1
|
Lehr AW, McDaniel KF, Roche KW. Analyses of Human Genetic Data to Identify Clinically Relevant Domains of Neuroligins. Genes (Basel) 2024; 15:1601. [PMID: 39766868 PMCID: PMC11675371 DOI: 10.3390/genes15121601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/03/2024] [Accepted: 12/11/2024] [Indexed: 01/30/2025] Open
Abstract
Background/Objectives: Neuroligins (NLGNs) are postsynaptic adhesion molecules critical for neuronal development that are highly associated with autism spectrum disorder (ASD). Here, we provide an overview of the literature on NLGN rare variants. In addition, we introduce a new approach to analyze human variation within NLGN genes to identify sensitive regions that have an increased frequency of ASD-associated variants to better understand NLGN function. Methods: To identify critical protein subdomains within the NLGN gene family, we developed an algorithm that assesses tolerance to missense mutations in human genetic variation by comparing clinical variants from ClinVar to reference variants from gnomAD. This approach provides tolerance values to subdomains within the protein. Results: Our algorithm identified several critical regions that were conserved across multiple NLGN isoforms. Importantly, this approach also identified a previously reported cluster of pathogenic variants in NLGN4X (also conserved in NLGN1 and NLGN3) as well as a region around the highly characterized NLGN3 R451C ASD-associated mutation. Additionally, we highlighted other, as of yet, uncharacterized regions enriched with mutations. Conclusions: The systematic analysis of NLGN ASD-associated variants compared to variants identified in the unaffected population (gnomAD) reveals conserved domains in NLGN isoforms that are tolerant to variation or are enriched in clinically relevant variants. Examination of databases also allows for predictions of the presumed tolerance to loss of an allele. The use of the algorithm we developed effectively allowed the evaluation of subdomains of NLGNs and can be used to examine other ASD-associated genes.
Collapse
Affiliation(s)
- Alexander W. Lehr
- Receptor Biology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA; (A.W.L.); (K.F.M.)
- Department of Neuroscience, Brown University, Providence, RI 02906, USA
| | - Kathryn F. McDaniel
- Receptor Biology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA; (A.W.L.); (K.F.M.)
- Department of Neuroscience, Brown University, Providence, RI 02906, USA
| | - Katherine W. Roche
- Receptor Biology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA; (A.W.L.); (K.F.M.)
| |
Collapse
|
2
|
Denessiouk K, Denesyuk AI, Johnson MS, Uversky VN. Two groups and three classes of the conserved structural organization of nucleophile and non-canonical ElbowFlankOxy networks in different superfamily proteins. J Biomol Struct Dyn 2024:1-16. [PMID: 39546335 DOI: 10.1080/07391102.2024.2429798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 04/15/2024] [Indexed: 11/17/2024]
Abstract
The nucleophile elbow is a well-known structural motif, which exists in proteins with catalytic triads and contains a catalytic nucleophile and the first node of an oxyanion hole. Here, we show that structural similarities of proteins with the nucleophile elbow extend beyond simple nucleophile elbow motifs. The motifs are incorporated into larger conserved structural organizations, the ElbowFlankOxy networks, incorporating motifs and flanking residues and networks of conserved interactions. A detailed structural analysis shows two major types of ElbowFlankOxy networks, depending on the formation of the oxyanion hole. Additionally, the ElbowFlankOxy networks show three classes: Class 1-2-3, 3-1-2, and 2-3-1, defined by the order in which the catalytic nucleophile and key interacting residues are located in the amino acid sequence, giving rise to six ElbowFlankOxy network variations. This makes it possible to properly position homologous non-catalytic, non-standard, and unusual catalytic triad active sites of proteins with the nucleophile elbow within the fold classification.
Collapse
Affiliation(s)
- Konstantin Denessiouk
- Structural Bioinformatics Laboratory, Biochemistry, InFLAMES Research Flagship Center, Faculty of Science and Engineering, Biochemistry, Åbo Akademi University, Turku, Finland
| | - Alexander I Denesyuk
- Structural Bioinformatics Laboratory, Biochemistry, InFLAMES Research Flagship Center, Faculty of Science and Engineering, Biochemistry, Åbo Akademi University, Turku, Finland
| | - Mark S Johnson
- Structural Bioinformatics Laboratory, Biochemistry, InFLAMES Research Flagship Center, Faculty of Science and Engineering, Biochemistry, Åbo Akademi University, Turku, Finland
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| |
Collapse
|
3
|
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
|
4
|
Szíber Z, Drouet A, Mondin M, Levet F, Thoumine O. Neuroligin-1 dependent phosphotyrosine signaling in excitatory synapse differentiation. Front Mol Neurosci 2024; 17:1359067. [PMID: 38813439 PMCID: PMC11133670 DOI: 10.3389/fnmol.2024.1359067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 04/22/2024] [Indexed: 05/31/2024] Open
Abstract
Introduction The synaptic adhesion molecule neuroligin-1 (NLGN1) is involved in the differentiation of excitatory synapses, but the precise underlying molecular mechanisms are still debated. Here, we explored the role of NLGN1 tyrosine phosphorylation in this process, focusing on a subset of receptor tyrosine kinases (RTKs), namely FGFR1 and Trks, that were previously described to phosphorylate NLGN1 at a unique intracellular residue (Y782). Methods We used pharmacological inhibitors and genetic manipulation of those RTKs in dissociated hippocampal neurons, followed by biochemical measurement of NLGN1 phosphorylation and immunocytochemical staining of excitatory synaptic scaffolds. Results This study shows that: (i) the accumulation of PSD-95 at de novo NLGN1 clusters induced by neurexin crosslinking is reduced by FGFR and Trk inhibitors; (ii) the increase in PSD-95 puncta caused by NLGN1 over-expression is impaired by FGFR and Trk inhibitors; (iii) TrkB activation by BDNF increases NLGN1 phosphorylation; and (iv) TrkB knock-down impairs the increase of PSD-95 puncta caused by NLGN1 over-expression, an effect which is not seen with the NLGN1 Y782A mutant. Discussion Together, our data identify TrkB as one of the major RTKs responsible for NLGN1 tyrosine phosphorylation, and reveal that TrkB activity is necessary for the synaptogenic effects of NLGN1.
Collapse
Affiliation(s)
- Zsófia Szíber
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France
| | - Adèle Drouet
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France
| | - Magali Mondin
- University of Bordeaux, CNRS, INSERM, Bordeaux Imaging Center, BIC, UAR 3420, US 4, Bordeaux, France
| | - Florian Levet
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France
- University of Bordeaux, CNRS, INSERM, Bordeaux Imaging Center, BIC, UAR 3420, US 4, Bordeaux, France
| | - Olivier Thoumine
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France
| |
Collapse
|
5
|
Benner O, Cast TP, Minamide LS, Lenninger Z, Bamburg JR, Chanda S. Multiple N-linked glycosylation sites critically modulate the synaptic abundance of neuroligin isoforms. J Biol Chem 2023; 299:105361. [PMID: 37865312 PMCID: PMC10679506 DOI: 10.1016/j.jbc.2023.105361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/05/2023] [Accepted: 10/11/2023] [Indexed: 10/23/2023] Open
Abstract
In recent years, elegant glycomic and glycoproteomic approaches have revealed an intricate glycosylation profile of mammalian brain with enormous spatial and temporal diversities. Nevertheless, at a cellular level, it is unclear how these post-translational modifications affect various proteins to influence crucial neuronal properties. Here, we have investigated the impact of N-linked glycosylation on neuroligins (NLGNs), a class of cell-adhesion molecules that play instructive roles in synapse organization. We found that endogenous NLGN proteins are differentially glycosylated across several regions of murine brain in a sex-independent but isoform-dependent manner. In both rodent primary neurons derived from brain sections and human neurons differentiated from stem cells, all NLGN variants were highly enriched with multiple N-glycan subtypes, which cumulatively ensured their efficient trafficking to the cell surface. Removal of these N-glycosylation residues only had a moderate effect on NLGNs' stability or expression levels but particularly enhanced their retention at the endoplasmic reticulum. As a result, the glycosylation-deficient NLGNs exhibited considerable impairments in their dendritic distribution and postsynaptic accumulation, which in turn, virtually eliminated their ability to recruit presynaptic terminals and significantly reduced NLGN overexpression-induced assemblies of both glutamatergic and GABAergic synapse structures. Therefore, our results highlight an essential mechanistic contribution of N-linked glycosylations in facilitating the appropriate secretory transport of a major synaptic cell-adhesion molecule and promoting its cellular function in neurons.
Collapse
Affiliation(s)
- Orion Benner
- Biochemistry & Molecular Biology, Colorado State University, Fort Collins, USA
| | - Thomas P Cast
- Biochemistry & Molecular Biology, Colorado State University, Fort Collins, USA
| | - Laurie S Minamide
- Biochemistry & Molecular Biology, Colorado State University, Fort Collins, USA
| | - Zephyr Lenninger
- Molecular, Cellular & Integrated Neurosciences, Colorado State University, Fort Collins, Colorado, USA
| | - James R Bamburg
- Biochemistry & Molecular Biology, Colorado State University, Fort Collins, USA; Molecular, Cellular & Integrated Neurosciences, Colorado State University, Fort Collins, Colorado, USA; Cell & Molecular Biology, Colorado State University, Fort Collins, Colorado, USA
| | - Soham Chanda
- Biochemistry & Molecular Biology, Colorado State University, Fort Collins, USA; Molecular, Cellular & Integrated Neurosciences, Colorado State University, Fort Collins, Colorado, USA; Cell & Molecular Biology, Colorado State University, Fort Collins, Colorado, USA.
| |
Collapse
|
6
|
Cortés E, Pak JS, Özkan E. Structure and evolution of neuronal wiring receptors and ligands. Dev Dyn 2023; 252:27-60. [PMID: 35727136 PMCID: PMC10084454 DOI: 10.1002/dvdy.512] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 06/13/2022] [Accepted: 06/14/2022] [Indexed: 01/04/2023] Open
Abstract
One of the fundamental properties of a neuronal circuit is the map of its connections. The cellular and developmental processes that allow for the growth of axons and dendrites, selection of synaptic targets, and formation of functional synapses use neuronal surface receptors and their interactions with other surface receptors, secreted ligands, and matrix molecules. Spatiotemporal regulation of the expression of these receptors and cues allows for specificity in the developmental pathways that wire stereotyped circuits. The families of molecules controlling axon guidance and synapse formation are generally conserved across animals, with some important exceptions, which have consequences for neuronal connectivity. Here, we summarize the distribution of such molecules across multiple taxa, with a focus on model organisms, evolutionary processes that led to the multitude of such molecules, and functional consequences for the diversification or loss of these receptors.
Collapse
Affiliation(s)
- Elena Cortés
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, Illinois, USA.,The Neuroscience Institute, University of Chicago, Chicago, Illinois, USA
| | - Joseph S Pak
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, Illinois, USA.,The Neuroscience Institute, University of Chicago, Chicago, Illinois, USA
| | - Engin Özkan
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, Illinois, USA.,The Neuroscience Institute, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
7
|
Israel LL, Galstyan A, Cox A, Shatalova ES, Sun T, Rashid MH, Grodzinski Z, Chiechi A, Fuchs DT, Patil R, Koronyo-Hamaoui M, Black KL, Ljubimova JY, Holler E. Signature Effects of Vector-Guided Systemic Nano Bioconjugate Delivery Across Blood-Brain Barrier of Normal, Alzheimer's, and Tumor Mouse Models. ACS NANO 2022; 16:11815-11832. [PMID: 35961653 PMCID: PMC9413444 DOI: 10.1021/acsnano.1c10034] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
The ability to cross the blood-brain barrier (BBB) is critical for targeted therapy of the central nerve system (CNS). Six peptide vectors were covalently attached to a 50 kDa poly(β-l-malic acid)-trileucine polymer forming P/LLL(40%)/vector conjugates. The vectors were Angiopep-2 (AP2), B6, Miniap-4 (M4), and d-configurated peptides D1, D3, and ACI-89, with specificity for transcytosis receptors low-density lipoprotein receptor-related protein-1 (LRP-1), transferrin receptor (TfR), bee venom-derived ion channel, and Aβ/LRP-1 related transcytosis complex, respectively. The BBB-permeation efficacies were substantially increased ("boosted") in vector conjugates of P/LLL(40%). We have found that the copolymer group binds at the endothelial membrane and, by an allosterically membrane rearrangement, exposes the sites for vector-receptor complex formation. The specificity of vectors is indicated by competition experiments with nonconjugated vectors. P/LLL(40%) does not function as an inhibitor, suggesting that the copolymer binding site is eliminated after binding of the vector-nanoconjugate. The two-step mechanism, binding to endothelial membrane and allosteric exposure of transcytosis receptors, is supposed to be an integral feature of nanoconjugate-transcytosis pathways. In vivo brain delivery signatures of the nanoconjugates were recapitulated in mouse brains of normal, tumor (glioblastoma), and Alzheimer's disease (AD) models. BBB permeation of the tumor was most efficient, followed by normal and then AD-like brain. In tumor-bearing and normal brains, AP2 was the top performing vector; however, in AD models, D3 and D1 peptides were superior ones. The TfR vector B6 was equally efficient in normal and AD-model brains. Cross-permeation efficacies are manifested through modulated vector coligation and dosage escalation such as supra-linear dose dependence and crossover transcytosis activities.
