111/j.1460-9568.2007.05842.x] [Citation(s) in RCA: 237] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Synaptic adhesion molecules are thought to play a critical role in the formation, function and plasticity of neuronal networks. Neuroligins (NL1-4) are a family of presumptive postsynaptic cell adhesion molecules. NL1 and NL2 isoforms are concentrated at glutamatergic and GABAergic synapses, respectively, but the cellular expression and synaptic localization of the endogenous NL3 and NL4 isoforms are unknown. We generated a panel of NL isoform-specific antibodies and examined the expression, developmental regulation and synaptic specificity of NL3. We found that NL3 was enriched in brain, where NL3 protein levels increased during postnatal development, coinciding with the peak of synaptogenesis. Subcellular fractionation revealed a concentration of NL3 in synaptic plasma membranes and postsynaptic densities. In cultured hippocampal neurons, endogenous NL3 was highly expressed and was localized at both glutamatergic and GABAergic synapses. Clustering of NL3 in hippocampal neurons by neurexin-expressing cells resulted in coaggregation of NL3 with glutamatergic and GABAergic scaffolding proteins. Finally, individual synapses contained colocalized NL2 and NL3 proteins, and coimmunoprecipitation studies revealed the presence of NL1-NL3 and NL2-NL3 complexes in brain extracts. These findings suggest that rodent NL3 is a synaptic adhesion molecule that is a shared component of glutamatergic and GABAergic synapses.
Collapse
Affiliation(s)
- Elaine C Budreck
- Department of Physiology & Cellular Biophysics, Columbia University, College of Physicians & Surgeons, 630 West 168th Street, P&S 11-511, New York, NY 10032, USA
| | | |
Collapse
|