1
|
Jacob TV, Doshi GM. New Promising Routes in Peptic Ulcers: Toll-like Receptors and Semaphorins. Endocr Metab Immune Disord Drug Targets 2024; 24:865-878. [PMID: 37605412 DOI: 10.2174/1871530323666230821102718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 07/09/2023] [Accepted: 07/12/2023] [Indexed: 08/23/2023]
Abstract
Peptic ulcers (PU) are one of the commonest yet problematic diseases found to be existing in the majority of the population. Today, drugs from a wide range of therapeutic classes are available for the management of the disease. Still, the complications of the condition are difficult to tackle and the side effect profile is quite a concern. The literature indicates that Toll-like receptors (TLRs) and Semaphorins (SEMAs) have been under study for their various pharmacological actions over the past few decades. Both these signalling pathways are found to regulate immunological and inflammatory responses. Moreover, receptors and signalling molecules from the family of TLRs and SEMAs are found to have bacterial recognition and antibacterial properties which are essential in eradicating Helicobacter pylori (H. pylori), one of the major causative agents of PU. Our understanding of SEMAs, a class of proteins involved in cell signalling, is relatively less developed compared to TLRs, another class of proteins involved in the immune response. SEMAs and TLRs play different roles in biological processes, with SEMAs primarily involved in guiding cell migration and axon guidance during development, while TLRs are responsible for recognizing pathogens and initiating an immune response. Here, in this review, we will discuss in detail the signalling cascade of TLRs and SEMAs and thereby understand its association with PU for future therapeutic targeting. The review also aims at providing an overview of the study that has been into exploring the role of these signalling pathways in the management of PU.
Collapse
Affiliation(s)
- Teresa V Jacob
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V.M. Road, Vile Parle (W), Mumbai, 400056, India
| | - Gaurav M Doshi
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V.M. Road, Vile Parle (W), Mumbai, 400056, India
| |
Collapse
|
2
|
Sema4C Is Required for Vascular and Primary Motor Neuronal Patterning in Zebrafish. Cells 2022; 11:cells11162527. [PMID: 36010604 PMCID: PMC9406964 DOI: 10.3390/cells11162527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/29/2022] [Accepted: 08/04/2022] [Indexed: 12/03/2022] Open
Abstract
Endothelial cells (ECs) and neurons share a number of common signaling pathways and molecular mediators to orchestrate directional migration and guide the pattern of the vascular network and nervous system. So far, research concerning the functional coupling between vascular and neuronal pathfinding remains insufficient. Semaphorin4C (sema4C), a member of class 4 semaphorins, is initially described in the nervous system, whose role has been demonstrated in diverse biological developments. The present study focused on the role of sema4C in the vascular and neural development process in zebrafish embryos. It confirmed that sema4C is expressed in both the nervous system and intersegmental vessels (ISVs) in zebrafish embryos by diverse expression analysis. It also showed that the knockdown of sema4C caused a serious pathfinding anomaly both in the ISVs and primary motor neurons (PMNs) of zebrafish embryos. In addition, overexpressing exogenous sema4C mRNA in sema4C morphants remarkably neutralized the defective pattern of the vascular and neural system. Collectively, this report suggests that sema4C acts as a dual guiding factor regulating vascular and neuronal development. These findings elucidate a new molecular mechanism underlying blood vessel and nerve development and might serve as groundwork for future research on functional coupling between both systems.
Collapse
|
3
|
Cordovado A, Schaettin M, Jeanne M, Panasenkava V, Denommé-Pichon AS, Keren B, Mignot C, Doco-Fenzy M, Rodan L, Ramsey K, Narayanan V, Jones JR, Prijoles EJ, Mitchell WG, Ozmore JR, Juliette K, Torti E, Normand EA, Granger L, Petersen AK, Au MG, Matheny JP, Phornphutkul C, Chambers MK, Fernández-Ramos JA, López-Laso E, Kruer MC, Bakhtiari S, Zollino M, Morleo M, Marangi G, Mei D, Pisano T, Guerrini R, Louie RJ, Childers A, Everman DB, Isidor B, Audebert-Bellanger S, Odent S, Bonneau D, Gilbert-Dussardier B, Redon R, Bézieau S, Laumonnier F, Stoeckli ET, Toutain A, Vuillaume ML. SEMA6B variants cause intellectual disability and alter dendritic spine density and axon guidance. Hum Mol Genet 2022; 31:3325-3340. [PMID: 35604360 DOI: 10.1093/hmg/ddac114] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/25/2022] [Accepted: 05/12/2022] [Indexed: 11/14/2022] Open
Abstract
Intellectual disability is a neurodevelopmental disorder frequently caused by monogenic defects. In this study, we collected 14 SEMA6B heterozygous variants in 16 unrelated patients referred for intellectual disability to different centres. Whereas until now SEMA6B variants have mainly been reported in patients with progressive myoclonic epilepsy, our study indicates that the clinical spectrum is wider, and also includes non-syndromic intellectual disability without epilepsy or myoclonus. To assess the pathogenicity of these variants, selected mutated forms of Sema6b were overexpressed in HEK293T cells and in primary neuronal cultures. shRNAs targeting Sema6b were also used in neuronal cultures to measure the impact of the decreased Sema6b expression on morphogenesis and synaptogenesis. The overexpression of some variants leads to a subcellular mislocalisation of SEMA6B protein in HEK293T cells and to a reduced spine density due to loss of mature spines in neuronal cultures. Sema6b knock-down also impairs spine density and spine maturation. In addition, we conducted in vivo rescue experiments in chicken embryos with the selected mutated forms of Sema6b expressed in commissural neurons after knock-down of endogenous SEMA6B. We observed that expression of these variants in commissural neurons fails to rescue the normal axon pathway. In conclusion, identification of SEMA6B variants in patients presenting with an overlapping phenotype with intellectual disability, and functional studies highlight the important role of SEMA6B in neuronal development, notably in spine formation and maturation, and in axon guidance. This study adds SEMA6B to the list of intellectual disability-related genes.
Collapse
Affiliation(s)
- Amélie Cordovado
- UMR 1253, iBrain, University of Tours, Inserm, 37032 Tours, France
| | - Martina Schaettin
- Department of Molecular Life Sciences and Neuroscience Center Zurich, University of Zurich, Zurich 8057, Switzerland
| | - Médéric Jeanne
- UMR 1253, iBrain, University of Tours, Inserm, 37032 Tours, France.,Genetics Department, University Hospital of Tours, 37044 Tours, France
| | | | - Anne-Sophie Denommé-Pichon
- Functional Unit in Innovative Genomic Diagnosis of Rare Diseases, FHU-TRANSLAD, Dijon-Bourgogne University Hospital, Dijon, France.,UMR1231 GAD, Inserm - Bourgogne-Franche Comté University, Dijon, France
| | - Boris Keren
- Genetics Department, Pitié-Salpêtrière Hospital, AP-HP. Sorbonne University, 75651 Paris, France
| | - Cyril Mignot
- Genetics Department, Pitié-Salpêtrière Hospital, AP-HP. Sorbonne University, 75651 Paris, France
| | - Martine Doco-Fenzy
- University Hospital Reims, AMH2, Genetics Division, SFR CAP santé EA3801, Reims, France
| | - Lance Rodan
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA.,Department of Neurology, Boston Children's Hospital, Boston, MA, USA
| | - Keri Ramsey
- Center for Rare Childhood Disorders, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Vinodh Narayanan
- Center for Rare Childhood Disorders, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Julie R Jones
- Molecular Diagnostic Laboratory, Greenwood Genetic Center, Greenwood, SC, USA
| | | | - Wendy G Mitchell
- Neurology Division, Keck School of Medicine, University of Southern California, Children's Hospital Los Angeles, USA
| | | | - Kali Juliette
- Gillette Children's Specialty Healthcare: Neurology Department, St Paul, MN 55101, USA
| | | | | | - Leslie Granger
- Genetics Division, Department of Pediatric Development and Rehabilitation, Randall Children's Hospital, Portland, OR 97227, USA
| | - Andrea K Petersen
- Genetics Division, Department of Pediatric Development and Rehabilitation, Randall Children's Hospital, Portland, OR 97227, USA
| | - Margaret G Au
- University of Kentucky: Department of Genetics and Metabolism, Lexington, KY 40536, USA
| | - Juliann P Matheny
- University of Kentucky: Department of Genetics and Metabolism, Lexington, KY 40536, USA
| | - Chanika Phornphutkul
- Division of Human Genetics, Department of Pediatrics, Warren Alpert Medical School of Brown University, Hasbro Children's Hospital, Providence, RI 02903, USA
| | - Mary-Kathryn Chambers
- Division of Genetics, Rhode Island Hospital, Hasbro Children's Hospital, Providence, RI 02903, USA
| | | | - Eduardo López-Laso
- Pediatric Neurology Unit, department of Pediatrics, University Hospital Reina Sofía, IMIBIC and CIBERER, Córdoba, Spain
| | - Michael C Kruer
- Pediatric Movement Disorders Program, Division of Pediatric Neurology, Barrow Neurological Institute, Phoenix Children's Hospital, Phoenix, AZ, USA.,Departments of Child Health, Neurology, and Cellular & Molecular Medicine, and Program in Genetics, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Somayeh Bakhtiari
- Pediatric Movement Disorders Program, Division of Pediatric Neurology, Barrow Neurological Institute, Phoenix Children's Hospital, Phoenix, AZ, USA.,Departments of Child Health, Neurology, and Cellular & Molecular Medicine, and Program in Genetics, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Marcella Zollino
- Università Cattolica Sacro Cuore, Dipartimento Scienze della Vita e Sanità Pubblica, Sezione di Medicina Genomica, Roma, Italy.,Fondazione Policlinico A. Gemelli IRCCS, U. O. C. Genetica Medica, Roma, Italy