Collapse
Affiliation(s)
- Liron L. Israel
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Anna Galstyan
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Alysia Cox
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Ekaterina S. Shatalova
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Tao Sun
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Mohammad-Harun Rashid
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Zachary Grodzinski
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Antonella Chiechi
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Dieu-Trang Fuchs
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Rameshwar Patil
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Maya Koronyo-Hamaoui
- Department of Neurosurgery and Department of Biomedical Sciences,
Division of Applied Cell Biology and Physiology, Cedars-Sinai Medical Center, Los
Angeles, California 90048, United States
| | - Keith L. Black
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Julia Y. Ljubimova
- Terasaki Institute for Biomedical Innovation
(TIBI), 1018 Westwood
Boulevard, Los Angeles, California 90024, United States
| | - Eggehard Holler
- Terasaki Institute for Biomedical Innovation
(TIBI), 1018 Westwood
Boulevard, Los Angeles, California 90024, United States
| |
Collapse
|
8
|
Nozawa K, Sogabe T, Hayashi A, Motohashi J, Miura E, Arai I, Yuzaki M. In vivo nanoscopic landscape of neurexin ligands underlying anterograde synapse specification. Neuron 2022; 110:3168-3185.e8. [PMID: 36007521 DOI: 10.1016/j.neuron.2022.07.027] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 05/04/2022] [Accepted: 07/27/2022] [Indexed: 11/17/2022]
Abstract
Excitatory synapses are formed and matured by the cooperative actions of synaptic organizers, such as neurexins (Nrxns), neuroligins (Nlgns), LRRTMs, and Cbln1. Recent super-resolution nanoscopy developments have revealed that many synaptic organizers, as well as glutamate receptors and glutamate release machinery, exist as nanoclusters within synapses. However, it is unclear how such nanodomains interact with each other to organize excitatory synapses in vivo. By applying X10 expansion microscopy to epitope tag knockin mice, we found that Cbln1, Nlgn1, and LRRTM1, which share Nrxn as a common presynaptic receptor, form overlapping or separate nanodomains depending on Nrxn with or without a sequence encoded by splice site 4. The size and position of glutamate receptor nanodomains of GluD1, NMDA, and AMPA receptors were regulated by Cbln1, Nlgn1, and LRRTM1 nanodomains, respectively. These findings indicate that Nrxns anterogradely regulate the postsynaptic nanoscopic architecture of glutamate receptors through competition and coordination of Nrxn ligands.
Collapse
Affiliation(s)
- Kazuya Nozawa
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Taku Sogabe
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Ayumi Hayashi
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Junko Motohashi
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Eriko Miura
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Itaru Arai
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Michisuke Yuzaki
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan.
| |
Collapse
|
9
|
Pohl TT, Hörnberg H. Neuroligins in neurodevelopmental conditions: how mouse models of de novo mutations can help us link synaptic function to social behavior. Neuronal Signal 2022; 6:NS20210030. [PMID: 35601025 PMCID: PMC9093077 DOI: 10.1042/ns20210030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 04/08/2022] [Accepted: 04/20/2022] [Indexed: 11/19/2022] Open
Abstract
Neurodevelopmental conditions (or neurodevelopmental disorders, NDDs) are highly heterogeneous with overlapping characteristics and shared genetic etiology. The large symptom variability and etiological heterogeneity have made it challenging to understand the biological mechanisms underpinning NDDs. To accommodate this individual variability, one approach is to move away from diagnostic criteria and focus on distinct dimensions with relevance to multiple NDDs. This domain approach is well suited to preclinical research, where genetically modified animal models can be used to link genetic variability to neurobiological mechanisms and behavioral traits. Genetic factors associated with NDDs can be grouped functionally into common biological pathways, with one prominent functional group being genes associated with the synapse. These include the neuroligins (Nlgns), a family of postsynaptic transmembrane proteins that are key modulators of synaptic function. Here, we review how research using Nlgn mouse models has provided insight into how synaptic proteins contribute to behavioral traits associated with NDDs. We focus on how mutations in different Nlgns affect social behaviors, as differences in social interaction and communication are a common feature of most NDDs. Importantly, mice carrying distinct mutations in Nlgns share some neurobiological and behavioral phenotypes with other synaptic gene mutations. Comparing the functional implications of mutations in multiple synaptic proteins is a first step towards identifying convergent neurobiological pathways in multiple brain regions and circuits.
Collapse
Affiliation(s)
- Tobias T. Pohl
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Straße 10, Berlin 13125, Germany
| | - Hanna Hörnberg
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Straße 10, Berlin 13125, Germany
| |
Collapse
|
10
|
Toledo A, Letellier M, Bimbi G, Tessier B, Daburon S, Favereaux A, Chamma I, Vennekens K, Vanderlinden J, Sainlos M, de Wit J, Choquet D, Thoumine O. MDGAs are fast-diffusing molecules that delay excitatory synapse development by altering neuroligin behavior. eLife 2022; 11:75233. [PMID: 35532105 PMCID: PMC9084894 DOI: 10.7554/elife.75233] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 04/11/2022] [Indexed: 12/28/2022] Open
Abstract
MDGA molecules can bind neuroligins and interfere with trans-synaptic interactions to neurexins, thereby impairing synapse development. However, the subcellular localization and dynamics of MDGAs, or their specific action mode in neurons remain unclear. Here, surface immunostaining of endogenous MDGAs and single molecule tracking of recombinant MDGAs in dissociated hippocampal neurons reveal that MDGAs are homogeneously distributed and exhibit fast membrane diffusion, with a small reduction in mobility across neuronal maturation. Knocking-down/out MDGAs using shRNAs and CRISPR/Cas9 strategies increases the density of excitatory synapses, the membrane confinement of neuroligin-1, and the phosphotyrosine level of neuroligins associated with excitatory post-synaptic differentiation. Finally, MDGA silencing reduces the mobility of AMPA receptors, increases the frequency of miniature EPSCs (but not IPSCs), and selectively enhances evoked AMPA-receptor-mediated EPSCs in CA1 pyramidal neurons. Overall, our results support a mechanism by which interactions between MDGAs and neuroligin-1 delays the assembly of functional excitatory synapses containing AMPA receptors.
Collapse
Affiliation(s)
- Andrea Toledo
- University of Bordeaux, CNRS UMR 5297, Interdisciplinary Institute for Neuroscience
| | - Mathieu Letellier
- University of Bordeaux, CNRS UMR 5297, Interdisciplinary Institute for Neuroscience
| | - Giorgia Bimbi
- University of Bordeaux, CNRS UMR 5297, Interdisciplinary Institute for Neuroscience
| | - Béatrice Tessier
- University of Bordeaux, CNRS UMR 5297, Interdisciplinary Institute for Neuroscience
| | - Sophie Daburon
- University of Bordeaux, CNRS UMR 5297, Interdisciplinary Institute for Neuroscience
| | - Alexandre Favereaux
- University of Bordeaux, CNRS UMR 5297, Interdisciplinary Institute for Neuroscience
| | - Ingrid Chamma
- University of Bordeaux, CNRS UMR 5297, Interdisciplinary Institute for Neuroscience
| | - Kristel Vennekens
- VIB Center for Brain & Disease Research and KU Leuven, Department of Neurosciences, Leuven Brain Institute
| | - Jeroen Vanderlinden
- VIB Center for Brain & Disease Research and KU Leuven, Department of Neurosciences, Leuven Brain Institute
| | - Matthieu Sainlos
- University of Bordeaux, CNRS UMR 5297, Interdisciplinary Institute for Neuroscience
| | - Joris de Wit
- VIB Center for Brain & Disease Research and KU Leuven, Department of Neurosciences, Leuven Brain Institute
| | - Daniel Choquet
- University of Bordeaux, CNRS UMR 5297, Interdisciplinary Institute for Neuroscience
- University of Bordeaux, CNRS UAR 3420, INSERM, Bordeaux Imaging Center
| | - Olivier Thoumine
- University of Bordeaux, CNRS UMR 5297, Interdisciplinary Institute for Neuroscience
| |
Collapse
|
11
|
Lagardère M, Drouet A, Sainlos M, Thoumine O. High-Resolution Fluorescence Imaging Combined With Computer Simulations to Quantitate Surface Dynamics and Nanoscale Organization of Neuroligin-1 at Synapses. Front Synaptic Neurosci 2022; 14:835427. [PMID: 35546899 PMCID: PMC9083120 DOI: 10.3389/fnsyn.2022.835427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 03/09/2022] [Indexed: 11/13/2022] Open
Abstract
Neuroligins (NLGNs) form a family of cell adhesion molecules implicated in synapse development, but the mechanisms that retain these proteins at synapses are still incompletely understood. Recent studies indicate that surface-associated NLGN1 is diffusionally trapped at synapses, where it interacts with quasi-static scaffolding elements of the post-synaptic density. Whereas single molecule tracking reveals rapid diffusion and transient immobilization of NLGN1 at synapses within seconds, fluorescence recovery after photobleaching experiments indicate instead a long-term turnover of NLGN1 at synapse, in the hour time range. To gain insight into the mechanisms supporting NLGN1 anchorage at post-synapses and try to reconcile those experimental paradigms, we quantitatively analyzed here live-cell and super-resolution imaging experiments performed on NLGN1 using a newly released simulator of membrane protein dynamics for fluorescence microscopy, FluoSim. Based on a small set of parameters including diffusion coefficients, binding constants, and photophysical rates, the framework describes fairly well the dynamic behavior of extra-synaptic and synaptic NLGN1 over both short and long time ranges, and provides an estimate of NLGN1 copy numbers in post-synaptic densities at steady-state (around 50 dimers). One striking result is that the residence time of NLGN1 at synapses is much longer than what can be expected from extracellular interactions with pre-synaptic neurexins only, suggesting that NLGN1 is stabilized at synapses through multivalent interactions with intracellular post-synaptic scaffolding proteins.
Collapse
Affiliation(s)
| | | | | | - Olivier Thoumine
- CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, University of Bordeaux, Bordeaux, France
| |
Collapse
|
12
|
Zhang Z, Hou M, Ou H, Wang D, Li Z, Zhang H, Lu J. Expression and structural analysis of human neuroligin 2 and neuroligin 3 implicated in autism spectrum disorders. Front Endocrinol (Lausanne) 2022; 13:1067529. [PMID: 36479216 PMCID: PMC9719943 DOI: 10.3389/fendo.2022.1067529] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 10/31/2022] [Indexed: 11/22/2022] Open
Abstract
The development of autism spectrum disorders (ASDs) involves both environmental factors such as maternal diabetes and genetic factors such as neuroligins (NLGNs). NLGN2 and NLGN3 are two members of NLGNs with distinct distributions and functions in synapse development and plasticity. The relationship between maternal diabetes and NLGNs, and the distinct working mechanisms of different NLGNs currently remain unclear. Here, we first analyzed the expression levels of NLGN2 and NLGN3 in a streptozotocin-induced ASD mouse model and different brain regions to reveal their differences and similarities. Then, cryogenic electron microscopy (cryo-EM) structures of human NLGN2 and NLGN3 were determined. The overall structures are similar to their homologs in previous reports. However, structural comparisons revealed the relative rotations of two protomers in the homodimers of NLGN2 and NLGN3. Taken together with the previously reported NLGN2-MDGA1 complex, we speculate that the distinct assembly adopted by NLGN2 and NLGN3 may affect their interactions with MDGAs. Our results provide structural insights into the potential distinct mechanisms of NLGN2 and NLGN3 implicated in the development of ASD.