| | - Manuela Morleo
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Naples, Italy.,Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples 80138, Italy
| | - Giuseppe Marangi
- Università Cattolica Sacro Cuore, Dipartimento Scienze della Vita e Sanità Pubblica, Sezione di Medicina Genomica, Roma, Italy.,Fondazione Policlinico A. Gemelli IRCCS, U. O. C. Genetica Medica, Roma, Italy
| | - Davide Mei
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Meyer Children's Hospital, Member of ERN Epicare, University of Florence, Florence, Italy
| | - Tiziana Pisano
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Meyer Children's Hospital, Member of ERN Epicare, University of Florence, Florence, Italy
| | - Renzo Guerrini
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Meyer Children's Hospital, Member of ERN Epicare, University of Florence, Florence, Italy
| | - Raymond J Louie
- Molecular Diagnostic Laboratory, Greenwood Genetic Center, Greenwood, SC, USA
| | - Anna Childers
- Molecular Diagnostic Laboratory, Greenwood Genetic Center, Greenwood, SC, USA
| | - David B Everman
- Molecular Diagnostic Laboratory, Greenwood Genetic Center, Greenwood, SC, USA
| | - Betrand Isidor
- Medical Genetics Service, Clinical Genetics Unit, University Hospital of Nantes, Hôtel Dieu, 44093 Nantes, France
| | | | - Sylvie Odent
- Clinical Genetics Service, University Hospital, Genetic and Development Institute of Rennes IGDR, UMR 6290 University of Rennes, ITHACA ERN, 35203 Rennes, France
| | - Dominique Bonneau
- Department of Medical Genetics, University Hospital of Angers and Mitovasc INSERM 1083, CNRS 6015, 49000 Angers, France
| | | | - Richard Redon
- INSERM, CNRS, UNIV Nantes, Thorax Institute, 44007 Nantes, France
| | - Stéphane Bézieau
- INSERM, CNRS, UNIV Nantes, Thorax Institute, 44007 Nantes, France.,University Hospital of Nantes, Medical Genetics Service 44093 Nantes, France
| | | | - Esther T Stoeckli
- Department of Molecular Life Sciences and Neuroscience Center Zurich, University of Zurich, Zurich 8057, Switzerland
| | - Annick Toutain
- UMR 1253, iBrain, University of Tours, Inserm, 37032 Tours, France.,Genetics Department, University Hospital of Tours, 37044 Tours, France
| | - Marie-Laure Vuillaume
- UMR 1253, iBrain, University of Tours, Inserm, 37032 Tours, France.,Genetics Department, University Hospital of Tours, 37044 Tours, France
| |
Collapse
|
4
|
Wang H, Parra M, Conboy JG, Hillyer CD, Mohandas N, An X. Selective effects of protein 4.1N deficiency on neuroendocrine and reproductive systems. Sci Rep 2020; 10:16947. [PMID: 33046791 PMCID: PMC7550591 DOI: 10.1038/s41598-020-73795-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 09/14/2020] [Indexed: 12/02/2022] Open
Abstract
Protein 4.1N, a member of the protein 4.1 family, is highly expressed in the brain. But its function remains to be fully defined. Using 4.1N−/− mice, we explored the function of 4.1N in vivo. We show that 4.1N−/− mice were born at a significantly reduced Mendelian ratio and exhibited high mortality between 3 to 5 weeks of age. Live 4.1N−/− mice were smaller than 4.1N+/+ mice. Notably, while there were no significant differences in organ/body weight ratio for most of the organs, the testis/body and ovary/body ratio were dramatically decreased in 4.1N−/− mice, demonstrating selective effects of 4.1N deficiency on the development of the reproductive systems. Histopathology of the reproductive organs showed atrophy of both testis and ovary. Specifically, in the testis there is a lack of spermatogenesis, lack of leydig cells and lack of mature sperm. Similarly, in the ovary there is a lack of follicular development and lack of corpora lutea formation, as well as lack of secretory changes in the endometrium. Examination of pituitary glands revealed that the secretory granules were significantly decreased in pituitary glands of 4.1N−/− compared to 4.1N+/+. Moreover, while GnRH was expressed in both neuronal cell body and axons in the hypothalamus of 4.1N+/+ mice, it was only expressed in the cell body but not the axons of 4.1N-/- mice. Our findings uncover a novel role for 4.1N in the axis of hypothalamus-pituitary gland-reproductive system.
Collapse
Affiliation(s)
- Hua Wang
- Red Cell Physiology Laboratory, New York Blood Center, New York, NY, 10065, USA.,Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, and Peking University Third Hospital, Beijing, 100191, China
| | - Marilyn Parra
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - John G Conboy
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | | | - Narla Mohandas
- Red Cell Physiology Laboratory, New York Blood Center, New York, NY, 10065, USA
| | - Xiuli An
- Laboratory of Membrane Biology, New York Blood Center, 310 East 67th St, New York, NY, 10065, USA.
| |
Collapse
|
5
|
Elimination of the four extracellular matrix molecules tenascin-C, tenascin-R, brevican and neurocan alters the ratio of excitatory and inhibitory synapses. Sci Rep 2019; 9:13939. [PMID: 31558805 PMCID: PMC6763627 DOI: 10.1038/s41598-019-50404-9] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 09/11/2019] [Indexed: 01/12/2023] Open
Abstract
The synaptic transmission in the mammalian brain is not limited to the interplay between the pre- and the postsynapse of neurons, but involves also astrocytes as well as extracellular matrix (ECM) molecules. Glycoproteins, proteoglycans and hyaluronic acid of the ECM pervade the pericellular environment and condense to special superstructures termed perineuronal nets (PNN) that surround a subpopulation of CNS neurons. The present study focuses on the analysis of PNNs in a quadruple knockout mouse deficient for the ECM molecules tenascin-C (TnC), tenascin-R (TnR), neurocan and brevican. Here, we analysed the proportion of excitatory and inhibitory synapses and performed electrophysiological recordings of the spontaneous neuronal network activity of hippocampal neurons in vitro. While we found an increase in the number of excitatory synaptic molecules in the quadruple knockout cultures, the number of inhibitory synaptic molecules was significantly reduced. This observation was complemented with an enhancement of the neuronal network activity level. The in vivo analysis of PNNs in the hippocampus of the quadruple knockout mouse revealed a reduction of PNN size and complexity in the CA2 region. In addition, a microarray analysis of the postnatal day (P) 21 hippocampus was performed unravelling an altered gene expression in the quadruple knockout hippocampus.
Collapse
|
6
|
Gurrapu S, Franzolin G, Fard D, Accardo M, Medico E, Sarotto I, Sapino A, Isella C, Tamagnone L. Reverse signaling by semaphorin 4C elicits SMAD1/5- and ID1/3-dependent invasive reprogramming in cancer cells. Sci Signal 2019; 12:12/595/eaav2041. [DOI: 10.1126/scisignal.aav2041] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Semaphorins are a family of molecular signals that guide cell migration and are implicated in the regulation of cancer cells. In particular, transmembrane semaphorins are postulated to act as both ligands (“forward” mode) and signaling receptors (“reverse” mode); however, reverse semaphorin signaling in cancer is relatively less understood. Here, we identified a previously unknown function of transmembrane semaphorin 4C (Sema4C), acting in reverse mode, to elicit nonconventional TGF-β/BMP receptor activation and selective SMAD1/5 phosphorylation. Sema4C coimmunoprecipitated with TGFBRII and BMPR1, supporting its role as modifier of this pathway. Sema4C reverse signaling led to the increased abundance of ID1/3 transcriptional factors and to extensive reprogramming of gene expression, which suppressed the typical features of the epithelial-mesenchymal transition in invasive carcinoma cells. This phenotype was nevertheless coupled with burgeoning metastatic behavior in vivo, consistent with evidence that Sema4C expression correlates with metastatic progression in human breast cancers. Thus, Sema4C reverse signaling promoted SMAD1/5- and ID1/3-dependent gene expression reprogramming and phenotypic plasticity in invasive cancer cells.
Collapse
|
7
|
Zhou H, Kann MG, Mallory EK, Yang YH, Bugshan A, Binmadi NO, Basile JR. Recruitment of Tiam1 to Semaphorin 4D Activates Rac and Enhances Proliferation, Invasion, and Metastasis in Oral Squamous Cell Carcinoma. Neoplasia 2016; 19:65-74. [PMID: 28038319 PMCID: PMC5198113 DOI: 10.1016/j.neo.2016.12.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 12/01/2016] [Accepted: 12/05/2016] [Indexed: 12/25/2022] Open
Abstract
The semaphorins and the plexins are a family of large, cysteine-rich proteins originally identified as regulators of axon growth and lymphocyte activation that are now known to provide motility and positional information for a number of cell and tissue types. For example, our group and others have shown that some malignancies over express Semaphorin 4D (S4D), which acts through its receptor Plexin-B1 (PB1) on endothelial cells to attract blood vessels from the surrounding stroma for the purpose of supporting tumor growth. While plexins are the known functional receptors for the semaphorins, there is evidence that transmembrane semaphorins may transmit a signal themselves through their short cytoplasmic tail, a phenomenon known as ‘reverse signaling.’ We used computational methods based upon correlated evolution of sequences of interacting proteins, mutational analysis and in vitro and in vivo measurements of tumor aggressiveness to show that when bound to PB1, transmembrane S4D associates with the Rac GTPase exchange factor T lymphoma invasion and metastasis (Tiam) 1, which activates Rac and promotes proliferation, invasion and metastasis in oral squamous cell carcinoma (OSCC) cells. These results suggest that not only can S4D production by tumor cells affect the microenvironment, but engagement of this semaphorin at the cell surface activates a reverse signaling mechanism that influences tumor aggressiveness in OSCC.
Collapse
Affiliation(s)
- Hua Zhou
- Department of Oncology and Diagnostic Sciences, University of Maryland Dental School, 650 W. Baltimore Street, 7-North, Baltimore, MD 21201, USA
| | - Maricel G Kann
- Dept of Biological Sciences, University of Maryland Baltimore County, 1000 Hilltop Circle, Baltimore, MD 21250, USA
| | - Emily K Mallory
- Biomedical Informatics Training Program, Stanford University, 1265 Welch Road, Stanford, CA 94305, USA
| | - Ying-Hua Yang
- Department of Oncology and Diagnostic Sciences, University of Maryland Dental School, 650 W. Baltimore Street, 7-North, Baltimore, MD 21201, USA
| | - Amr Bugshan
- Department of Oncology and Diagnostic Sciences, University of Maryland Dental School, 650 W. Baltimore Street, 7-North, Baltimore, MD 21201, USA
| | - Nada O Binmadi
- Department of Oncology and Diagnostic Sciences, University of Maryland Dental School, 650 W. Baltimore Street, 7-North, Baltimore, MD 21201, USA; Department of Oral Basic & Clinical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - John R Basile
- Department of Oncology and Diagnostic Sciences, University of Maryland Dental School, 650 W. Baltimore Street, 7-North, Baltimore, MD 21201, USA; Greenebaum Cancer Center, 22 S. Greene Street, Baltimore, MD 21201, USA.
| |
Collapse
|
8
|
Sun T, Yang L, Kaur H, Pestel J, Looso M, Nolte H, Krasel C, Heil D, Krishnan RK, Santoni MJ, Borg JP, Bünemann M, Offermanns S, Swiercz JM, Worzfeld T. A reverse signaling pathway downstream of Sema4A controls cell migration via Scrib. J Cell Biol 2016; 216:199-215. [PMID: 28007914 PMCID: PMC5223600 DOI: 10.1083/jcb.201602002] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 09/30/2016] [Accepted: 11/13/2016] [Indexed: 11/22/2022] Open
Abstract
Semaphorins comprise a large family of ligands that regulate key cellular functions through their receptors, plexins. In this study, we show that the transmembrane semaphorin 4A (Sema4A) can also function as a receptor, rather than a ligand, and transduce signals triggered by the binding of Plexin-B1 through reverse signaling. Functionally, reverse Sema4A signaling regulates the migration of various cancer cells as well as dendritic cells. By combining mass spectrometry analysis with small interfering RNA screening, we identify the polarity protein Scrib as a downstream effector of Sema4A. We further show that binding of Plexin-B1 to Sema4A promotes the interaction of Sema4A with Scrib, thereby removing Scrib from its complex with the Rac/Cdc42 exchange factor βPIX and decreasing the activity of the small guanosine triphosphatase Rac1 and Cdc42. Our data unravel a role for Plexin-B1 as a ligand and Sema4A as a receptor and characterize a reverse signaling pathway downstream of Sema4A, which controls cell migration.
Collapse
Affiliation(s)
- Tianliang Sun
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Lida Yang
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Harmandeep Kaur
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Jenny Pestel
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Mario Looso
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Hendrik Nolte
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Cornelius Krasel
- Institute of Pharmacology and Clinical Pharmacy, Biochemical-Pharmacological Center, University of Marburg, 35043 Marburg, Germany
| | - Daniel Heil
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Ramesh K Krishnan
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Marie-Josée Santoni
- Cell Polarity, Cell Signaling and Cancer, Equipe labellisée Ligue Contre le Cancer, Institut National de la Santé et de la Recherche Médicale, U1068, 13009 Marseille, France.,Institut Paoli-Calmettes, 13009 Marseille, France.,Aix-Marseille Université, 13284 Marseille, France.,Centre National de la Recherche Scientifique, UMR7258, 13273 Marseille, France
| | - Jean-Paul Borg
- Cell Polarity, Cell Signaling and Cancer, Equipe labellisée Ligue Contre le Cancer, Institut National de la Santé et de la Recherche Médicale, U1068, 13009 Marseille, France.,Institut Paoli-Calmettes, 13009 Marseille, France.,Aix-Marseille Université, 13284 Marseille, France.,Centre National de la Recherche Scientifique, UMR7258, 13273 Marseille, France
| | - Moritz Bünemann
- Institute of Pharmacology and Clinical Pharmacy, Biochemical-Pharmacological Center, University of Marburg, 35043 Marburg, Germany
| | - Stefan Offermanns
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany.,Medical Faculty, University of Frankfurt, 60590 Frankfurt am Main, Germany
| | - Jakub M Swiercz
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Thomas Worzfeld
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany .,Institute of Pharmacology, Biochemical-Pharmacological Center, University of Marburg, 35043 Marburg, Germany
| |
Collapse
|
9
|
Xue D, Desjardins M, Kaufman GN, Béland M, Al-Tamemi S, Ahmed E, Tao S, Friedel RH, Mourad W, Mazer BD. Semaphorin 4C: A Novel Component of B-Cell Polarization in Th2-Driven Immune Responses. Front Immunol 2016; 7:558. [PMID: 28003812 PMCID: PMC5141245 DOI: 10.3389/fimmu.2016.00558] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 11/21/2016] [Indexed: 12/27/2022] Open
Abstract
Background Semaphorins are important molecules in embryonic development and multiple semaphorins have been identified as having key roles in immune regulation. To date, there is little known about Semaphorin 4C (Sema4C) in immune biology. We report for the first time that Sema4C is inducible in human and murine B-cells and may be important for normal B-cell development. Methods Human tonsillar B-cells were studied following activation via anti-CD40 antibodies in the presence or absence of representative Th1, Th2, and regulatory cytokines. Murine B-cells from WT and Sema4C−/− mice were similarly stimulated. B-cell phenotyping in WT and Sema4C mutant mice was performed by flow cytometry and lymphoid architecture was studied by immunohistochemistry. Sema4C expression and synapse formation were analyzed by confocal microscopy. Results Gene array studies performed on human tonsillar B-cells stimulated to produce IgE revealed that Sema4C was among the top genes expressed at 24 h, and the only semaphorin to be increased under Th2 conditions. Validation studies demonstrated that human and murine B-cells expressed Sema4C under similar conditions. Sema4C−/− mice had impaired maturation of B-cell follicles in spleens and associated decreases in follicular and marginal zone B-cells as well as impaired IgG and IgA production. In keeping with a potential role in maturation of B-cells, Sema4C was expressed predominantly on CD27+ human B-cells. Within 72 h of B-cell activation, Sema4C was localized to one pole in a synapse-like structure, in association with F-actin, B-cell receptor, and Plexin-B2. Cell polarization was impaired in Sema4C−/− mice. Conclusion We have identified a novel immune semaphorin induced in human and murine B-cells under Th2 conditions. Sema4C appears to be a marker for human memory B-cells. It may be important for B-cell polarization and for the formation of normal splenic follicles.
Collapse
Affiliation(s)
- Di Xue
- Translational Research in Respiratory Diseases, The Research Institute of the McGill University Health Center , Montreal, QC , Canada
| | - Marylin Desjardins
- Translational Research in Respiratory Diseases, The Research Institute of the McGill University Health Center, Montreal, QC, Canada; McGill University Health Center, Montreal Children's Hospital, Montreal, QC, Canada
| | - Gabriel N Kaufman
- Translational Research in Respiratory Diseases, The Research Institute of the McGill University Health Center , Montreal, QC , Canada
| | - Marianne Béland
- Translational Research in Respiratory Diseases, The Research Institute of the McGill University Health Center , Montreal, QC , Canada
| | - Salem Al-Tamemi
- McGill University Health Center, Montreal Children's Hospital, Montreal, QC, Canada; Sultan Qaboos University Hospital, Muscat, Oman
| | - Eisha Ahmed
- Translational Research in Respiratory Diseases, The Research Institute of the McGill University Health Center , Montreal, QC , Canada
| | - Shao Tao
- Translational Research in Respiratory Diseases, The Research Institute of the McGill University Health Center , Montreal, QC , Canada
| | | | - Walid Mourad
- Department of Medicine, University de Montreal , Montreal, QC , Canada
| | - Bruce D Mazer
- Translational Research in Respiratory Diseases, The Research Institute of the McGill University Health Center, Montreal, QC, Canada; McGill University Health Center, Montreal Children's Hospital, Montreal, QC, Canada
| |
Collapse
|
10
|
Gurrapu S, Tamagnone L. Transmembrane semaphorins: Multimodal signaling cues in development and cancer. Cell Adh Migr 2016; 10:675-691. [PMID: 27295627 DOI: 10.1080/19336918.2016.1197479] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Semaphorins constitute a large family of membrane-bound and secreted proteins that provide guidance cues for axon pathfinding and cell migration. Although initially discovered as repelling cues for axons in nervous system, they have been found to regulate cell adhesion and motility, angiogenesis, immune function and tumor progression. Notably, semaphorins are bifunctional cues and for instance can mediate both repulsive and attractive functions in different contexts. While many studies focused so far on the function of secreted family members, class 1 semaphorins in invertebrates and class 4, 5 and 6 in vertebrate species comprise around 14 transmembrane semaphorin molecules with emerging functional relevance. These can signal in juxtacrine, paracrine and autocrine fashion, hence mediating long and short range repulsive and attractive guidance cues which have a profound impact on cellular morphology and functions. Importantly, transmembrane semaphorins are capable of bidirectional signaling, acting both in "forward" mode via plexins (sometimes in association with receptor tyrosine kinases), and in "reverse" manner through their cytoplasmic domains. In this review, we will survey known molecular mechanisms underlying the functions of transmembrane semaphorins in development and cancer.