Collapse
Affiliation(s)
- Zhenzhen Zhang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Mengzhuo Hou
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Huaxing Ou
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Daping Wang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
- Department of Orthopedics, Shenzhen Intelligent Orthopaedics and Biomedical Innovation Platform, Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Zhifang Li
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
- *Correspondence: Jianping Lu, ; Huawei Zhang, ; Zhifang Li,
| | - Huawei Zhang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, China
- *Correspondence: Jianping Lu, ; Huawei Zhang, ; Zhifang Li,
| | - Jianping Lu
- Department of Child and Adolescent Psychiatry, Shenzhen Kangning Hospital, Shenzhen Mental Health Center, Shenzhen, China
- *Correspondence: Jianping Lu, ; Huawei Zhang, ; Zhifang Li,
| |
Collapse
|
13
|
Uchigashima M, Cheung A, Futai K. Neuroligin-3: A Circuit-Specific Synapse Organizer That Shapes Normal Function and Autism Spectrum Disorder-Associated Dysfunction. Front Mol Neurosci 2021; 14:749164. [PMID: 34690695 PMCID: PMC8526735 DOI: 10.3389/fnmol.2021.749164] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/06/2021] [Indexed: 01/02/2023] Open
Abstract
Chemical synapses provide a vital foundation for neuron-neuron communication and overall brain function. By tethering closely apposed molecular machinery for presynaptic neurotransmitter release and postsynaptic signal transduction, circuit- and context- specific synaptic properties can drive neuronal computations for animal behavior. Trans-synaptic signaling via synaptic cell adhesion molecules (CAMs) serves as a promising mechanism to generate the molecular diversity of chemical synapses. Neuroligins (Nlgns) were discovered as postsynaptic CAMs that can bind to presynaptic CAMs like Neurexins (Nrxns) at the synaptic cleft. Among the four (Nlgn1-4) or five (Nlgn1-3, Nlgn4X, and Nlgn4Y) isoforms in rodents or humans, respectively, Nlgn3 has a heterogeneous expression and function at particular subsets of chemical synapses and strong association with non-syndromic autism spectrum disorder (ASD). Several lines of evidence have suggested that the unique expression and function of Nlgn3 protein underlie circuit-specific dysfunction characteristic of non-syndromic ASD caused by the disruption of Nlgn3 gene. Furthermore, recent studies have uncovered the molecular mechanism underlying input cell-dependent expression of Nlgn3 protein at hippocampal inhibitory synapses, in which trans-synaptic signaling of specific alternatively spliced isoforms of Nlgn3 and Nrxn plays a critical role. In this review article, we overview the molecular, anatomical, and physiological knowledge about Nlgn3, focusing on the circuit-specific function of mammalian Nlgn3 and its underlying molecular mechanism. This will provide not only new insight into specific Nlgn3-mediated trans-synaptic interactions as molecular codes for synapse specification but also a better understanding of the pathophysiological basis for non-syndromic ASD associated with functional impairment in Nlgn3 gene.
Collapse
Affiliation(s)
- Motokazu Uchigashima
- Department of Cellular Neuropathology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Amy Cheung
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester, MA, United States
| | - Kensuke Futai
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester, MA, United States
| |
Collapse
|
14
|
Blumenthal DK, Cheng X, Fajer M, Ho KY, Rohrer J, Gerlits O, Taylor P, Juneja P, Kovalevsky A, Radić Z. Covalent inhibition of hAChE by organophosphates causes homodimer dissociation through long-range allosteric effects. J Biol Chem 2021; 297:101007. [PMID: 34324828 PMCID: PMC8384907 DOI: 10.1016/j.jbc.2021.101007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 07/20/2021] [Accepted: 07/23/2021] [Indexed: 11/26/2022] Open
Abstract
Acetylcholinesterase (EC 3.1.1.7), a key acetylcholine-hydrolyzing enzyme in cholinergic neurotransmission, is present in a variety of states in situ, including monomers, C-terminally disulfide-linked homodimers, homotetramers, and up to three tetramers covalently attached to structural subunits. Could oligomerization that ensures high local concentrations of catalytic sites necessary for efficient neurotransmission be affected by environmental factors? Using small-angle X-ray scattering (SAXS) and cryo-EM, we demonstrate that homodimerization of recombinant monomeric human acetylcholinesterase (hAChE) in solution occurs through a C-terminal four-helix bundle at micromolar concentrations. We show that diethylphosphorylation of the active serine in the catalytic gorge or isopropylmethylphosphonylation by the RP enantiomer of sarin promotes a 10-fold increase in homodimer dissociation. We also demonstrate the dissociation of organophosphate (OP)-conjugated dimers is reversed by structurally diverse oximes 2PAM, HI6, or RS194B, as demonstrated by SAXS of diethylphosphoryl-hAChE. However, binding of oximes to the native ligand-free hAChE, binding of high-affinity reversible ligands, or formation of an SP-sarin-hAChE conjugate had no effect on homodimerization. Dissociation monitored by time-resolved SAXS occurs in milliseconds, consistent with rates of hAChE covalent inhibition. OP-induced dissociation was not observed in the SAXS profiles of the double-mutant Y337A/F338A, where the active center gorge volume is larger than in wildtype hAChE. These observations suggest a key role of the tightly packed acyl pocket in allosterically triggered OP-induced dimer dissociation, with the potential for local reduction of acetylcholine-hydrolytic power in situ. Computational models predict allosteric correlated motions extending from the acyl pocket toward the four-helix bundle dimerization interface 25 Å away.
Collapse
Affiliation(s)
- Donald K Blumenthal
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah, USA
| | - Xiaolin Cheng
- Division of Medicinal Chemistry & Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | - Mikolai Fajer
- Division of Medicinal Chemistry & Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | - Kwok-Yiu Ho
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UC San Diego, La Jolla, California, USA
| | - Jacqueline Rohrer
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UC San Diego, La Jolla, California, USA
| | - Oksana Gerlits
- Department of Natural Sciences, Tennessee Wesleyan University, Athens, Tennessee, USA
| | - Palmer Taylor
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UC San Diego, La Jolla, California, USA
| | - Puneet Juneja
- Cryo-EM Facility, Iowa State University, Ames, Iowa, USA
| | - Andrey Kovalevsky
- Neutron Scattering Division, Oak Ridge National Laboratory, Oak Ridge, Tennessee, USA
| | - Zoran Radić
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UC San Diego, La Jolla, California, USA.
| |
Collapse
|
15
|
Steffen DM, Ferri SL, Marcucci CG, Blocklinger KL, Molumby MJ, Abel T, Weiner JA. The γ-Protocadherins Interact Physically and Functionally with Neuroligin-2 to Negatively Regulate Inhibitory Synapse Density and Are Required for Normal Social Interaction. Mol Neurobiol 2021; 58:2574-2589. [PMID: 33471287 PMCID: PMC8137559 DOI: 10.1007/s12035-020-02263-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 12/15/2020] [Indexed: 12/16/2022]
Abstract
Cell adhesion molecules (CAMs) are key players in the formation of neural circuits during development. The γ-protocadherins (γ-Pcdhs), a family of 22 CAMs encoded by the Pcdhg gene cluster, are known to play important roles in dendrite arborization, axon targeting, and synapse development. We showed previously that multiple γ-Pcdhs interact physically with the autism-associated CAM neuroligin-1, and inhibit the latter's ability to promote excitatory synapse maturation. Here, we show that γ-Pcdhs can also interact physically with the related neuroligin-2, and inhibit this CAM's ability to promote inhibitory synapse development. In an artificial synapse assay, γ-Pcdhs co-expressed with neuroligin-2 in non-neuronal cells reduce inhibitory presynaptic maturation in contacting hippocampal axons. Mice lacking the γ-Pcdhs from the forebrain (including the cortex, the hippocampus, and portions of the amygdala) exhibit increased inhibitory synapse density and increased co-localization of neuroligin-2 with inhibitory postsynaptic markers in vivo. These Pcdhg mutants also exhibit defective social affiliation and an anxiety-like phenotype in behavioral assays. Together, these results suggest that γ-Pcdhs negatively regulate neuroligins to limit synapse density in a manner that is important for normal behavior.
Collapse
Affiliation(s)
- David M Steffen
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, 52242, USA
- Department of Biology, The University of Iowa, Iowa City, IA, 52242, USA
| | - Sarah L Ferri
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, 52242, USA
- Department of Neuroscience and Pharmacology, Carver College of Medicine, The University of Iowa, Iowa City, IA, 52242, USA
| | - Charles G Marcucci
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, 52242, USA
- Department of Biology, The University of Iowa, Iowa City, IA, 52242, USA
| | - Kelsey L Blocklinger
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, 52242, USA
- Department of Neuroscience and Pharmacology, Carver College of Medicine, The University of Iowa, Iowa City, IA, 52242, USA
| | - Michael J Molumby
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, 52242, USA
- Department of Biology, The University of Iowa, Iowa City, IA, 52242, USA
| | - Ted Abel
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, 52242, USA
- Department of Neuroscience and Pharmacology, Carver College of Medicine, The University of Iowa, Iowa City, IA, 52242, USA
| | - Joshua A Weiner
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, 52242, USA.
- Department of Biology, The University of Iowa, Iowa City, IA, 52242, USA.
| |
Collapse
|
16
|
Xue K, Hu Y, Gu S, Wang C, Kong R, Xie W, Li J. Using structural analysis to clarify the impact of single nucleotide variants in neurexin/neuroligin revealed in clinical genomic sequencing. J Biomol Struct Dyn 2021; 40:8085-8099. [PMID: 33818307 DOI: 10.1080/07391102.2021.1907225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The synapse is a highly specialized and dynamic structure, which is involved in regulating neurotransmission. Nerve cell adhesion molecule is a kind of transmembrane protein that mediates the interaction between cells and cells, cells and extracellular matrix, and plays a role in cell recognition, metastasis, and transmembrane signal transduction. Among nerve cell adhesion molecules, Neurexins (NRXNs) and Neuroligins (NLGNs) have been focused due to the relation with autism and other neuropsychiatric diseases. The previous research discovered numerous variants in NRXNs and NLGNs reported in neurodevelopmental disorders by genomic sequencing. However, structural variants in synaptic molecules caused by genome variants still prevent us from understanding the molecular mechanism of diseases. Thus, we sought to conduct a comprehensive risk assessment of the known NRXN and NLGN gene variants by protein structure analysis. In this study, we analyzed the structural properties of the NRXN/NLGN complex by calculating free energy in residue scanning, in combination with existing risk evaluation tools to focus on candidate missense mutations. Our calculations show that five candidate missense mutations in NLGNs can reduce the stability of NLGNs and even prevent the formation of NRXN/NLGN complexes, namely R87W, R204H, R437H, R437C and R583W. In addition, we found that the affinity of the amino acid substitution (Leu593Phe) (ΔΔG(affinity)) changes the affinity of the NLGN dimer. Overall, we have identified important potential pathological variants that provide clues to biomarkers.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Kaiyu Xue
- Key Laboratory of DGHD, MOE, Institute of Life Sciences, Southeast University, Nanjing, China
| | - Yunyun Hu
- Key Laboratory of DGHD, MOE, Institute of Life Sciences, Southeast University, Nanjing, China
| | - Shuanglin Gu
- Key Laboratory of DGHD, MOE, Institute of Life Sciences, Southeast University, Nanjing, China
| | - Chao Wang
- Key Laboratory of DGHD, MOE, Institute of Life Sciences, Southeast University, Nanjing, China
| | - Ren Kong
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou, China
| | - Wei Xie
- Key Laboratory of DGHD, MOE, Institute of Life Sciences, Southeast University, Nanjing, China
| | - Jian Li
- Key Laboratory of DGHD, MOE, Institute of Life Sciences, Southeast University, Nanjing, China
| |
Collapse
|
17
|
Yoshida T, Yamagata A, Imai A, Kim J, Izumi H, Nakashima S, Shiroshima T, Maeda A, Iwasawa-Okamoto S, Azechi K, Osaka F, Saitoh T, Maenaka K, Shimada T, Fukata Y, Fukata M, Matsumoto J, Nishijo H, Takao K, Tanaka S, Okabe S, Tabuchi K, Uemura T, Mishina M, Mori H, Fukai S. Canonical versus non-canonical transsynaptic signaling of neuroligin 3 tunes development of sociality in mice. Nat Commun 2021; 12:1848. [PMID: 33758193 PMCID: PMC7988105 DOI: 10.1038/s41467-021-22059-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 02/25/2021] [Indexed: 12/31/2022] Open
Abstract
Neuroligin 3 (NLGN3) and neurexins (NRXNs) constitute a canonical transsynaptic cell-adhesion pair, which has been implicated in autism. In autism spectrum disorder (ASD) development of sociality can be impaired. However, the molecular mechanism underlying NLGN3-mediated social development is unclear. Here, we identify non-canonical interactions between NLGN3 and protein tyrosine phosphatase δ (PTPδ) splice variants, competing with NRXN binding. NLGN3-PTPδ complex structure revealed a splicing-dependent interaction mode and competition mechanism between PTPδ and NRXNs. Mice carrying a NLGN3 mutation that selectively impairs NLGN3-NRXN interaction show increased sociability, whereas mice where the NLGN3-PTPδ interaction is impaired exhibit impaired social behavior and enhanced motor learning, with imbalance in excitatory/inhibitory synaptic protein expressions, as reported in the Nlgn3 R451C autism model. At neuronal level, the autism-related Nlgn3 R451C mutation causes selective impairment in the non-canonical pathway. Our findings suggest that canonical and non-canonical NLGN3 pathways compete and regulate the development of sociality.