Collapse
Affiliation(s)
- Sreeharsha Gurrapu
- a Department of Oncology , University of Torino c/o IRCCS , Candiolo ( TO ), Italy.,b Candiolo Cancer Institute, IRCCS-FPO , Candiolo ( TO ), Italy
| | - Luca Tamagnone
- a Department of Oncology , University of Torino c/o IRCCS , Candiolo ( TO ), Italy.,b Candiolo Cancer Institute, IRCCS-FPO , Candiolo ( TO ), Italy
| |
Collapse
|
11
|
Puthiyedth N, Riveros C, Berretta R, Moscato P. Identification of Differentially Expressed Genes through Integrated Study of Alzheimer's Disease Affected Brain Regions. PLoS One 2016; 11:e0152342. [PMID: 27050411 PMCID: PMC4822961 DOI: 10.1371/journal.pone.0152342] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 03/11/2016] [Indexed: 11/28/2022] Open
Abstract
Background Alzheimer’s disease (AD) is the most common form of dementia in older adults that damages the brain and results in impaired memory, thinking and behaviour. The identification of differentially expressed genes and related pathways among affected brain regions can provide more information on the mechanisms of AD. In the past decade, several studies have reported many genes that are associated with AD. This wealth of information has become difficult to follow and interpret as most of the results are conflicting. In that case, it is worth doing an integrated study of multiple datasets that helps to increase the total number of samples and the statistical power in detecting biomarkers. In this study, we present an integrated analysis of five different brain region datasets and introduce new genes that warrant further investigation. Methods The aim of our study is to apply a novel combinatorial optimisation based meta-analysis approach to identify differentially expressed genes that are associated to AD across brain regions. In this study, microarray gene expression data from 161 samples (74 non-demented controls, 87 AD) from the Entorhinal Cortex (EC), Hippocampus (HIP), Middle temporal gyrus (MTG), Posterior cingulate cortex (PC), Superior frontal gyrus (SFG) and visual cortex (VCX) brain regions were integrated and analysed using our method. The results are then compared to two popular meta-analysis methods, RankProd and GeneMeta, and to what can be obtained by analysing the individual datasets. Results We find genes related with AD that are consistent with existing studies, and new candidate genes not previously related with AD. Our study confirms the up-regualtion of INFAR2 and PTMA along with the down regulation of GPHN, RAB2A, PSMD14 and FGF. Novel genes PSMB2, WNK1, RPL15, SEMA4C, RWDD2A and LARGE are found to be differentially expressed across all brain regions. Further investigation on these genes may provide new insights into the development of AD. In addition, we identified the presence of 23 non-coding features, including four miRNA precursors (miR-7, miR570, miR-1229 and miR-6821), dysregulated across the brain regions. Furthermore, we compared our results with two popular meta-analysis methods RankProd and GeneMeta to validate our findings and performed a sensitivity analysis by removing one dataset at a time to assess the robustness of our results. These new findings may provide new insights into the disease mechanisms and thus make a significant contribution in the near future towards understanding, prevention and cure of AD.
Collapse
Affiliation(s)
- Nisha Puthiyedth
- Information Based Medicine Program, Hunter Medical Research Institute, New Lambton Heights NSW, Australia
- Centre for Bioinformatics, Biomarker Discovery and Information-Based Medicine, School of Electrical Engineering and Computer Science, The University of Newcastle, Callaghan NSW, Australia
| | - Carlos Riveros
- Clinical Research Design, Information Technology and Statistics Suport Unit, Hunter Medical Research Institute, New Lambton Heights NSW, Australia
| | - Regina Berretta
- Information Based Medicine Program, Hunter Medical Research Institute, New Lambton Heights NSW, Australia
- Centre for Bioinformatics, Biomarker Discovery and Information-Based Medicine, School of Electrical Engineering and Computer Science, The University of Newcastle, Callaghan NSW, Australia
| | - Pablo Moscato
- Information Based Medicine Program, Hunter Medical Research Institute, New Lambton Heights NSW, Australia
- Centre for Bioinformatics, Biomarker Discovery and Information-Based Medicine, School of Electrical Engineering and Computer Science, The University of Newcastle, Callaghan NSW, Australia
- * E-mail:
| |
Collapse
|
12
|
Battistini C, Tamagnone L. Transmembrane semaphorins, forward and reverse signaling: have a look both ways. Cell Mol Life Sci 2016; 73:1609-22. [PMID: 26794845 PMCID: PMC11108563 DOI: 10.1007/s00018-016-2137-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 01/07/2016] [Accepted: 01/11/2016] [Indexed: 01/06/2023]
Abstract
Semaphorins are signaling molecules playing pivotal roles not only as axon guidance cues, but are also involved in the regulation of a range of biological processes, such as immune response, angiogenesis and invasive tumor growth. The main functional receptors for semaphorins are plexins, which are large single-pass transmembrane molecules. Semaphorin signaling through plexins-the "classical" forward signaling-affects cytoskeletal remodeling and integrin-dependent adhesion, consequently influencing cell migration. Intriguingly, semaphorins and plexins can interact not only in trans, but also in cis, leading to differentiated and highly regulated signaling outputs. Moreover, transmembrane semaphorins can also mediate a so-called "reverse" signaling, by acting not as ligands but rather as receptors, and initiate a signaling cascade through their own cytoplasmic domains. Semaphorin reverse signaling has been clearly demonstrated in fruit fly Sema1a, which is required to control motor axon defasciculation and target recognition during neuromuscular development. Sema1a invertebrate semaphorin is most similar to vertebrate class-6 semaphorins, and examples of semaphorin reverse signaling in mammalians have been described for these family members. Reverse signaling is also reported for other vertebrate semaphorin subsets, e.g. class-4 semaphorins, which bear potential PDZ-domain interaction motifs in their cytoplasmic regions. Therefore, thanks to their various signaling abilities, transmembrane semaphorins can play multifaceted roles both in developmental processes and in physiological as well as pathological conditions in the adult.
Collapse
Affiliation(s)
- Chiara Battistini
- Department of Oncology, University of Torino c/o IRCCS, Str. Prov. 142, 10060, Candiolo (TO), Italy
- Candiolo Cancer Institute, IRCCS-FPO, Str. Prov. 142, 10060, Candiolo (TO), Italy
| | - Luca Tamagnone
- Department of Oncology, University of Torino c/o IRCCS, Str. Prov. 142, 10060, Candiolo (TO), Italy.
- Candiolo Cancer Institute, IRCCS-FPO, Str. Prov. 142, 10060, Candiolo (TO), Italy.
| |
Collapse
|
13
|
Synaptic P-Rex1 signaling regulates hippocampal long-term depression and autism-like social behavior. Proc Natl Acad Sci U S A 2015; 112:E6964-72. [PMID: 26621702 DOI: 10.1073/pnas.1512913112] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Autism spectrum disorders (ASDs) are a group of highly inheritable mental disorders associated with synaptic dysfunction, but the underlying cellular and molecular mechanisms remain to be clarified. Here we report that autism in Chinese Han population is associated with genetic variations and copy number deletion of P-Rex1 (phosphatidylinositol-3,4,5-trisphosphate-dependent Rac exchange factor 1). Genetic deletion or knockdown of P-Rex1 in the CA1 region of the hippocampus in mice resulted in autism-like social behavior that was specifically linked to the defect of long-term depression (LTD) in the CA1 region through alteration of AMPA receptor endocytosis mediated by the postsynaptic PP1α (protein phosphase 1α)-P-Rex1-Rac1 (Ras-related C3 botulinum toxin substrate 1) signaling pathway. Rescue of the LTD in the CA1 region markedly alleviated autism-like social behavior. Together, our findings suggest a vital role of P-Rex1 signaling in CA1 LTD that is critical for social behavior and cognitive function and offer new insight into the etiology of ASDs.
Collapse
|
14
|
Plexin-B3 suppresses excitatory and promotes inhibitory synapse formation in rat hippocampal neurons. Exp Cell Res 2015; 335:269-78. [PMID: 25989221 DOI: 10.1016/j.yexcr.2015.05.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Revised: 04/23/2015] [Accepted: 05/10/2015] [Indexed: 11/21/2022]
Abstract
Molecular mechanisms underlying synaptogenesis and synaptic plasticity have become one of the main topics in neurobiology. Increasing evidence suggests that axon guidance molecules including semaphorins and plexins participate in synapse formation and elimination. Although class B plexins are widely expressed in the brain, their role in the nervous system remains poorly characterized. We previously identified that B-plexins modulate microtubule dynamics and through this impact dendrite growth in rat hippocampal neurons. Here, we demonstrate that Plexin-B2 and Plexin-B3 are present in dendrites, but do not localize in synapses. We find that overexpression of all B-plexins leads to decreased volume of excitatory synapses, and at the same time Plexin-B1 and Plexin-B3 promote inhibitory synapse assembly. Plexin-B3 mutants revealed that these processes use different downstream pathways. While elimination of excitatory synapses is the result of Plexin-B3 binding to microtubule end binding proteins EB1 and EB3, the increase in inhibitory synapses is mediated by regulation of Ras and Rho GTPases. Overall, our findings demonstrate that Plexin-B3 contributes to regulating synapse formation.
Collapse
|
15
|
Giacobini P. Shaping the Reproductive System: Role of Semaphorins in Gonadotropin-Releasing Hormone Development and Function. Neuroendocrinology 2015; 102:200-15. [PMID: 25967979 DOI: 10.1159/000431021] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 04/28/2015] [Indexed: 11/19/2022]
Abstract
The semaphorin proteins, which contribute to the morphogenesis and homeostasis of a wide range of systems, are among the best-studied families of guidance cues. Much recent research has focused on the role of semaphorins in the development and adult activity of hormone systems and, reciprocally, how circulating reproductive hormones regulate their expression and function. Specifically, several reports have focused on the molecular mechanisms underlying the effects of semaphorins on the migration, survival and structural and functional plasticity of neurons that secrete gonadotropin-releasing hormone (GnRH), essential for the acquisition and maintenance of reproductive competence in mammals. Alterations in the development of this neuroendocrine system lead to anomalous or absent GnRH secretion, resulting in heterogeneous reproductive disorders such as congenital hypogonadotropic hypogonadism (CHH) or other conditions characterized by infertility or subfertility. This review summarizes current knowledge of the role of semaphorins and their receptors on the development, differentiation and plasticity of the GnRH system. In addition, the involvement of genetic deficits in semaphorin signaling in some forms of CHH in humans is discussed.
Collapse
Affiliation(s)
- Paolo Giacobini
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Centre, U1172, School of Medicine, University of Lille, and Institut de Médecine Prédictive et de Recherche Thérapeutique, IFR114, Lille, France
| |
Collapse
|
16
|
Staudt N, Müller-Sienerth N, Fane-Dremucheva A, Yusaf SP, Millrine D, Wright GJ. A panel of recombinant monoclonal antibodies against zebrafish neural receptors and secreted proteins suitable for wholemount immunostaining. Biochem Biophys Res Commun 2014; 456:527-33. [PMID: 25490391 DOI: 10.1016/j.bbrc.2014.11.123] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 11/27/2014] [Indexed: 12/18/2022]
Abstract
Cell surface receptors and secreted proteins play important roles in neural recognition processes, but because their site of action can be a long distance from neuron cell bodies, antibodies that label these proteins are valuable to understand their function. The zebrafish embryo is a popular vertebrate model for neurobiology, but suffers from a paucity of validated antibody reagents. Here, we use the entire ectodomain of neural zebrafish cell surface or secreted proteins expressed in mammalian cells to select monoclonal antibodies to ten different antigens. The antibodies were characterised by Western blotting and the sensitivity of their epitopes to formalin fixation was determined. The rearranged antigen binding regions of the antibodies were amplified and cloned which enabled expression in a recombinant form from a single plasmid. All ten antibodies gave specific staining patterns within formalin-treated embryonic zebrafish brains, demonstrating that this generalised approach is particularly efficient to elicit antibodies that stain native antigen in fixed wholemount tissue. Finally, we show that additional tags can be easily added to the recombinant antibodies for convenient multiplex staining. The antibodies and the approaches described here will help to address the lack of well-defined antibody reagents in zebrafish research.
Collapse
Affiliation(s)
- Nicole Staudt
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1HH, UK
| | | | | | - Shahnaz P Yusaf
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1HH, UK
| | - David Millrine
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1HH, UK
| | - Gavin J Wright
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1HH, UK.
| |
Collapse
|
17
|
Semaphorins and the dynamic regulation of synapse assembly, refinement, and function. Curr Opin Neurobiol 2014; 27:1-7. [PMID: 24598309 DOI: 10.1016/j.conb.2014.02.005] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Revised: 01/21/2014] [Accepted: 02/06/2014] [Indexed: 01/13/2023]
Abstract
Semaphorins are phylogenetically conserved proteins expressed in most organ systems, including the nervous system. Following their description as axon guidance cues, semaphorins have been implicated in multiple aspects of nervous system development. Semaphorins are key regulators of neural circuit assembly, neuronal morphogenesis, assembly of excitatory and inhibitory synapses, and synaptic refinement. Semaphorins contribute to the balance between excitatory and inhibitory synaptic transmission, and electrical activity can modulate semaphorin signaling in neurons. This interplay between guidance cue signaling and electrical activity has the potential to sculpt the wiring of neural circuits and to modulate their function.
Collapse
|
18
|
Zhou QD, Ning Y, Zeng R, Chen L, Kou P, Xu CO, Pei GC, Han M, Xu G. Erbin interacts with Sema4C and inhibits Sema4C-induced epithelial-mesenchymal transition in HK2 cells. ACTA ACUST UNITED AC 2013; 33:672-679. [DOI: 10.1007/s11596-013-1179-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Revised: 08/18/2013] [Indexed: 11/29/2022]
|
19
|
Tillo M, Ruhrberg C, Mackenzie F. Emerging roles for semaphorins and VEGFs in synaptogenesis and synaptic plasticity. Cell Adh Migr 2012; 6:541-6. [PMID: 23076132 PMCID: PMC3547901 DOI: 10.4161/cam.22408] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Synapse formation, maintenance and plasticity are critical for the correct function of the nervous system and its target organs. During development, these processes enable the establishment of appropriate neural circuits. During adulthood, they allow adaptation to both physiological and environmental changes. In this review, we discuss emerging roles for two families of classical axon and vascular guidance cues in synaptogenesis and synaptic plasticity, the semaphorins and the vascular endothelial growth factors (VEGFs). Their contribution to synapse formation and function add a new facet to the spectrum of overlapping and complementary roles for these molecules in development, adulthood and disease.
Collapse
Affiliation(s)
- Miguel Tillo
- Institute of Ophthalmology, University College London, London, UK
| | | | | |
Collapse
|
20
|
Abstract
Semaphorins are key players in the control of neural circuit development. Recent studies have uncovered several exciting and novel aspects of neuronal semaphorin signalling in various cellular processes--including neuronal polarization, topographical mapping and axon sorting--that are crucial for the assembly of functional neuronal connections. This progress is important for further understanding the many neuronal and non-neuronal functions of semaphorins and for gaining insight into their emerging roles in the perturbed neural connectivity that is observed in some diseases. This Review discusses recent advances in semaphorin research, focusing on novel aspects of neuronal semaphorin receptor regulation and previously unexplored cellular functions of semaphorins in the nervous system.
Collapse
|
21
|
Sema4D as an inhibitory regulator in oligodendrocyte development. Mol Cell Neurosci 2012; 49:290-9. [DOI: 10.1016/j.mcn.2011.12.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Revised: 11/15/2011] [Accepted: 12/06/2011] [Indexed: 12/28/2022] Open
|
22
|
Plexin a-semaphorin-1a reverse signaling regulates photoreceptor axon guidance in Drosophila. J Neurosci 2010; 30:12151-6. [PMID: 20826677 DOI: 10.1523/jneurosci.1494-10.2010] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
While it is well established that Semaphorin family proteins function as axon guidance ligands in invertebrates and vertebrates, several recent studies indicate that the Drosophila Semaphorin-1a (Sema1a), a transmembrane Semaphorin, can also function as a receptor during neural development. The regulator of Sema1a reverse signaling, however, remains unknown. In this study, we show that like Sema1a, the well known Semaphorin receptor Plexin A (PlexA), is required for the proper guidance of photoreceptor (R cell) axons in the Drosophila visual system. Loss of PlexA, like loss of semala, disrupted the association of R-cell growth cones in the optic lobe. Conversely, overexpression of PlexA, like overexpression of sema1a, induced the hyperfasciculation of R-cell axons. Unlike Sema1a, however, the cytoplasmic domain of PlexA is dispensable. Epistasis analysis suggests that PlexA functions upstream of semala. And PlexA and sema1a interact genetically with Rho1. We propose that PlexA regulates Semala reverse signaling in the Drosophila visual system.
Collapse
|
23
|
Zielonka M, Xia J, Friedel RH, Offermanns S, Worzfeld T. A systematic expression analysis implicates Plexin-B2 and its ligand Sema4C in the regulation of the vascular and endocrine system. Exp Cell Res 2010; 316:2477-86. [PMID: 20478304 DOI: 10.1016/j.yexcr.2010.05.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2010] [Revised: 05/04/2010] [Accepted: 05/06/2010] [Indexed: 12/15/2022]
Abstract
Plexins serve as receptors for semaphorins and play important roles in the developing nervous system. Plexin-B2 controls decisive developmental programs in the neural tube and cerebellum. However, whether Plexin-B2 also regulates biological functions in adult nonneuronal tissues is unknown. Here we show by two methodologically independent approaches that Plexin-B2 is expressed in discrete cell types of several nonneuronal tissues in the adult mouse. In the vasculature, Plexin-B2 is selectively expressed in functionally specialized endothelial cells. In endocrine organs, Plexin-B2 localizes to the pancreatic islets of Langerhans and to both cortex and medulla of the adrenal gland. Plexin-B2 expression is also detected in certain types of immune and epithelial cells. In addition, we report on a systematic comparison of the expression patterns of Plexin-B2 and its ligand Sema4C, which show complementarity or overlap in some but not all tissues. Furthermore, we demonstrate that Plexin-B2 and its family member Plexin-B1 display largely nonredundant expression patterns. This work establishes Plexin-B2 and Sema4C as potential regulators of the vascular and endocrine system and provides an anatomical basis to understand the biological functions of this ligand-receptor pair.
Collapse
Affiliation(s)
- Matthias Zielonka
- Max-Planck-Institute for Heart and Lung Research, Department of Pharmacology, Ludwigstr. 43, 61231 Bad Nauheim, Germany
| | | | | | | | | |
Collapse
|
24
|
The PtdIns(3,4)P(2) phosphatase INPP4A is a suppressor of excitotoxic neuronal death. Nature 2010; 465:497-501. [PMID: 20463662 DOI: 10.1038/nature09023] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2009] [Accepted: 03/12/2010] [Indexed: 11/08/2022]
Abstract
Phosphorylated derivatives of phosphatidylinositol, collectively referred to as phosphoinositides, occur in the cytoplasmic leaflet of cellular membranes and regulate activities such as vesicle transport, cytoskeletal reorganization and signal transduction. Recent studies have indicated an important role for phosphoinositide metabolism in the aetiology of diseases such as cancer, diabetes, myopathy and inflammation. Although the biological functions of the phosphatases that regulate phosphatidylinositol-3,4,5-trisphosphate (PtdIns(3,4,5)P(3)) have been well characterized, little is known about the functions of the phosphatases regulating the closely related molecule phosphatidylinositol-3,4-bisphosphate (PtdIns(3,4)P(2)). Here we show that inositol polyphosphate phosphatase 4A (INPP4A), a PtdIns(3,4)P(2) phosphatase, is a suppressor of glutamate excitotoxicity in the central nervous system. Targeted disruption of the Inpp4a gene in mice leads to neurodegeneration in the striatum, the input nucleus of the basal ganglia that has a central role in motor and cognitive behaviours. Notably, Inpp4a(-/-) mice show severe involuntary movement disorders. In vitro, Inpp4a gene silencing via short hairpin RNA renders cultured primary striatal neurons vulnerable to cell death mediated by N-methyl-d-aspartate-type glutamate receptors (NMDARs). Mechanistically, INPP4A is found at the postsynaptic density and regulates synaptic NMDAR localization and NMDAR-mediated excitatory postsynaptic current. Thus, INPP4A protects neurons from excitotoxic cell death and thereby maintains the functional integrity of the brain. Our study demonstrates that PtdIns(3,4)P(2), PtdIns(3,4,5)P(3) and the phosphatases acting on them can have distinct regulatory roles, and provides insight into the unique aspects and physiological significance of PtdIns(3,4)P(2) metabolism. INPP4A represents, to our knowledge, the first signalling protein with a function in neurons to suppress excitotoxic cell death. The discovery of a direct link between PtdIns(3,4)P(2) metabolism and the regulation of neurodegeneration and involuntary movements may aid the development of new approaches for the treatment of neurodegenerative disorders.