Collapse
Affiliation(s)
- Tomoyuki Yoshida
- Department of Molecular Neuroscience, Faculty of Medicine, University of Toyama, Toyama, Japan. .,Research Center for Idling Brain Science, University of Toyama, Toyama, Japan. .,JST PRESTO, Saitama, Japan.
| | | | - Ayako Imai
- Department of Molecular Neuroscience, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Juhyon Kim
- Division of Bio-Information Engineering, Faculty of Engineering, University of Toyama, Toyama, Japan
| | - Hironori Izumi
- Department of Molecular Neuroscience, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Shogo Nakashima
- Department of System Emotional Science, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Tomoko Shiroshima
- Department of Anatomy, Kitasato University School of Medicine, Kanagawa, Japan
| | - Asami Maeda
- Research Institute for Diseases of Old Age, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shiho Iwasawa-Okamoto
- Department of Molecular Neuroscience, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Kenji Azechi
- Department of Molecular Neuroscience, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Fumina Osaka
- Center for Research and Education on Drug Discovery, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Takashi Saitoh
- Center for Research and Education on Drug Discovery, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Katsumi Maenaka
- Center for Research and Education on Drug Discovery, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan.,Laboratory of Biomolecular Science, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Takashi Shimada
- SHIMADZU Bioscience Research Partnership, Innovation Center, Shimadzu Scientific Instruments, Bothell, WA, USA
| | - Yuko Fukata
- Division of Membrane Physiology, Department of Molecular and Cellular Physiology, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Aichi, Japan
| | - Masaki Fukata
- Division of Membrane Physiology, Department of Molecular and Cellular Physiology, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Aichi, Japan
| | - Jumpei Matsumoto
- Research Center for Idling Brain Science, University of Toyama, Toyama, Japan.,Department of System Emotional Science, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Hisao Nishijo
- Research Center for Idling Brain Science, University of Toyama, Toyama, Japan.,Department of System Emotional Science, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Keizo Takao
- Research Center for Idling Brain Science, University of Toyama, Toyama, Japan.,Life Science Research Center, University of Toyama, Toyama, Japan
| | - Shinji Tanaka
- Department of Cellular Neurobiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shigeo Okabe
- Department of Cellular Neurobiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Katsuhiko Tabuchi
- JST PRESTO, Saitama, Japan.,Department of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano, Japan.,Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Nagano, Japan
| | - Takeshi Uemura
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Nagano, Japan.,Division of Gene Research, Research Center for Supports to Advanced Science, Shinshu University, Nagano, Japan
| | - Masayoshi Mishina
- Brain Science Laboratory, Research Organization of Science and Technology, Ritsumeikan University, Shiga, Japan
| | - Hisashi Mori
- Department of Molecular Neuroscience, Faculty of Medicine, University of Toyama, Toyama, Japan.,Research Center for Idling Brain Science, University of Toyama, Toyama, Japan
| | - Shuya Fukai
- Department of Chemistry, Graduate School of Science, Kyoto University, Kyoto, Japan.
| |
Collapse
|
18
|
Abstract
The function of neuronal circuits relies on the properties of individual neuronal cells and their synapses. We propose that a substantial degree of synapse formation and function is instructed by molecular codes resulting from transcriptional programmes. Recent studies on the Neurexin protein family and its ligands provide fundamental insight into how synapses are assembled and remodelled, how synaptic properties are specified and how single gene mutations associated with neurodevelopmental and psychiatric disorders might modify the operation of neuronal circuits and behaviour. In this Review, we first summarize insights into Neurexin function obtained from various model organisms. We then discuss the mechanisms and logic of the cell type-specific regulation of Neurexin isoforms, in particular at the level of alternative mRNA splicing. Finally, we propose a conceptual framework for how combinations of synaptic protein isoforms act as 'senders' and 'readers' to instruct synapse formation and the acquisition of cell type-specific and synapse-specific functional properties.
Collapse
|
19
|
The multiple biological roles of the cholinesterases. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2020; 162:41-56. [PMID: 33307019 DOI: 10.1016/j.pbiomolbio.2020.12.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 11/30/2020] [Accepted: 12/03/2020] [Indexed: 12/14/2022]
Abstract
It is tacitly assumed that the biological role of acetylcholinesterase is termination of synaptic transmission at cholinergic synapses. However, together with its structural homolog, butyrylcholinesterase, it is widely distributed both within and outside the nervous system, and, in many cases, the role of both enzymes remains obscure. The transient appearance of the cholinesterases in embryonic tissues is especially enigmatic. The two enzymes' extra-synaptic roles, which are known as 'non-classical' roles, are the topic of this review. Strong evidence has been presented that AChE and BChE play morphogenetic roles in a variety of eukaryotic systems, and they do so either by acting as adhesion proteins, or as trophic factors. As trophic factors, one mode of action is to directly regulate morphogenesis, such as neurite outgrowth, by poorly understood mechanisms. The other mode is by regulating levels of acetylcholine, which acts as the direct trophic factor. Alternate substrates have been sought for the cholinesterases. Quite recently, it was shown that levels of the aggression hormone, ghrelin, which also controls appetite, are regulated by butyrylcholinesterase. The rapid hydrolysis of acetylcholine by acetylcholinesterase generates high local proton concentrations. The possible biophysical and biological consequences of this effect are discussed. The biological significance of the acetylcholinesterases secreted by parasitic nematodes is reviewed, and, finally, the involvement of acetylcholinesterase in apoptosis is considered.
Collapse
|
20
|
An Autism-Associated Mutation Impairs Neuroligin-4 Glycosylation and Enhances Excitatory Synaptic Transmission in Human Neurons. J Neurosci 2020; 41:392-407. [PMID: 33268543 DOI: 10.1523/jneurosci.0404-20.2020] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 11/18/2020] [Accepted: 11/20/2020] [Indexed: 02/06/2023] Open
Abstract
Neuroligins (NLGNs) are a class of postsynaptic cell adhesion molecules that interact with presynaptic neurexins (NRXNs) and regulate synapse function. NLGN4 is a member of the NLGN family and consists of a unique amino acid sequence in humans that is not evolutionarily well conserved in rodents. The human-specific NLGN4 gene has been reported to be mutated in many patients with autism and other neurodevelopmental disorders. However, it remained unclear how these mutations might alter the molecular properties of NLGN4 and affect synaptic transmission in human neurons. Here, we describe a severely autistic male patient carrying a single amino acid substitution (R101Q) in the NLGN4 gene. When expressed in HEK293 cells, the R101Q mutation in NLGN4 did not affect its binding affinity for NRXNs or its capacity to form homodimers. This mutation, however, impaired the maturation of NLGN4 protein by inhibiting N-linked glycosylation at an adjacent residue (N102), which is conserved in all NLGNs. As a result, the R101Q substitution significantly decreased the surface trafficking of NLGN4 and increased its retention in the endoplasmic reticulum and Golgi apparatus. In human neurons derived from male stem cell lines, the R101Q mutation also similarly reduced the synaptic localization of NLGN4, resulting in a loss-of-function phenotype. This mutation-induced trafficking defect substantially diminished the ability of NLGN4 to form excitatory synapses and modulate their functional properties. Viewed together, our findings suggest that the R101Q mutation is pathogenic for NLGN4 and can lead to synaptic dysfunction in autism.
Collapse
|
21
|
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
|
22
|
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
|
23
|
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
|
24
|
Yumoto T, Kimura M, Nagatomo R, Sato T, Utsunomiya S, Aoki N, Kitaura M, Takahashi K, Takemoto H, Watanabe H, Okano H, Yoshida F, Nao Y, Tomita T. Autism-associated variants of neuroligin 4X impair synaptogenic activity by various molecular mechanisms. Mol Autism 2020; 11:68. [PMID: 32873342 PMCID: PMC7465329 DOI: 10.1186/s13229-020-00373-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 08/20/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Several genetic alterations, including point mutations and copy number variations in NLGN genes, have been associated with psychiatric disorders, such as autism spectrum disorder (ASD) and X-linked mental retardation (XLMR). NLGN genes encode neuroligin (NL) proteins, which are adhesion molecules that are important for proper synaptic formation and maturation. Previously, we and others found that the expression level of murine NL1 is regulated by proteolytic processing in a synaptic activity-dependent manner. METHODS In this study, we analyzed the effects of missense variants associated with ASD and XLMR on the metabolism and function of NL4X, a protein which is encoded by the NLGN4X gene and is expressed only in humans, using cultured cells, primary neurons from rodents, and human induced pluripotent stem cell-derived neurons. RESULTS NL4X was found to undergo proteolytic processing in human neuronal cells. Almost all NL4X variants caused a substantial decrease in the levels of mature NL4X and its synaptogenic activity in a heterologous culture system. Intriguingly, the L593F variant of NL4X accelerated the proteolysis of mature NL4X proteins located on the cell surface. In contrast, other variants decreased the cell-surface trafficking of NL4X. Notably, protease inhibitors as well as chemical chaperones rescued the expression of mature NL4X. LIMITATIONS Our study did not reveal whether these dysfunctional phenotypes occurred in individuals carrying NLGN4X variant. Moreover, though these pathological mechanisms could be exploited as potential drug targets for ASD, it remains unclear whether these compounds would have beneficial effects on ASD model animals and patients. CONCLUSIONS These data suggest that reduced amounts of the functional NL4X protein on the cell surface is a common mechanism by which point mutants of the NL4X protein cause psychiatric disorders, although different molecular mechanisms are thought to be involved. Furthermore, these results highlight that the precision medicine approach based on genetic and cell biological analyses is important for the development of therapeutics for psychiatric disorders.
Collapse
Affiliation(s)
- Takafumi Yumoto
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Misaki Kimura
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Ryota Nagatomo
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Tsukika Sato
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Shun Utsunomiya
- Neuroscience 2, Laboratory for Drug Discovery and Disease Research, Shionogi, Osaka, Japan
- Business-Academia Collaborative Laboratory (Shionogi), Graduate School of Pharmaceutical Science, The University of Tokyo, Tokyo, Japan
| | - Natsue Aoki
- Neuroscience 2, Laboratory for Drug Discovery and Disease Research, Shionogi, Osaka, Japan
- Business-Academia Collaborative Laboratory (Shionogi), Graduate School of Pharmaceutical Science, The University of Tokyo, Tokyo, Japan
| | - Motoji Kitaura
- Research Administration SPRC, R&D General Administration Unit, General Administration Division, Shionogi Administration Service, Osaka, Japan
| | - Koji Takahashi
- Drug Discovery Technology 3, Laboratory for Innovative Therapy Research, Shionogi, Osaka, Japan
| | - Hiroshi Takemoto
- Neuroscience 2, Laboratory for Drug Discovery and Disease Research, Shionogi, Osaka, Japan
- Business-Academia Collaborative Laboratory (Shionogi), Graduate School of Pharmaceutical Science, The University of Tokyo, Tokyo, Japan
| | - Hirotaka Watanabe
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Fumiaki Yoshida
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Yosuke Nao
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Taisuke Tomita
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| |
Collapse
|
25
|
Nguyen TA, Lehr AW, Roche KW. Neuroligins and Neurodevelopmental Disorders: X-Linked Genetics. Front Synaptic Neurosci 2020; 12:33. [PMID: 32848696 PMCID: PMC7431521 DOI: 10.3389/fnsyn.2020.00033] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 07/22/2020] [Indexed: 12/24/2022] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder that results in social-communication impairments, as well as restricted and repetitive behaviors. Moreover, ASD is more prevalent in males, with a male to female ratio of 4 to 1. Although the underlying etiology of ASD is generally unknown, recent advances in genome sequencing have facilitated the identification of a host of associated genes. Among these, synaptic proteins such as cell adhesion molecules have been strongly linked with ASD. Interestingly, many large genome sequencing studies exclude sex chromosomes, which leads to a shift in focus toward autosomal genes as targets for ASD research. However, there are many genes on the X chromosome that encode synaptic proteins, including strong candidate genes. Here, we review findings regarding two members of the neuroligin (NLGN) family of postsynaptic adhesion molecules, NLGN3 and NLGN4. Neuroligins have multiple isoforms (NLGN1-4), which are both autosomal and sex-linked. The sex-linked genes, NLGN3 and NLGN4, are both on the X chromosome and were among the first few genes to be linked with ASD and intellectual disability (ID). In addition, there is a less studied human neuroligin on the Y chromosome, NLGN4Y, which forms an X-Y pair with NLGN4X. We will discuss recent findings of these neuroligin isoforms regarding function at the synapse in both rodent models and human-derived differentiated neurons, and highlight the exciting challenges moving forward to a better understanding of ASD/ID.