Collapse
|
25
|
Identification of beta-secretase (BACE1) substrates using quantitative proteomics. PLoS One 2009; 4:e8477. [PMID: 20041192 PMCID: PMC2793532 DOI: 10.1371/journal.pone.0008477] [Citation(s) in RCA: 149] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2009] [Accepted: 12/01/2009] [Indexed: 11/19/2022] Open
Abstract
β-site APP cleaving enzyme 1 (BACE1) is a transmembrane aspartyl protease with a lumenal active site that sheds the ectodomains of membrane proteins through juxtamembrane proteolysis. BACE1 has been studied principally for its role in Alzheimer's disease as the β-secretase responsible for generating the amyloid-β protein. Emerging evidence from mouse models has identified the importance of BACE1 in myelination and cognitive performance. However, the substrates that BACE1 processes to regulate these functions are unknown, and to date only a few β-secretase substrates have been identified through candidate-based studies. Using an unbiased approach to substrate identification, we performed quantitative proteomic analysis of two human epithelial cell lines stably expressing BACE1 and identified 68 putative β-secretase substrates, a number of which we validated in a cell culture system. The vast majority were of type I transmembrane topology, although one was type II and three were GPI-linked proteins. Intriguingly, a preponderance of these proteins are involved in contact-dependent intercellular communication or serve as receptors and have recognized roles in the nervous system and other organs. No consistent sequence motif predicting BACE1 cleavage was identified in substrates versus non-substrates. These findings expand our understanding of the proteins and cellular processes that BACE1 may regulate, and suggest possible mechanisms of toxicity arising from chronic BACE1 inhibition.
Collapse
|
26
|
Taniguchi Y, Amazaki M, Furuyama T, Yamaguchi W, Takahara M, Saino O, Wada T, Niwa H, Tashiro F, Miyazaki JI, Kogo M, Matsuyama T, Inagaki S. Sema4D deficiency results in an increase in the number of oligodendrocytes in healthy and injured mouse brains. J Neurosci Res 2009; 87:2833-41. [DOI: 10.1002/jnr.22124] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
27
|
Li N, Mei H, MacDonald IM, Jiao X, Hejtmancik JF. Mutations in ASCC3L1 on 2q11.2 are associated with autosomal dominant retinitis pigmentosa in a Chinese family. Invest Ophthalmol Vis Sci 2009; 51:1036-43. [PMID: 19710410 DOI: 10.1167/iovs.09-3725] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE To localize and identify the gene and mutations causing autosomal dominant retinitis pigmentosa in a Chinese Family. METHODS Families were ascertained and patients underwent complete ophthalmic examinations. Blood samples were collected and DNA was extracted. A linkage scan of genomic regions containing known candidate genes was performed by using 34 polymorphic microsatellite markers on genomic DNA from affected and unaffected family members, and lod scores were calculated. Candidate genes were sequenced and mutations analyzed. RESULTS A genome-wide scan yielded a lod score of 3.5 at theta = 0 for D2S2333 and 3.46 at theta = 0 for D2S2216. This region harbors the ASCC3L1 gene. Sequencing of ASCC3L1 in an affected family member showed a heterozygous single-base-pair change; c.3269G-->T, predicted to result in an Arg1090Leu amino acid change. CONCLUSIONS The results provide strong evidence that mutations in ASCC3L1 have resulted in autosomal dominant retinitis pigmentosa in this Chinese family.
Collapse
|
28
|
Hevezi P, Moyer BD, Lu M, Gao N, White E, Echeverri F, Kalabat D, Soto H, Laita B, Li C, Yeh SA, Zoller M, Zlotnik A. Genome-wide analysis of gene expression in primate taste buds reveals links to diverse processes. PLoS One 2009; 4:e6395. [PMID: 19636377 PMCID: PMC2712080 DOI: 10.1371/journal.pone.0006395] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2009] [Accepted: 05/27/2009] [Indexed: 11/19/2022] Open
Abstract
Efforts to unravel the mechanisms underlying taste sensation (gustation) have largely focused on rodents. Here we present the first comprehensive characterization of gene expression in primate taste buds. Our findings reveal unique new insights into the biology of taste buds. We generated a taste bud gene expression database using laser capture microdissection (LCM) procured fungiform (FG) and circumvallate (CV) taste buds from primates. We also used LCM to collect the top and bottom portions of CV taste buds. Affymetrix genome wide arrays were used to analyze gene expression in all samples. Known taste receptors are preferentially expressed in the top portion of taste buds. Genes associated with the cell cycle and stem cells are preferentially expressed in the bottom portion of taste buds, suggesting that precursor cells are located there. Several chemokines including CXCL14 and CXCL8 are among the highest expressed genes in taste buds, indicating that immune system related processes are active in taste buds. Several genes expressed specifically in endocrine glands including growth hormone releasing hormone and its receptor are also strongly expressed in taste buds, suggesting a link between metabolism and taste. Cell type-specific expression of transcription factors and signaling molecules involved in cell fate, including KIT, reveals the taste bud as an active site of cell regeneration, differentiation, and development. IKBKAP, a gene mutated in familial dysautonomia, a disease that results in loss of taste buds, is expressed in taste cells that communicate with afferent nerve fibers via synaptic transmission. This database highlights the power of LCM coupled with transcriptional profiling to dissect the molecular composition of normal tissues, represents the most comprehensive molecular analysis of primate taste buds to date, and provides a foundation for further studies in diverse aspects of taste biology.
Collapse
Affiliation(s)
- Peter Hevezi
- Senomyx, Inc, San Diego, California, United States of America
| | - Bryan D. Moyer
- Senomyx, Inc, San Diego, California, United States of America
- * E-mail:
| | - Min Lu
- Senomyx, Inc, San Diego, California, United States of America
| | - Na Gao
- Senomyx, Inc, San Diego, California, United States of America
| | - Evan White
- Senomyx, Inc, San Diego, California, United States of America
| | | | - Dalia Kalabat
- Senomyx, Inc, San Diego, California, United States of America
| | - Hortensia Soto
- Senomyx, Inc, San Diego, California, United States of America
| | - Bianca Laita
- Senomyx, Inc, San Diego, California, United States of America
| | - Cherry Li
- Senomyx, Inc, San Diego, California, United States of America
| | | | - Mark Zoller
- Senomyx, Inc, San Diego, California, United States of America
| | - Albert Zlotnik
- Senomyx, Inc, San Diego, California, United States of America
| |
Collapse
|
29
|
Toguchi M, Gonzalez D, Furukawa S, Inagaki S. Involvement of Sema4D in the control of microglia activation. Neurochem Int 2009; 55:573-80. [PMID: 19467284 DOI: 10.1016/j.neuint.2009.05.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2009] [Revised: 05/10/2009] [Accepted: 05/15/2009] [Indexed: 11/29/2022]
Abstract
Microglia normally exist in a resting state characterized by a ramified morphology, and are responsible for immune surveillance in the CNS. However, the resting microglia rapidly transform towards an activated phenotype in response to brain injury or immunological stimuli. In certain pathological conditions, the unregulated response or over-activation of microglia can provoke severe neuronal damage. Here, we have investigated whether Semaphorin4D (Sema4D/CD100) could function as a potential factor to control activation. Microglia were cultured, activated by bacterial endotoxin lipopolysaccharide (LPS) and then, exposed to Sema4D/CD100 or conditioned medium. We found that Sema4D/CD100 negatively controlled LPS-induced morphological activation. Moreover, intracerebral injection of LPS-induced abundant microglial activated forms in mice lacking Sema4D/CD100. Sema4D/CD100 also inhibited other relevant aspects of cell activation. Treatment with Sema4D/CD100 inhibited the production of nitrites and LPS-induced microglia migration. We also provide evidence that LPS markedly upregulated Plexin-B1 expression in microglia and Sema4D/CD100 stimulated RhoA-activation in LPS-activated microglia. Taken together, these findings suggest a novel role of Sema4D/CD100 in the regulation of microglia activation providing a valuable neuroprotective tool to the CNS.
Collapse
|
30
|
Sema4C Expression in Neural Stem/Progenitor Cells and in Adult Neurogenesis Induced by Cerebral Ischemia. J Mol Neurosci 2009; 39:27-39. [DOI: 10.1007/s12031-009-9177-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2008] [Accepted: 01/07/2009] [Indexed: 12/28/2022]
|
31
|
Morishige N, Ko JA, Liu Y, Chikama TI, Nishida T. Localization of semaphorin 3A in the rat cornea. Exp Eye Res 2008; 86:669-74. [PMID: 18308303 DOI: 10.1016/j.exer.2008.01.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2007] [Revised: 01/08/2008] [Accepted: 01/11/2008] [Indexed: 12/11/2022]
Abstract
Semaphorin 3A (Sema3A) functions to guide the growth of neurons during development. We investigated the localization of Sema3A in the cornea, one of the most sensitive tissues in the body. Immunoblot analysis and reverse transcription-polymerase chain reaction analysis revealed that Sema3A protein and mRNA are expressed in the normal rat cornea. Immunofluorescence staining of frozen sections or tissue blocks prepared from the cornea revealed the presence of Sema3A in wing cells and basal cells (but not superficial cells) of the corneal epithelium, in keratocytes, and in the corneal endothelium. The expression pattern of Sema3A in the corneal epithelium differed from those of zonula occludens-1 and connexin43. These observations show that Sema3A is expressed in all cells of the rat cornea with the exception of superficial epithelial cells.
Collapse
Affiliation(s)
- Naoyuki Morishige
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan.
| | | | | | | | | |
Collapse
|
32
|
Gregg JP, Lit L, Baron CA, Hertz-Picciotto I, Walker W, Davis RA, Croen LA, Ozonoff S, Hansen R, Pessah IN, Sharp FR. Gene expression changes in children with autism. Genomics 2007; 91:22-9. [PMID: 18006270 DOI: 10.1016/j.ygeno.2007.09.003] [Citation(s) in RCA: 130] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2007] [Revised: 09/11/2007] [Accepted: 09/15/2007] [Indexed: 01/30/2023]
Abstract
The objective of this study was to identify gene expression differences in blood differences in children with autism (AU) and autism spectrum disorder (ASD) compared to general population controls. Transcriptional profiles were compared with age- and gender-matched, typically developing children from the general population (GP). The AU group was subdivided based on a history of developmental regression (A-R) or a history of early onset (A-E without regression). Total RNA from blood was processed on human Affymetrix microarrays. Thirty-five children with AU (17 with early onset autism and 18 with autism with regression) and 14 ASD children (who did not meet criteria for AU) were compared to 12 GP children. Unpaired t tests (corrected for multiple comparisons with a false discovery rate of 0.05) detected a number of genes that were regulated more than 1.5-fold for AU versus GP (n=55 genes), for A-E versus GP (n=140 genes), for A-R versus GP (n=20 genes), and for A-R versus A-E (n=494 genes). No genes were significantly regulated for ASD versus GP. There were 11 genes shared between the comparisons of all autism subgroups to GP (AU, A-E, and A-R versus GP) and these genes were all expressed in natural killer cells and many belonged to the KEGG natural killer cytotoxicity pathway (p=0.02). A subset of these genes (n=7) was tested with qRT-PCR and all genes were found to be differentially expressed (p<0.05). We conclude that the gene expression data support emerging evidence for abnormalities in peripheral blood leukocytes in autism that could represent a genetic and/or environmental predisposition to the disorder.
Collapse
Affiliation(s)
- Jeffrey P Gregg
- Department of Pathology, University of California at Davis Medical Center, Sacramento, CA 95817, USA. University of California at Davis, Davis, CA 95616, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Lin X, Ogiya M, Takahara M, Yamaguchi W, Furuyama T, Tanaka H, Tohyama M, Inagaki S. Sema4D-plexin-B1 implicated in regulation of dendritic spine density through RhoA/ROCK pathway. Neurosci Lett 2007; 428:1-6. [PMID: 17950529 DOI: 10.1016/j.neulet.2007.09.045] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2007] [Revised: 09/02/2007] [Accepted: 09/19/2007] [Indexed: 12/21/2022]
Abstract
Plexin-B1, Sema4D receptor, mediates retraction and extension signals in axon guidance by associating with PDZ-containing Rho guanine nucleotide exchange factors (PDZ-RhoGEFs) which can activate a small Rho GTPase RhoA. RhoA is implicated in spine formation by rearranging actin cytoskeleton. Exogenous application of Sema4D to cultured neurons caused activation of RhoA, increase of spine density and changes in spine shape. Sema4D-induced changes in spine density were blocked by either Rho-kinase (a downstream of RhoA, ROCK) inhibitor Y-27632 or by overexpression of plexin-B1 mutant lacking the C-terminus which no longer associates with PDZ-RhoGEFs. This study suggests that Sema4D-plexin-B1 play a crucial role in spine formation by regulating RhoA/ROCK pathway.
Collapse
Affiliation(s)
- Xianzong Lin
- Department of Anatomy and Neuroscience, Osaka University Graduate School of Medicine, Yamadaoka 2-2, Suita, Osaka, Japan
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Svensson A, Libelius R, Tågerud S. Semaphorin 6C expression in innervated and denervated skeletal muscle. J Mol Histol 2007; 39:5-13. [PMID: 17605078 DOI: 10.1007/s10735-007-9113-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2007] [Accepted: 06/07/2007] [Indexed: 10/23/2022]
Abstract
Semaphorins are secreted or transmembrane proteins important for axonal guidance and for the structuring of neuronal systems. Semaphorin 6C, a transmembrane Semaphorin, has growth cone collapsing activity and is expressed in adult skeletal muscle. In the present study the expression of Semaphorin 6C mRNA and immunoreactivity has been compared in innervated and denervated mouse hind-limb and hemidiaphragm muscles. Microscopic localization of immunoreactivity was studied in innervated and denervated rat skeletal muscle. The results show that Semaphorin 6C mRNA expression and immunoreactivity on Western blots are down-regulated following denervation. The mRNA of Semaphorin 6C as well as immunoreactivity determined by Western blots are expressed in extrasynaptic as well as perisynaptic regions of muscle. Immunohistochemical studies, however, show Semaphorin 6C-like immunoreactivity to be concentrated at neuromuscular junctions. The results suggest a role for Semaphorin 6C in neuromuscular communication.
Collapse
Affiliation(s)
- Anna Svensson
- School of Pure and Applied Natural Sciences, University of Kalmar, Kalmar, 391 82, Sweden.
| | | | | |
Collapse
|
35
|
Wu H, Wang X, Liu S, Wu Y, Zhao T, Chen X, Zhu L, Wu Y, Ding X, Peng X, Yuan J, Wang X, Fan W, Fan M. Sema4C participates in myogenic differentiation in vivo and in vitro through the p38 MAPK pathway. Eur J Cell Biol 2007; 86:331-44. [PMID: 17498836 DOI: 10.1016/j.ejcb.2007.03.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2006] [Revised: 03/03/2007] [Accepted: 03/05/2007] [Indexed: 11/19/2022] Open
Abstract
Sema4C is a member of transmembrane semaphorin proteins which regulate axonal guidance in the developing nervous system. The expression of Sema4C was dramatically induced not only during differentiation of C2C12 mouse myoblasts, but also during injury-induced skeletal muscle regeneration. C2C12 cells stably or transiently expressing Sema4C both showed increased myogenic differentiation reflected by accelerated myotube formation and expression of muscle-specific proteins. Overexpression of Sema4C elicited p38 phosphorylation directly, and the effects of Sema4C during myogenic differentiation could be abolished by the p38alpha-specific inhibitor SB203580. Knockdown of Sema4C by siRNA transfection during C2C12 myoblasts differentiation could suppress the phosphorylation of p38 followed by dramatically diminished myotube formation. Sema4C could activate the myogenin promoter during myogenic differentiation. This activation could be abolished by p38 inhibitor SB203580. Taken together, these observations reveal novel functional potentialities of Sema4C which suggest that Sema4C promotes terminal myogenic differentiation in a p38 MAPK-dependent manner.
Collapse
Affiliation(s)
- Haitao Wu
- Department of Brain Protection & Plasticity Research, Beijing Institute of Basic Medical Sciences, Taiping Road 27, Beijing 100850, PR China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Mann F, Chauvet S, Rougon G. Semaphorins in development and adult brain: Implication for neurological diseases. Prog Neurobiol 2007; 82:57-79. [PMID: 17537564 DOI: 10.1016/j.pneurobio.2007.02.011] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2006] [Revised: 01/18/2007] [Accepted: 02/26/2007] [Indexed: 01/17/2023]
Abstract
As a group, Semaphorins are expressed in most tissues and this distribution varies considerably with age. Semaphorins are dynamically expressed during embryonic development and their expression is often associated with growing axons. This expression decreases with maturity and several observations support the idea that in adult brain the expression of secreted Semaphorins is sensitive to electrical activity and experience. The functional role of Semaphorins in guiding axonal projections is well established and more recent evidence points to additional roles in the development, function and reorganization of synaptic complexes. Semaphorins exert the majority of their effects by binding to cognate receptor proteins through their extracellular domains. A common theme is that Semaphorin-triggered signalling induces the rearrangement of the actin and microtubule cytoskeleton. Mutations in Semaphorin genes are linked to several human diseases associated with neurological changes, but their actual influence in the pathogenesis of these diseases remains to be demonstrated. In addition, Semaphorins and their receptors are likely to mediate cross-talk between neurons and other cell types, including in pathological situations where their influence can be damaging or favourable depending on the context. We discuss how the manipulation of Semaphorin function might be crucial for future clinical studies.
Collapse
Affiliation(s)
- Fanny Mann
- CNRS UMR 6216, Université de la Méditerranée, Developmental Biology Institute of Marseille Luminy, Case 907, Parc Scientifique de Luminy, 13288 Marseille Cedex 09, France
| | | | | |
Collapse
|
37
|
Nagai H, Sugito N, Matsubara H, Tatematsu Y, Hida T, Sekido Y, Nagino M, Nimura Y, Takahashi T, Osada H. CLCP1 interacts with semaphorin 4B and regulates motility of lung cancer cells. Oncogene 2007; 26:4025-31. [PMID: 17213806 DOI: 10.1038/sj.onc.1210183] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
We previously established a highly metastatic subline, LNM35, from the NCI-H460 lung cancer cell line, and demonstrated upregulation of a novel gene, CLCP1 (CUB, LCCL-homology, coagulation factor V/VIII homology domains protein), in LNM35 and lung cancer specimens. In this study, we focused on the potential roles of that gene in cancer metastasis. First, we established stable LNM35 RNAi clones, in which CLCP1 expression was suppressed by RNAi, and found that their motility was significantly reduced, although growth rates were not changed. Next, in vitro selection of a phage display library demonstrated that a phage clone displaying a peptide similar to a sequence within the Sema domain of semaphorin 4B (SEMA4B) interacted with LNM35. Immunoprecipitation experiments confirmed interaction of CLCP1 with SEMA4B, regulation of CLCP1 protein by ubiquitination and proteasome degradation enhanced in the presence of SEMA4B. These results are the first to indicate that CLCP1 plays a role in cell motility, whereas they also showed that at least one of its ligands is SEMA4B and that their interaction mediates proteasome degradation by CLCP1. Although the physiological role of the interaction between CLCP1 and SEMA4B remains to be investigated, this novel gene may become a target of therapy to inhibit metastasis of lung cancers.
Collapse
Affiliation(s)
- H Nagai
- Division of Molecular Carcinogenesis, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Maynard KR, McCarthy SS, Sheldon E, Horch HW. Developmental and adult expression of semaphorin 2a in the cricketGryllus bimaculatus. J Comp Neurol 2007; 503:169-81. [PMID: 17480023 DOI: 10.1002/cne.21392] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Developmental guidance cues act to direct growth cones to their correct targets in the nervous system. Recent experiments also demonstrate that developmental cues are expressed in the adult mammalian nervous system, although their function in the brain is not yet clear. The semaphorin gene family has been implicated in the growth of dendrites and axons in a number of different species. While the expression of semaphorin and its influence on tibial pioneer neurons in the developing limb bud have been well characterized in the grasshopper, the expression of semaphorin 2a (sema2a) has not been explored in the adult insect. In this study we used polymerase chain reaction (PCR) with degenerate and gene-specific primers to clone part of the secreted form of sema2a from Gryllus bimaculatus. Using in situ hybridization and immunohistochemistry, we confirmed that sema2a mRNA and protein expression patterns in the embryonic cricket were similar to that seen in the grasshopper. We also showed that tibial neuron development in crickets was comparable to that described in grasshopper. An examination of both developing and adult cricket brains showed that sema2a mRNA and protein were expressed in the Kenyon cells in mushroom bodies, an area involved in learning and memory. Sema2a expression was most obvious near the apex of the mushroom body in a region surrounding the neurogenic tip, which produces neurons throughout the life of the cricket. We discuss the role of neurogenesis in learning and memory and the potential involvement of semaphorin in this process.