Collapse
Affiliation(s)
- Thien A. Nguyen
- Receptor Biology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
- Department of Pharmacology and Physiology, Georgetown University, Washington, DC, United States
| | - Alexander W. Lehr
- Receptor Biology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Katherine W. Roche
- Receptor Biology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
26
|
Nguyen TA, Wu K, Pandey S, Lehr AW, Li Y, Bemben MA, Badger JD, Lauzon JL, Wang T, Zaghloul KA, Thurm A, Jain M, Lu W, Roche KW. A Cluster of Autism-Associated Variants on X-Linked NLGN4X Functionally Resemble NLGN4Y. Neuron 2020; 106:759-768.e7. [PMID: 32243781 DOI: 10.1016/j.neuron.2020.03.008] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 02/12/2020] [Accepted: 03/11/2020] [Indexed: 12/20/2022]
Abstract
Autism spectrum disorder (ASD) is more prevalent in males; however, the etiology for this sex bias is not well understood. Many mutations on X-linked cell adhesion molecule NLGN4X result in ASD or intellectual disability. NLGN4X is part of an X-Y pair, with NLGN4Y sharing ∼97% sequence homology. Using biochemistry, electrophysiology, and imaging, we show that NLGN4Y displays severe deficits in maturation, surface expression, and synaptogenesis regulated by one amino acid difference with NLGN4X. Furthermore, we identify a cluster of ASD-associated mutations surrounding the critical amino acid in NLGN4X, and these mutations phenocopy NLGN4Y. We show that NLGN4Y cannot compensate for the functional deficits observed in ASD-associated NLGN4X mutations. Altogether, our data reveal a potential pathogenic mechanism for male bias in NLGN4X-associated ASD.
Collapse
Affiliation(s)
- Thien A Nguyen
- Receptor Biology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA; Department of Pharmacology and Physiology, Georgetown University, Washington DC 20057, USA
| | - Kunwei Wu
- Synapse and Neural Circuit Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Saurabh Pandey
- Synapse and Neural Circuit Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Alexander W Lehr
- Receptor Biology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yan Li
- Protein/Peptide Sequencing Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michael A Bemben
- Receptor Biology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - John D Badger
- Receptor Biology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Julie L Lauzon
- Department of Medical Genetics and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Tongguang Wang
- Translational Neuroscience Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kareem A Zaghloul
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Audrey Thurm
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mahim Jain
- Department of Bone and OI, Kennedy Krieger Institute, Baltimore, MD 21205, USA
| | - Wei Lu
- Synapse and Neural Circuit Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Katherine W Roche
- Receptor Biology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
27
|
Spieker J, Frieß JL, Sperling L, Thangaraj G, Vogel-Höpker A, Layer PG. Cholinergic control of bone development and beyond. Int Immunopharmacol 2020; 83:106405. [PMID: 32208165 DOI: 10.1016/j.intimp.2020.106405] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 03/06/2020] [Accepted: 03/11/2020] [Indexed: 12/15/2022]
Abstract
There is ample evidence that cholinergic actions affect the health status of bones in vertebrates including man. Nicotine smoking, but also exposure to pesticides or medical drugs point to the significance of cholinergic effects on bone status, as reviewed here in Introduction. Then, we outline processes of endochondral ossification, and review respective cholinergic actions. In Results, we briefly summarize our in vivo and in vitro studies on bone development of chick and mouse [1,2], including (i) expressions of cholinergic components (AChE, BChE, ChAT) in chick embryo, (ii) characterisation of defects during skeletogenesis in prenatal ChE knockout mice, (iii) loss-of-function experiments with beads soaked in cholinergic components and implanted into chicken limb buds, and finally (iv) we use an in vitro mesenchymal 3D-micromass model that mimics cartilage and bone formation, which also had revealed complex crosstalks between cholinergic, radiation and inflammatory mechanisms [3]. In Discussion, we evaluate non-cholinergic actions of cholinesterases during bone formation by considering: (i) how cholinesterases could function in adhesive mechanisms; (ii) whether and how cholinesterases can form bone-regulatory complexes with alkaline phosphatase (ALP) and/or ECM components, which could regulate cell division, migration and adhesion. We conclude that cholinergic actions in bone development are driven mainly by classic cholinergic, but non-neural cycles (e.g., by acetylcholine); in addition, both cholinesterases can exert distinct ACh-independent roles. Considering their tremendous medical impact, these results bring forward novel research directions that deserve to be pursued.
Collapse
Affiliation(s)
- Janine Spieker
- Developmental Biology and Neurogenetics, Technische Universität Darmstadt, Schnittspahnstrasse 13, D-64287 Darmstadt, Germany
| | - Johannes L Frieß
- Developmental Biology and Neurogenetics, Technische Universität Darmstadt, Schnittspahnstrasse 13, D-64287 Darmstadt, Germany
| | - Laura Sperling
- Developmental Biology and Neurogenetics, Technische Universität Darmstadt, Schnittspahnstrasse 13, D-64287 Darmstadt, Germany
| | - Gopenath Thangaraj
- Developmental Biology and Neurogenetics, Technische Universität Darmstadt, Schnittspahnstrasse 13, D-64287 Darmstadt, Germany
| | - Astrid Vogel-Höpker
- Developmental Biology and Neurogenetics, Technische Universität Darmstadt, Schnittspahnstrasse 13, D-64287 Darmstadt, Germany
| | - Paul G Layer
- Developmental Biology and Neurogenetics, Technische Universität Darmstadt, Schnittspahnstrasse 13, D-64287 Darmstadt, Germany.
| |
Collapse
|
28
|
Bhandari R, Paliwal JK, Kuhad A. Neuropsychopathology of Autism Spectrum Disorder: Complex Interplay of Genetic, Epigenetic, and Environmental Factors. ADVANCES IN NEUROBIOLOGY 2020; 24:97-141. [PMID: 32006358 DOI: 10.1007/978-3-030-30402-7_4] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Autism spectrum disorder (ASD) is a complex heterogeneous consortium of pervasive development disorders (PDD) which ranges from atypical autism, autism, and Asperger syndrome affecting brain in the developmental stage. This debilitating neurodevelopmental disorder results in both core as well as associated symptoms. Core symptoms observed in autistic patients are lack of social interaction, pervasive, stereotyped, and restricted behavior while the associated symptoms include irritability, anxiety, aggression, and several comorbid disorders.ASD is a polygenic disorder and is multifactorial in origin. Copy number variations (CNVs) of several genes that regulate the synaptogenesis and signaling pathways are one of the major factors responsible for the pathogenesis of autism. The complex integration of various CNVs cause mutations in the genes which code for molecules involved in cell adhesion, voltage-gated ion-channels, scaffolding proteins as well as signaling pathways (PTEN and mTOR pathways). These mutated genes are responsible for affecting synaptic transmission by causing plasticity dysfunction responsible, in turn, for the expression of ASD.Epigenetic modifications affecting DNA transcription and various pre-natal and post-natal exposure to a variety of environmental factors are also precipitating factors for the occurrence of ASD. All of these together cause dysregulation of glutamatergic signaling as well as imbalance in excitatory: inhibitory pathways resulting in glial cell activation and release of inflammatory mediators responsible for the aberrant social behavior which is observed in autistic patients.In this chapter we review and provide insight into the intricate integration of various genetic, epigenetic, and environmental factors which play a major role in the pathogenesis of this disorder and the mechanistic approach behind this integration.
Collapse
Affiliation(s)
- Ranjana Bhandari
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Study, Panjab University, Chandigarh, India
| | - Jyoti K Paliwal
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Study, Panjab University, Chandigarh, India
| | - Anurag Kuhad
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Study, Panjab University, Chandigarh, India.
| |
Collapse
|
29
|
Zhang Y, Wang L, Li Z, Chen D, Han W, Wu Z, Shang F, Hai E, Wei Y, Su R, Liu Z, Wang R, Wang Z, Zhao Y, Wang Z, Zhang Y, Li J. Transcriptome profiling reveals transcriptional and alternative splicing regulation in the early embryonic development of hair follicles in the cashmere goat. Sci Rep 2019; 9:17735. [PMID: 31780728 PMCID: PMC6882815 DOI: 10.1038/s41598-019-54315-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 10/31/2019] [Indexed: 01/30/2023] Open
Abstract
The undercoat fiber of the cashmere goat, from the secondary hair follicle (HF), possesses commercial value. However, very few studies have focused on the molecular details of primary and secondary HF initiation and development in goat embryos. In this study, skin samples at embryonic day 45, 55, and 65 (E45, E55, and E65) were collected and prepared for RNA sequencing (RNA-seq). We found that the HF probably initiated from E55 to E65 by analyzing the functional pathways of differentially expressed genes (DEGs). Most key genes in canonical signaling pathways, including WNT, TGF-β, FGF, Hedgehog, NOTCH, and other factors showed clear expression changes from E55 to E65. We, for the first time, explored alternative splicing (AS) alterations, which showed distinct patterns among these three stages. Functional pathways of AS-regulated genes showed connections to HF development. By comparing the published RNA-seq samples from the E60, E120, and newborn (NB) stages, we found the majority of WNT/β-catenin signaling genes were important in the initiation of HF development, while other factors including FOXN1, GATA3, and DLX3 may have a consistent influence on HF development. Our investigation supported the time points of embryonic HF initiation and identified genes that have potential functions of embryonic HF initiation and development. We further explored the potential regulatory roles of AS in HF initiation, which extended our knowledge about the molecular mechanisms of HF development.
Collapse
Affiliation(s)
- Yanjun Zhang
- College of Animal Science, Inner Mongolia Agricultural University, 010018, Hohhot, Inner Mongolia Autonomous Region, China
| | - Lele Wang
- Ulanqab Medical College, 010020, Ulanqab, Inner Mongolia Autonomous Region, China
| | - Zhen Li
- Center for Genome Analysis, ABLife Inc., Wuhan, Hubei, 430072, China
| | - Dong Chen
- Center for Genome Analysis, ABLife Inc., Wuhan, Hubei, 430072, China
| | - Wenjing Han
- College of Animal Science, Inner Mongolia Agricultural University, 010018, Hohhot, Inner Mongolia Autonomous Region, China
| | - Zhihong Wu
- College of Animal Science, Inner Mongolia Agricultural University, 010018, Hohhot, Inner Mongolia Autonomous Region, China
| | - Fangzheng Shang
- College of Animal Science, Inner Mongolia Agricultural University, 010018, Hohhot, Inner Mongolia Autonomous Region, China
| | - Erhan Hai
- College of Animal Science, Inner Mongolia Agricultural University, 010018, Hohhot, Inner Mongolia Autonomous Region, China
| | - Yaxun Wei
- Center for Genome Analysis, ABLife Inc., Wuhan, Hubei, 430072, China
| | - Rui Su
- College of Animal Science, Inner Mongolia Agricultural University, 010018, Hohhot, Inner Mongolia Autonomous Region, China
| | - Zhihong Liu
- College of Animal Science, Inner Mongolia Agricultural University, 010018, Hohhot, Inner Mongolia Autonomous Region, China
| | - Ruijun Wang
- College of Animal Science, Inner Mongolia Agricultural University, 010018, Hohhot, Inner Mongolia Autonomous Region, China
| | - Zhiying Wang
- College of Animal Science, Inner Mongolia Agricultural University, 010018, Hohhot, Inner Mongolia Autonomous Region, China
| | - Yanhong Zhao
- College of Animal Science, Inner Mongolia Agricultural University, 010018, Hohhot, Inner Mongolia Autonomous Region, China
| | - Zhixin Wang
- College of Animal Science, Inner Mongolia Agricultural University, 010018, Hohhot, Inner Mongolia Autonomous Region, China
| | - Yi Zhang
- Center for Genome Analysis, ABLife Inc., Wuhan, Hubei, 430072, China.
| | - Jinquan Li
- College of Animal Science, Inner Mongolia Agricultural University, 010018, Hohhot, Inner Mongolia Autonomous Region, China.
- Key Laboratory of Mutton Sheep Genetics and Breeding, Ministry of Agriculture, 010018, Hohhot, Inner Mongolia Autonomous Region, China.
- Key Laboratory of Animal Genetics, Breeding and Reproduction in Inner Mongolia Autonomous Region, 010018, Hohhot, Inner Mongolia Autonomous Region, China.
- Engineering Research Center for Goat Genetics and Breeding, Inner Mongolia Autonomous Region, 010018, Hohhot, Inner Mongolia Autonomous Region, China.
| |
Collapse
|
30
|
Pronker MF, van den Hoek H, Janssen BJC. Design and structural characterisation of olfactomedin-1 variants as tools for functional studies. BMC Mol Cell Biol 2019; 20:50. [PMID: 31726976 PMCID: PMC6857237 DOI: 10.1186/s12860-019-0232-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 10/10/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Olfactomedin-1 (Olfm1; also known as Noelin or Pancortin) is a highly-expressed secreted brain and retina protein and its four isoforms have different roles in nervous system development and function. Structural studies showed that the long Olfm1 isoform BMZ forms a disulfide-linked tetramer with a V-shaped architecture. The tips of the Olfm1 "V" each consist of two C-terminal β-propeller domains that enclose a calcium binding site. Functional characterisation of Olfm1 may be aided by new biochemical tools derived from these core structural elements. RESULTS Here we present the production, purification and structural analysis of three novel monomeric, dimeric and tetrameric forms of mammalian Olfm1 for functional studies. We characterise these constructs structurally by high-resolution X-ray crystallography and small-angle X-ray scattering. The crystal structure of the Olfm1 β-propeller domain (to 1.25 Å) represents the highest-resolution structure of an olfactomedin family member to date, revealing features such as a hydrophilic tunnel containing water molecules running into the core of the domain where the calcium binding site resides. The shorter Olfactomedin-1 isoform BMY is a disulfide-linked tetramer with a shape similar to the corresponding region in the longer BMZ isoform. CONCLUSIONS These recombinantly-expressed protein tools should assist future studies, for example of biophysical, electrophysiological or morphological nature, to help elucidate the functions of Olfm1 in the mature mammalian brain. The control over the oligomeric state of Olfm1 provides a firm basis to better understand the role of Olfm1 in the (trans-synaptic) tethering or avidity-mediated clustering of synaptic receptors such as post-synaptic AMPA receptors and pre-synaptic amyloid precursor protein. In addition, the variation in domain composition of these protein tools provides a means to dissect the Olfm1 regions important for receptor binding.
Collapse
Affiliation(s)
- Matti F Pronker
- MRC Laboratory of Molecular Biology, Division of Neurobiology, Francis Crick Avenue, Cambridge, CB2 0QH, UK. .,Bijvoet Center for Biomolecular Research, Utrecht University, Crystal and Structural Chemistry, Kruytgebouw, Padualaan 8, 3584 CH, Utrecht, The Netherlands.
| | - Hugo van den Hoek
- Bijvoet Center for Biomolecular Research, Utrecht University, Crystal and Structural Chemistry, Kruytgebouw, Padualaan 8, 3584 CH, Utrecht, The Netherlands.,Department of Molecular Structural Biology, Max Planck institute for Biochemistry, Am Klopferspitz 18, 82152, Martinsried, Germany
| | - Bert J C Janssen
- Bijvoet Center for Biomolecular Research, Utrecht University, Crystal and Structural Chemistry, Kruytgebouw, Padualaan 8, 3584 CH, Utrecht, The Netherlands.
| |
Collapse
|
31
|
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
|
32
|
Karki S, Maksimainen MM, Lehtiö L, Kajander T. Inhibitor screening assay for neurexin-LRRTM adhesion protein interaction involved in synaptic maintenance and neurological disorders. Anal Biochem 2019; 587:113463. [PMID: 31574254 DOI: 10.1016/j.ab.2019.113463] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 09/26/2019] [Accepted: 09/26/2019] [Indexed: 12/28/2022]
Abstract
Synaptic adhesion molecules, including presynaptic neurexins (NRXNs) and post-synaptic leucine-rich repeat transmembrane (LRRTM) proteins are important for development and maintenance of brain neuronal networks. NRXNs are probably the best characterized synaptic adhesion molecules, and one of the major presynaptic organizer proteins. The LRRTMs were found as ligands for NRXNs. Many of the synaptic adhesion proteins have been linked to neurological cognitive disorders, such as schizophrenia and autism spectrum disorders, making them targets of interest for both biological studies, and towards drug development. Therefore, we decided to develop a screening method to target the adhesion proteins, here the LRRTM-NRXN interaction, to find small molecule probes for further studies in cellular settings. To our knowledge, no potent small molecule compounds against the neuronal synaptic adhesion proteins are available. We utilized the AlphaScreen technology, and developed an assay targeting the NRXN-LRRTM2 interaction. We carried out screening of 2000 compounds and identified hits with moderate IC50-values. We also established an orthogonal in-cell Western blot assay to validate hits. This paves way for future development of specific high affinity compounds by further high throughput screening of larger compound libraries using the methods established here. The method could also be applied to screening other NRXN-ligand interactions.
Collapse
Affiliation(s)
- Sudeep Karki
- Institute of Biotechnology, University of Helsinki, Finland
| | - Mirko M Maksimainen
- Faculty of Biochemistry and Molecular Medicine and Biocenter Oulu University of Oulu, Finland
| | - Lari Lehtiö
- Faculty of Biochemistry and Molecular Medicine and Biocenter Oulu University of Oulu, Finland
| | - Tommi Kajander
- Institute of Biotechnology, University of Helsinki, Finland.
| |
Collapse
|
33
|
Kiss-Szemán AJ, Harmat V, Menyhárd DK. Achieving Functionality Through Modular Build-up: Structure and Size Selection of Serine Oligopeptidases. Curr Protein Pept Sci 2019; 20:1089-1101. [PMID: 31553292 DOI: 10.2174/1389203720666190925103339] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 03/13/2019] [Accepted: 04/12/2019] [Indexed: 01/13/2023]
Abstract
Enzymes of the prolyl oligopeptidase family (S9 family) recognize their substrates not only by the specificity motif to be cleaved but also by size - they hydrolyze oligopeptides smaller than 30 amino acids. They belong to the serine-protease family, but differ from classical serine-proteases in size (80 kDa), structure (two domains) and regulation system (size selection of substrates). This group of enzymes is an important target for drug design as they are linked to amnesia, schizophrenia, type 2 diabetes, trypanosomiasis, periodontitis and cell growth. By comparing the structure of various members of the family we show that the most important features contributing to selectivity and efficiency are: (i) whether the interactions weaving the two domains together play a role in stabilizing the catalytic triad and thus their absence may provide for its deactivation: these oligopeptidases can screen their substrates by opening up, and (ii) whether the interaction-prone β-edge of the hydrolase domain is accessible and thus can guide a multimerization process that creates shielded entrance or intricate inner channels for the size-based selection of substrates. These cornerstones can be used to estimate the multimeric state and selection strategy of yet undetermined structures.
Collapse
Affiliation(s)
- Anna J Kiss-Szemán
- Laboratory of Structural Chemistry and Biology, Institute of Chemistry, Eotvos Lorand University, Budapest, Hungary
| | - Veronika Harmat
- Laboratory of Structural Chemistry and Biology, Institute of Chemistry, Eotvos Lorand University, Budapest, Hungary.,MTA-ELTE Protein Modelling Research Group, Eötvös Loránd University, Budapest, Hungary
| | - Dóra K Menyhárd
- MTA-ELTE Protein Modelling Research Group, Eotvos Lorand University, Budapest, Hungary
| |
Collapse
|
34
|
Chatonnet A, Brazzolotto X, Hotelier T, Lenfant N, Marchot P, Bourne Y. An evolutionary perspective on the first disulfide bond in members of the cholinesterase-carboxylesterase (COesterase) family: Possible outcomes for cholinesterase expression in prokaryotes. Chem Biol Interact 2019; 308:179-184. [DOI: 10.1016/j.cbi.2019.05.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 04/16/2019] [Accepted: 05/13/2019] [Indexed: 02/07/2023]
|
35
|
Phenotypic and molecular features underlying neurodegeneration of motor neurons derived from spinal and bulbar muscular atrophy patients. Neurobiol Dis 2019; 124:1-13. [DOI: 10.1016/j.nbd.2018.10.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Revised: 10/11/2018] [Accepted: 10/28/2018] [Indexed: 12/17/2022] Open
|
36
|
Neurexins - versatile molecular platforms in the synaptic cleft. Curr Opin Struct Biol 2019; 54:112-121. [PMID: 30831539 DOI: 10.1016/j.sbi.2019.01.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 01/22/2019] [Accepted: 01/24/2019] [Indexed: 01/05/2023]
Abstract
Neurexins constitute a large family of synaptic organizers. Their extracellular domains protrude into the synaptic cleft where they can form transsynaptic bridges with different partners. A unique constellation of structural elements within their ectodomains enables neurexins to create molecular platforms within the synaptic cleft that permit a large portfolio of partners to be recruited, assembled and their interactions to be dynamically regulated. Neurexins and their partners are implicated in neuropsychiatric diseases including autism spectrum disorder and schizophrenia. Detailed understanding of the mechanisms that underlie neurexin interactions may in future guide the design of tools to manipulate synaptic connections and their function, in particular those involved in the pathogenesis of neuropsychiatric disease.
Collapse
|
37
|
Chamma I, Sainlos M, Thoumine O. Biophysical mechanisms underlying the membrane trafficking of synaptic adhesion molecules. Neuropharmacology 2019; 169:107555. [PMID: 30831159 DOI: 10.1016/j.neuropharm.2019.02.037] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 02/14/2019] [Accepted: 02/27/2019] [Indexed: 01/13/2023]
Abstract
Adhesion proteins play crucial roles at synapses, not only by providing a physical trans-synaptic linkage between axonal and dendritic membranes, but also by connecting to functional elements including the pre-synaptic neurotransmitter release machinery and post-synaptic receptors. To mediate these functions, adhesion proteins must be organized on the neuronal surface in a precise and controlled manner. Recent studies have started to describe the mobility, nanoscale organization, and turnover rate of key synaptic adhesion molecules including cadherins, neurexins, neuroligins, SynCAMs, and LRRTMs, and show that some of these proteins are highly mobile in the plasma membrane while others are confined at sub-synaptic compartments, providing evidence for different regulatory pathways. In this review article, we provide a biophysical view of the diffusional trapping of adhesion molecules at synapses, involving both extracellular and intracellular protein interactions. We review the methodology underlying these measurements, including biomimetic systems with purified adhesion proteins, means to perturb protein expression or function, single molecule imaging in cultured neurons, and analytical models to interpret the data. This article is part of the special issue entitled 'Mobility and trafficking of neuronal membrane proteins'.
Collapse
Affiliation(s)
- Ingrid Chamma
- Univ. Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, F-33000, Bordeaux, France; CNRS, Interdisciplinary Institute for Neuroscience, UMR 5297, F-33000, Bordeaux, France
| | - Matthieu Sainlos
- Univ. Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, F-33000, Bordeaux, France; CNRS, Interdisciplinary Institute for Neuroscience, UMR 5297, F-33000, Bordeaux, France
| | - Olivier Thoumine
- Univ. Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, F-33000, Bordeaux, France; CNRS, Interdisciplinary Institute for Neuroscience, UMR 5297, F-33000, Bordeaux, France.
| |
Collapse
|
38
|
Connor SA, Elegheert J, Xie Y, Craig AM. Pumping the brakes: suppression of synapse development by MDGA-neuroligin interactions. Curr Opin Neurobiol 2019; 57:71-80. [PMID: 30771697 DOI: 10.1016/j.conb.2019.01.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 01/03/2019] [Accepted: 01/04/2019] [Indexed: 12/22/2022]
Abstract
Synapse development depends on a dynamic balance between synapse promoters and suppressors. MDGAs, immunoglobulin superfamily proteins, negatively regulate synapse development through blocking neuroligin-neurexin interactions. Recent analyses of MDGA-neuroligin complexes revealed the structural basis of this activity and indicate that MDGAs interact with all neuroligins with differential affinities. Surprisingly, analyses of mouse mutants revealed a functional divergence, with targeted mutation of Mdga1 and Mdga2 elevating inhibitory and excitatory synapses, respectively, on hippocampal pyramidal neurons. Further research is needed to determine the synapse-specific organizing properties of MDGAs in neural circuits, which may depend on relative levels and subcellular distributions of each MDGA, neuroligin and neurexin. Behavioral deficits in Mdga mutant mice support genetic links to schizophrenia and autism spectrum disorders and raise the possibility of harnessing these interactions for therapeutic purposes.
Collapse
Affiliation(s)
- Steven A Connor
- Department of Biology, York University, 4700 Keele Street, Toronto, ON, M3J 1P3, Canada.
| | - Jonathan Elegheert
- Interdisciplinary Institute for Neuroscience, CNRS UMR 5297 and University of Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France
| | - Yicheng Xie
- The Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China
| | - Ann Marie Craig
- Djavad Mowafaghian Centre for Brain Health and Department of Psychiatry, University of British Columbia, 2211 Wesbrook Mall, Vancouver, BC, V6T 2B5, Canada.
| |
Collapse
|
39
|
Dimitriou PS, Denesyuk AI, Nakayama T, Johnson MS, Denessiouk K. Distinctive structural motifs co-ordinate the catalytic nucleophile and the residues of the oxyanion hole in the alpha/beta-hydrolase fold enzymes. Protein Sci 2018; 28:344-364. [PMID: 30311984 DOI: 10.1002/pro.3527] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/04/2018] [Accepted: 10/08/2018] [Indexed: 12/17/2022]
Abstract
The alpha/beta-hydrolases (ABH) are among the largest structural families of proteins that are found in nature. Although they vary in their sequence and function, the ABH enzymes use a similar acid-base-nucleophile catalytic mechanism to catalyze reactions on different substrates. Because ABH enzymes are biocatalysts with a wide range of potential applications, protein engineering has taken advantage of their catalytic versatility to develop enzymes with industrial applications. This study is a comprehensive analysis of 40 ABH enzyme families focusing on two identified substructures: the nucleophile zone and the oxyanion zone, which co-ordinate the catalytic nucleophile and the residues of the oxyanion hole, and independently reported as critical for the enzymatic activity. We also frequently observed an aromatic cluster near the nucleophile and oxyanion zones, and opposite the ligand-binding site. The nucleophile zone, the oxyanion zone and the residue cluster enriched in aromatic side chains comprise a three-dimensional structural organization that shapes the active site of ABH enzymes and plays an important role in the enzymatic function by structurally stabilizing the catalytic nucleophile and the residues of the oxyanion hole. The structural data support the notion that the aromatic cluster can participate in co-ordination of the catalytic histidine loop, and properly place the catalytic histidine next to the catalytic nucleophile.
Collapse
Affiliation(s)
- Polytimi S Dimitriou
- Structural Bioinformatics Laboratory, Biochemistry, Faculty of Science and Engineering, Åbo Akademi University, Turku, 20520, Finland
| | - Alexander I Denesyuk
- Structural Bioinformatics Laboratory, Biochemistry, Faculty of Science and Engineering, Åbo Akademi University, Turku, 20520, Finland.,Institute for Biological Instrumentation of the Russian Academy of Sciences, Pushchino, 142290, Russia
| | - Toru Nakayama
- Tohoku University, Biomolecular Engineering, Sendai, Miyagi, 980-8579, Japan
| | - Mark S Johnson
- Structural Bioinformatics Laboratory, Biochemistry, Faculty of Science and Engineering, Åbo Akademi University, Turku, 20520, Finland
| | - Konstantin Denessiouk
- Structural Bioinformatics Laboratory, Biochemistry, Faculty of Science and Engineering, Åbo Akademi University, Turku, 20520, Finland.,Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Pharmacy, Åbo Akademi University, Turku, 20520, Finland
| |
Collapse
|
40
|
Liu J, Misra A, Reddy MVVVS, White MA, Ren G, Rudenko G. Structural Plasticity of Neurexin 1α: Implications for its Role as Synaptic Organizer. J Mol Biol 2018; 430:4325-4343. [PMID: 30193986 PMCID: PMC6223652 DOI: 10.1016/j.jmb.2018.08.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 08/27/2018] [Accepted: 08/29/2018] [Indexed: 11/24/2022]
Abstract
α-Neurexins are synaptic organizing molecules implicated in neuropsychiatric disorders. They bind and arrange an array of different partners in the synaptic cleft. The extracellular region of neurexin 1α (n1α) contains six LNS domains (L1-L6) interspersed by three Egf-like repeats. N1α must encode highly evolved structure-function relationships in order to fit into the narrow confines of the synaptic cleft, and also recruit its large, membrane-bound partners. Internal molecular flexibility could provide a solution; however, it is challenging to delineate because currently no structural methods permit high-resolution structure determination of large, flexible, multi-domain protein molecules. To investigate the structural plasticity of n1α, in particular the conformation of domains that carry validated binding sites for different protein partners, we used a panel of structural techniques. Individual particle electron tomography revealed that the N-terminally and C-terminally tethered domains, L1 and L6, have a surprisingly limited range of conformational freedom with respect to the linear central core containing L2 through L5. A 2.8-Å crystal structure revealed an unexpected arrangement of the L2 and L3 domains. Small-angle X-ray scattering and electron tomography indicated that incorporation of the alternative splice insert SS6 relieves the restricted conformational freedom between L5 and L6, suggesting that SS6 may work as a molecular toggle. The architecture of n1α thus encodes a combination of rigid and flexibly tethered domains that are uniquely poised to work together to promote its organizing function in the synaptic cleft, and may permit allosterically regulated and/or concerted protein partner binding.
Collapse
Affiliation(s)
- Jianfang Liu
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA; Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Anurag Misra
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - M V V V Sekhar Reddy
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Mark Andrew White
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA; Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Gang Ren
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA.
| | - Gabby Rudenko
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA; Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX 77555, USA.
| |
Collapse
|
41
|
Dynamics, nanoscale organization, and function of synaptic adhesion molecules. Mol Cell Neurosci 2018; 91:95-107. [DOI: 10.1016/j.mcn.2018.04.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 04/12/2018] [Accepted: 04/13/2018] [Indexed: 12/13/2022] Open
|
42
|
Yu Y, Kong R, Cao H, Yin Z, Liu J, Nan X, Phan AT, Ding T, Zhao H, Wong ST. Two birds, one stone: hesperetin alleviates chemotherapy-induced diarrhea and potentiates tumor inhibition. Oncotarget 2018; 9:27958-27973. [PMID: 29963254 PMCID: PMC6021345 DOI: 10.18632/oncotarget.24563] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 10/30/2017] [Indexed: 12/14/2022] Open
Abstract
Chemotherapy-induced diarrhea (CID), with clinical high incidence, adversely affects the efficacy of cancer treatment and patients' quality of life. Our study demonstrates that the citrus flavonoid hesperetin (Hst) has a superior potential as a new agent to prevent and alleviate CID. In the animal model for irinotecan (CPT-11) induced CID, Hst could selectively inhibit intestinal carboxylesterase (CES2) and thus reduce the local conversion of CPT-11 to cytotoxic SN-38 which causes intestinal toxicity. Oral administration of Hst manifested an excellent anti-diarrhea efficacy, prohibiting 80% of severe and 100% of mild diarrhea in the CPT-11 administered tumor-bearing mice. In addition, a significant attenuation of intestinal inflammation contributed to the anti-diarrhea effect of Hst. Moreover, Hst was found to work synergistically with CPT-11 in tumor inhibition by suppressing the tumor's STAT3 activity and recruiting tumoricidal macrophages into the tumor microenvironment. The anti-intestinal inflammation and anti-STAT3 properties of Hst would contribute its broad benefits to the management of diarrhea caused by other chemo or targeted agents, and more importantly, enhance and reinforce the anti-tumor effects of these agents, to improve patient outcomes.
Collapse
Affiliation(s)
- Yaping Yu
- Department of Systems Medicine and Bioengineering, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX, 77030, USA
- Department of Gynecology and Obstetrics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Ren Kong
- Department of Systems Medicine and Bioengineering, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX, 77030, USA
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou, Jiangsu, 213001, P.R. China
| | - Huojun Cao
- Department of Systems Medicine and Bioengineering, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX, 77030, USA
- Iowa Institute for Oral Health Research, College of Dentistry, The University of Iowa, Iowa City, IA, 52246, USA
| | - Zheng Yin
- Department of Systems Medicine and Bioengineering, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX, 77030, USA
| | - Jiyong Liu
- Department of Systems Medicine and Bioengineering, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX, 77030, USA
- Department of Pharmacy, Changhai Hospital, Shanghai, 200433, P.R. China
| | - Xiang Nan
- Department of Systems Medicine and Bioengineering, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX, 77030, USA
- Center for Biomedical Engineering, Department of Electronic Science and Technology, University of Science and Technology of China, Hefei, Anhui, 230026, P.R. China
| | - Alexandria T. Phan
- Cancer Treatment Centers of America at South Eastern Regional Center, Atlanta, GA, 30265, USA
| | - Tian Ding
- Department of Systems Medicine and Bioengineering, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX, 77030, USA
| | - Hong Zhao
- Department of Systems Medicine and Bioengineering, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX, 77030, USA
| | - Stephen T.C. Wong
- Department of Systems Medicine and Bioengineering, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX, 77030, USA
- Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX, 77030, USA
| |
Collapse
|
43
|
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
|
44
|
Südhof TC. Synaptic Neurexin Complexes: A Molecular Code for the Logic of Neural Circuits. Cell 2017; 171:745-769. [PMID: 29100073 DOI: 10.1016/j.cell.2017.10.024] [Citation(s) in RCA: 544] [Impact Index Per Article: 68.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 10/04/2017] [Accepted: 10/15/2017] [Indexed: 10/18/2022]
Abstract
Synapses are specialized junctions between neurons in brain that transmit and compute information, thereby connecting neurons into millions of overlapping and interdigitated neural circuits. Here, we posit that the establishment, properties, and dynamics of synapses are governed by a molecular logic that is controlled by diverse trans-synaptic signaling molecules. Neurexins, expressed in thousands of alternatively spliced isoforms, are central components of this dynamic code. Presynaptic neurexins regulate synapse properties via differential binding to multifarious postsynaptic ligands, such as neuroligins, cerebellin/GluD complexes, and latrophilins, thereby shaping the input/output relations of their resident neural circuits. Mutations in genes encoding neurexins and their ligands are associated with diverse neuropsychiatric disorders, especially schizophrenia, autism, and Tourette syndrome. Thus, neurexins nucleate an overall trans-synaptic signaling network that controls synapse properties, which thereby determines the precise responses of synapses to spike patterns in a neuron and circuit and which is vulnerable to impairments in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Thomas C Südhof
- Department of Molecular and Cellular Physiology and Howard Hughes Medical Institute, Stanford University Medical School, 265 Campus Drive, CA 94305-5453, USA.
| |
Collapse
|
45
|
Scafuri B, Varriale A, Facchiano A, D'Auria S, Raggi ME, Marabotti A. Binding of mycotoxins to proteins involved in neuronal plasticity: a combined in silico/wet investigation. Sci Rep 2017; 7:15156. [PMID: 29123130 PMCID: PMC5680308 DOI: 10.1038/s41598-017-15148-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 10/16/2017] [Indexed: 12/27/2022] Open
Abstract
We have applied a combined computational procedure based on inverse and direct docking in order to identify putative protein targets of a panel of mycotoxins and xenobiotic compounds that can contaminate food and that are known to have several detrimental effects on human health. This procedure allowed us to identify a panel of human proteins as possible targets for aflatoxins, gliotoxin, ochratoxin A and deoxynivalenol. Steady-state fluorescence and microscale thermophoresis experiments allowed us to confirm the binding of some of these mycotoxins to acetylcholinesterase and X-linked neuroligin 4, two proteins involved in synapse activity and, particularly for the second protein, neuronal plasticity and development. Considering the possible involvement of X-linked neuroligin 4 in the etiopathogenesis of autism spectrum syndrome, this finding opens up a new avenue to explore the hypothetical role of these xenobiotic compounds in the onset of this pathology.
Collapse
Affiliation(s)
- Bernardina Scafuri
- CNR-ISA, National Research Council, Institute of Food Science, Via Roma 64, 83100, Avellino, Italy
- Scientific Institute, IRCCS "Eugenio Medea" Bosisio Parini, Via Don Luigi Monza 20, 23842, Bosisio Parini, LC, Italy
- Department of Chemistry and Biology, "A. Zambelli", University of Salerno, Via Giovanni Paolo II, 132, 84084, Fisciano, SA, Italy
| | - Antonio Varriale
- CNR-ISA, National Research Council, Institute of Food Science, Via Roma 64, 83100, Avellino, Italy
| | - Angelo Facchiano
- CNR-ISA, National Research Council, Institute of Food Science, Via Roma 64, 83100, Avellino, Italy
| | - Sabato D'Auria
- CNR-ISA, National Research Council, Institute of Food Science, Via Roma 64, 83100, Avellino, Italy
| | - Maria Elisabetta Raggi
- Scientific Institute, IRCCS "Eugenio Medea" Bosisio Parini, Via Don Luigi Monza 20, 23842, Bosisio Parini, LC, Italy
| | - Anna Marabotti
- Scientific Institute, IRCCS "Eugenio Medea" Bosisio Parini, Via Don Luigi Monza 20, 23842, Bosisio Parini, LC, Italy.
- Department of Chemistry and Biology, "A. Zambelli", University of Salerno, Via Giovanni Paolo II, 132, 84084, Fisciano, SA, Italy.
| |
Collapse
|
46
|
Wu J, Tao N, Tian Y, Xing G, Lv H, Han J, Lin C, Xie W. Proteolytic maturation of Drosophila Neuroligin 3 by tumor necrosis factor α-converting enzyme in the nervous system. Biochim Biophys Acta Gen Subj 2017; 1862:440-450. [PMID: 29107812 DOI: 10.1016/j.bbagen.2017.10.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 10/18/2017] [Accepted: 10/27/2017] [Indexed: 01/23/2023]
Abstract
BACKGROUND The functions of autism-associated Neuroligins (Nlgs) are modulated by their post-translational modifications, such as proteolytic cleavage. A previous study has shown that there are different endogenous forms of DNlg3 in Drosophila, indicating it may undergo proteolytic processing. However, the molecular mechanism underlying DNlg3 proteolytic processing is unknown. Here, we report a novel proteolytic mechanism that is essential for DNlg3 maturation and function in the nervous system. METHODS Molecular cloning, cell culture, immunohistochemistry, western blotting and genetic studies were employed to map the DNlg3 cleavage region, identify the protease and characterize the cleavage manner. Behavior analysis, immunohistochemistry and genetic manipulations were employed to study the functions of different DNlg3 forms in the nervous system and neuromuscular junction (NMJs). RESULTS Tumor necrosis factor α-converting enzyme (TACE) cleaved DNlg3 exclusively at its extracellular acetylcholinesterase-like domain to generate the N-terminal fragment and the short membrane-anchored fragment (sDNlg3). DNlg3 was constitutively processed in an activity-independent manner. Interestingly, DNlg3 was cleaved intracellularly in the Golgi apparatus before it arrived at the cell surface, a unique cleavage mechanism that is distinct from 'conventional' ectodomain shedding of membrane proteins, including rodent Nlg1. Genetic studies showed that sDNlg3 was essential for maintaining proper locomotor activity in Drosophila. CONCLUSIONS Our results revealed a unique cleavage mechanism of DNlg3 and a neuron-specific role for DNlg3 maturation which is important in locomotor activity. GENERAL SIGNIFICANCE Our study provides a new insight into a cleavage mechanism of Nlgs maturation in the nervous system.
Collapse
Affiliation(s)
- Jun Wu
- Institute of Life Sciences, The Collaborative Innovation Center for Brain Science, Southeast University, China
| | - Nana Tao
- Institute of Life Sciences, The Collaborative Innovation Center for Brain Science, Southeast University, China
| | - Yao Tian
- The Key Laboratory of Developmental Genes and Human Disease, Jiangsu Co-innovation Center of Neuroregeneration, Southeast University, Nanjing 210096, China
| | - Guanglin Xing
- The Key Laboratory of Developmental Genes and Human Disease, Jiangsu Co-innovation Center of Neuroregeneration, Southeast University, Nanjing 210096, China
| | - Huihui Lv
- The Key Laboratory of Developmental Genes and Human Disease, Jiangsu Co-innovation Center of Neuroregeneration, Southeast University, Nanjing 210096, China
| | - Junhai Han
- Institute of Life Sciences, The Collaborative Innovation Center for Brain Science, Southeast University, China; The Key Laboratory of Developmental Genes and Human Disease, Jiangsu Co-innovation Center of Neuroregeneration, Southeast University, Nanjing 210096, China
| | - Chengqi Lin
- The Key Laboratory of Developmental Genes and Human Disease, Jiangsu Co-innovation Center of Neuroregeneration, Southeast University, Nanjing 210096, China
| | - Wei Xie
- Institute of Life Sciences, The Collaborative Innovation Center for Brain Science, Southeast University, China; The Key Laboratory of Developmental Genes and Human Disease, Jiangsu Co-innovation Center of Neuroregeneration, Southeast University, Nanjing 210096, China.
| |
Collapse
|
47
|
Yamasaki T, Hoyos-Ramirez E, Martenson JS, Morimoto-Tomita M, Tomita S. GARLH Family Proteins Stabilize GABA A Receptors at Synapses. Neuron 2017; 93:1138-1152.e6. [PMID: 28279354 DOI: 10.1016/j.neuron.2017.02.023] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Revised: 05/26/2016] [Accepted: 02/09/2017] [Indexed: 10/20/2022]
Abstract
Ionotropic neurotransmitter receptors mediate fast synaptic transmission by functioning as ligand-gated ion channels. Fast inhibitory transmission in the brain is mediated mostly by ionotropic GABAA receptors (GABAARs), but their essential components for synaptic localization remain unknown. Here, we identify putative auxiliary subunits of GABAARs, which we term GARLHs, consisting of LH4 and LH3 proteins. LH4 forms a stable tripartite complex with GABAARs and neuroligin-2 in the brain. Moreover, LH4 is required for the synaptic localization of GABAARs and inhibitory synaptic transmission in the hippocampus. Our findings propose GARLHs as the first identified auxiliary subunits for anion channels. These findings provide new insights into the regulation of inhibitory transmission and the molecular constituents of native anion channels in vivo.
Collapse
Affiliation(s)
- Tokiwa Yamasaki
- Department of Cellular and Molecular Physiology, Program in Cellular Neuroscience, Neurodegeneration and Repair, Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Erika Hoyos-Ramirez
- Department of Cellular and Molecular Physiology, Program in Cellular Neuroscience, Neurodegeneration and Repair, Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06520, USA
| | - James S Martenson
- Department of Cellular and Molecular Physiology, Program in Cellular Neuroscience, Neurodegeneration and Repair, Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Megumi Morimoto-Tomita
- Department of Cellular and Molecular Physiology, Program in Cellular Neuroscience, Neurodegeneration and Repair, Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Susumu Tomita
- Department of Cellular and Molecular Physiology, Program in Cellular Neuroscience, Neurodegeneration and Repair, Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
48
|
Elegheert J, Cvetkovska V, Clayton AJ, Heroven C, Vennekens KM, Smukowski SN, Regan MC, Jia W, Smith AC, Furukawa H, Savas JN, de Wit J, Begbie J, Craig AM, Aricescu AR. Structural Mechanism for Modulation of Synaptic Neuroligin-Neurexin Signaling by MDGA Proteins. Neuron 2017; 95:896-913.e10. [PMID: 28817804 PMCID: PMC5563082 DOI: 10.1016/j.neuron.2017.07.040] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Revised: 06/22/2017] [Accepted: 07/28/2017] [Indexed: 01/30/2023]
Abstract
Neuroligin-neurexin (NL-NRX) complexes are fundamental synaptic organizers in the central nervous system. An accurate spatial and temporal control of NL-NRX signaling is crucial to balance excitatory and inhibitory neurotransmission, and perturbations are linked with neurodevelopmental and psychiatric disorders. MDGA proteins bind NLs and control their function and interaction with NRXs via unknown mechanisms. Here, we report crystal structures of MDGA1, the NL1-MDGA1 complex, and a spliced NL1 isoform. Two large, multi-domain MDGA molecules fold into rigid triangular structures, cradling a dimeric NL to prevent NRX binding. Structural analyses guided the discovery of a broad, splicing-modulated interaction network between MDGA and NL family members and helped rationalize the impact of autism-linked mutations. We demonstrate that expression levels largely determine whether MDGAs act selectively or suppress the synapse organizing function of multiple NLs. These results illustrate a potentially brain-wide regulatory mechanism for NL-NRX signaling modulation.
Collapse
Affiliation(s)
- Jonathan Elegheert
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK.
| | - Vedrana Cvetkovska
- Djavad Mowafaghian Centre for Brain Health and Department of Psychiatry, University of British Columbia, Vancouver, BC V6T 2B5, Canada
| | - Amber J Clayton
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Christina Heroven
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK; MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge CB2 0QH, UK
| | - Kristel M Vennekens
- VIB Center for Brain and Disease Research, Herestraat 49, B-3000 Leuven, Belgium; Department of Neurosciences, KU Leuven, Herestraat 49, B-3000 Leuven, Belgium
| | - Samuel N Smukowski
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Michael C Regan
- Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, USA
| | - Wanyi Jia
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Alexandra C Smith
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
| | - Hiro Furukawa
- Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, USA
| | - Jeffrey N Savas
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Joris de Wit
- VIB Center for Brain and Disease Research, Herestraat 49, B-3000 Leuven, Belgium; Department of Neurosciences, KU Leuven, Herestraat 49, B-3000 Leuven, Belgium
| | - Jo Begbie
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
| | - Ann Marie Craig
- Djavad Mowafaghian Centre for Brain Health and Department of Psychiatry, University of British Columbia, Vancouver, BC V6T 2B5, Canada.
| | - A Radu Aricescu
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK; MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge CB2 0QH, UK.
| |
Collapse
|
49
|
Kim JA, Kim D, Won SY, Han KA, Park D, Cho E, Yun N, An HJ, Um JW, Kim E, Lee JO, Ko J, Kim HM. Structural Insights into Modulation of Neurexin-Neuroligin Trans-synaptic Adhesion by MDGA1/Neuroligin-2 Complex. Neuron 2017. [PMID: 28641111 DOI: 10.1016/j.neuron.2017.05.034] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Membrane-associated mucin domain-containing glycosylphosphatidylinositol anchor proteins (MDGAs) bind directly to neuroligin-1 (NL1) and neuroligin-2 (NL2), thereby respectively regulating excitatory and inhibitory synapse development. However, the mechanisms by which MDGAs modulate NL activity to specify development of the two synapse types remain unclear. Here, we determined the crystal structures of human NL2/MDGA1 Ig1-3 complex, revealing their stable 2:2 arrangement with three interaction interfaces. Cell-based assays using structure-guided, site-directed MDGA1 mutants showed that all three contact patches were required for the MDGA's negative regulation of NL2-mediated synaptogenic activity. Furthermore, MDGA1 competed with neurexins for NL2 via its Ig1 domain. The binding affinities of both MDGA1 and MDGA2 for NL1 and NL2 were similar, consistent with the structural prediction of similar binding interfaces. However, MDGA1 selectively associated with NL2, but not NL1, in vivo. These findings collectively provide structural insights into the mechanism by which MDGAs negatively modulate synapse development governed by NLs/neurexins.
Collapse
Affiliation(s)
- Jung A Kim
- Graduate School of Nanoscience and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Doyoun Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Daejeon 34141, Korea
| | | | - Kyung Ah Han
- Department of Brain & Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| | - Dongseok Park
- Department of Brain & Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| | - Eunju Cho
- Department of Brain & Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| | - Nayoung Yun
- Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon 305-764, Korea
| | - Hyun Joo An
- Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon 305-764, Korea
| | - Ji Won Um
- Department of Brain & Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| | - Eunjoon Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Daejeon 34141, Korea; Department of Biological Sciences, KAIST, Daejeon 34141, Korea
| | - Jie-Oh Lee
- Department of Chemistry, KAIST, Daejeon 34141, Korea
| | - Jaewon Ko
- Department of Brain & Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea.
| | - Ho Min Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Daejeon 34141, Korea; Graduate School of Medical Science & Engineering, KAIST, Daejeon 34141, Korea.
| |
Collapse
|
50
|
Gangwar SP, Zhong X, Seshadrinathan S, Chen H, Machius M, Rudenko G. Molecular Mechanism of MDGA1: Regulation of Neuroligin 2:Neurexin Trans-synaptic Bridges. Neuron 2017. [PMID: 28641112 DOI: 10.1016/j.neuron.2017.06.009] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Neuroligins and neurexins promote synapse development and validation by forming trans-synaptic bridges spanning the synaptic cleft. Select pairs promote excitatory and inhibitory synapses, with neuroligin 2 (NLGN2) limited to inhibitory synapses and neuroligin 1 (NLGN1) dominating at excitatory synapses. The cell-surface molecules, MAM domain-containing glycosylphosphatidylinositol anchor 1 (MDGA1) and 2 (MDGA2), regulate trans-synaptic adhesion between neurexins and neuroligins, impacting NLGN2 and NLGN1, respectively. We have determined the molecular mechanism of MDGA action. MDGA1 Ig1-Ig2 is sufficient to bind NLGN2 with nanomolar affinity; its crystal structure reveals an unusual locked rod-shaped array. In the crystal structure of the complex, two MDGA1 Ig1-Ig2 molecules each span the entire NLGN2 dimer. Site-directed mutagenesis confirms the observed interaction interface. Strikingly, Ig1 from MDGA1 binds to the same region on NLGN2 as neurexins do. Thus, MDGAs regulate the formation of neuroligin-neurexin trans-synaptic bridges by sterically blocking access of neurexins to neuroligins.
Collapse
Affiliation(s)
- Shanti Pal Gangwar
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA; Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Xiaoying Zhong
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA; Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Suchithra Seshadrinathan
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA; Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Hui Chen
- University of Michigan, Ann Arbor, MI 48109, USA
| | - Mischa Machius
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA; Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Gabby Rudenko
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA; Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX 77555, USA.
| |
Collapse
|