Collapse
|
39
|
Burgaya F, Fontana X, Martínez A, Montolio M, Mingorance A, Simó S, del Río JA, Soriano E. Semaphorin 6C leads to GSK-3-dependent growth cone collapse and redistributes after entorhino-hippocampal axotomy. Mol Cell Neurosci 2006; 33:321-34. [PMID: 17029982 DOI: 10.1016/j.mcn.2006.08.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2006] [Revised: 07/24/2006] [Accepted: 08/17/2006] [Indexed: 10/24/2022] Open
Abstract
We studied the changes in the distribution of a specific variant of Semaphorin Y/6C (Sema6C) in mouse forebrain after axotomy of the entorhino-hippocampal perforant pathway. We found this isoform to be widely expressed during development, remaining in the adult and showing variations in distribution when the perforant pathway was axotomized. These changes were detected in both the hippocampal and entorhinal cortices. Sema6C1 immunoreactivity (IR) was high in the stratum radiatum of the hippocampus proper and the inner molecular layer of the dentate gyrus; the entorhinal cortex showed Sema6C1 IR in both cell bodies and in fibers of the II/III and V/VI layers. In axotomized animals, the IR of the ipsilateral, but not the contralateral, hemisphere showed that IR had moved into the stratum lacunosum-moleculare, the medial molecular layer of the dentate gyrus and the fibers, but not the cell bodies, of the entorhinal cortex. These results were not reproduced after lateral axotomy of the fimbria fornix, indicating a specific role for Sema6C variants in the generation and/or stability of entorhino-hippocampal synapses. Growth cone collapse of entorhinal and pyramidal neurons, as well as activation of glycogen synthase kinase-3 (GSK-3) through depletion of the inactive pool, induced by diffusible Sema6C1 further supports this view.
Collapse
Affiliation(s)
- Ferran Burgaya
- Institute for Research in Biomedicine (IRB) and Cell Biology Department, Faculty of Biology, University of Barcelona, Barcelona, Spain.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Piechotta K, Dudanova I, Missler M. The resilient synapse: insights from genetic interference of synaptic cell adhesion molecules. Cell Tissue Res 2006; 326:617-42. [PMID: 16855838 DOI: 10.1007/s00441-006-0267-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2006] [Accepted: 05/31/2006] [Indexed: 01/05/2023]
Abstract
Synaptic cell adhesion molecules (SCAMs) are mostly membrane-anchored molecules with extracellular domains that extend into the synaptic cleft. Prototypical SCAMs interact with homologous or heterologous molecules on the surface of adjacent cells, ensuring the precise apposition of pre- and postsynaptic elements. More recent definitions of SCAMs often include molecules involved in axon pathfinding, cell recognition and synaptic differentiation events, making SCAMs functionally and molecularly a highly diverse group. In this review, we summarize the proposed in vivo functions of a large variety of SCAMs. We mainly focus on results obtained from analyses of genetic model organisms, mostly mouse knockout mutants, lacking expression of the respective candidate genes. In contrast to the substantial effect yielded by some knockouts of molecules involved in synaptic vesicle release, no SCAM mutant has been reported thus far that shows a prominently altered structure of the majority of synapses or even lacks synapses altogether. This surprising resilience of synaptic structure might be explained by a high redundancy between different SCAMs, by the assumption that the crucial molecular players in synapse structure have yet to be discovered or by a grand variability in the mechanisms of synapse formation that underlies the diversity of synapses. Whatever the final answer turns out to be, the genetic dissection of the SCAM superfamilies has led to a much better understanding of the different steps required to form, differentiate and modify a synapse.
Collapse
Affiliation(s)
- Kerstin Piechotta
- Center for Physiology and Pathophysiology, Georg-August University, Humboldtallee 23, 37073 Göttingen, Germany
| | | | | |
Collapse
|
41
|
Abstract
Semaphorins are secreted, transmembrane, and GPI-linked proteins, defined by cysteine-rich semaphorin protein domains, that have important roles in a variety of tissues. Humans have 20 semaphorins, Drosophila has five, and two are known from DNA viruses; semaphorins are also found in nematodes and crustaceans but not in non-animals. They are grouped into eight classes on the basis of phylogenetic tree analyses and the presence of additional protein motifs. The expression of semaphorins has been described most fully in the nervous system, but they are also present in most, or perhaps all, other tissues. Functionally, semaphorins were initially characterized for their importance in the development of the nervous system and in axonal guidance. More recently, they have been found to be important for the formation and functioning of the cardiovascular, endocrine, gastrointestinal, hepatic, immune, musculoskeletal, renal, reproductive, and respiratory systems. A common theme in the mechanisms of semaphorin function is that they alter the cytoskeleton and the organization of actin filaments and the microtubule network. These effects occur primarily through binding of semaphorins to their receptors, although transmembrane semaphorins also serve as receptors themselves. The best characterized receptors for mediating semaphorin signaling are members of the neuropilin and plexin families of transmembrane proteins. Plexins, in particular, are thought to control many of the functional effects of semaphorins; the molecular mechanisms of semaphorin signaling are still poorly understood, however. Given the importance of semaphorins in a wide range of functions, including neural connectivity, angiogenesis, immunoregulation, and cancer, much remains to be learned about these proteins and their roles in pathology and human disease.
Collapse
Affiliation(s)
- Umar Yazdani
- Center for Basic Neuroscience, Department of Pharmacology, NA4.301/5323 Harry Hines Blvd, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jonathan R Terman
- Center for Basic Neuroscience, Department of Pharmacology, NA4.301/5323 Harry Hines Blvd, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
42
|
Abstract
Semaphorins are secreted or transmembrane proteins that regulate cell motility and attachment in axon guidance, vascular growth, immune cell regulation and tumour progression. The main receptors for semaphorins are plexins, which have established roles in regulating Rho-family GTPases. Recent work shows that plexins can also influence R-Ras, which, in turn, can regulate integrins. Such regulation is probably a common feature of semaphorin signalling and contributes substantially to our understanding of semaphorin biology.
Collapse
Affiliation(s)
- Robert P Kruger
- Neuroscience Program and Institute of Gerontology, Life Sciences Institute, University of Michigan, 210 Washtenaw Avenue, Ann Arbor, Michigan 48109, USA
| | | | | |
Collapse
|
43
|
Burkhardt C, Müller M, Badde A, Garner CC, Gundelfinger ED, Püschel AW. Semaphorin 4B interacts with the post-synaptic density protein PSD-95/SAP90 and is recruited to synapses through a C-terminal PDZ-binding motif. FEBS Lett 2005; 579:3821-8. [PMID: 15978582 DOI: 10.1016/j.febslet.2005.05.079] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2005] [Revised: 05/30/2005] [Accepted: 05/31/2005] [Indexed: 11/28/2022]
Abstract
The semaphorins are a large family of proteins that act as guidance signals for axons and dendrites. The class 4 semaphorins are integral membrane proteins that are widely expressed throughout the nervous system. Here, we show that a subclass of these semaphorins is characterized by a PDZ-binding motif at their carboxy-terminus. This sequence mediates the interaction with the post-synaptic density protein PSD-95/SAP90. Co-expression of Sema4B with PSD-95 in COS 7 cells results in the clustering of Sema4B. Sema4B co-localizes with PSD-95 at synaptic contacts between cultured hippocampal neurons. This synaptic localization depends on the presence of the PDZ-binding motif.
Collapse
Affiliation(s)
- Constanze Burkhardt
- Abt. Molekularbiologie, Institut für Allgemeine Zoologie und Genetik, Westfälische Wilhelms-Universität, Schlossplatz 5, D-48149 Münster, Germany
| | | | | | | | | | | |
Collapse
|
44
|
Ko JA, Gondo T, Inagaki S, Inui M. Requirement of the transmembrane semaphorin Sema4C for myogenic differentiation. FEBS Lett 2005; 579:2236-42. [PMID: 15811348 DOI: 10.1016/j.febslet.2005.03.022] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2005] [Revised: 03/03/2005] [Accepted: 03/03/2005] [Indexed: 12/28/2022]
Abstract
Semaphorins constitute a large family of signaling proteins that contribute to axonal guidance. Here we demonstrate that the transmembrane semaphorin Sema4C is up-regulated both in the early stage of differentiation of C2C12 mouse skeletal myoblasts into myotubes and during injury-induced muscle regeneration in vivo. Depletion of Sema4C in C2C12 cells resulted in marked attenuation of myotube formation. A fusion protein containing the extracellular Sema domain and a peptide corresponding to the intracellular COOH-terminal region of Sema4C each inhibited the differentiation of C2C12 cells. These findings indicate that Sema4C-mediated interaction among myoblasts plays an important role in terminal myogenic differentiation.
Collapse
Affiliation(s)
- Ji-Ae Ko
- Department of Pharmacology, Yamaguchi University School of Medicine, Yamaguchi University Hospital, Ube, Yamaguchi 755-8505, Japan
| | | | | | | |
Collapse
|
45
|
Abstract
Class 4 semaphorins are a large class of transmembrane proteins that contain a sema domain and that are expressed in the CNS, but their in vivo neural function is unknown. In zebrafish, the epithelial cells that line the pharyngeal arches express Sema4E. Extension of branchiomotor axons along the mesenchymal cells bounded by these epithelial cells suggests that Sema4E may act as a repulsive guidance molecule to restrict the branchiomotor axons to the mesenchymal cells. To test this hypothesis, Sema4E was misexpressed in hsp70 promoter-regulated transgenic zebrafish in which sema4E was heat-inducible, and Sema4E was knocked down by injection of antisense morpholino oligonucleotides that acted specifically against Sema4E. Ubiquitous induction of Sema4E retarded outgrowth by the facial and gill branchiomotor axons significantly. Furthermore, outgrowth by gill motor axons was specifically inhibited when Sema4E-expressing transgenic cells were transplanted to their pathway in nontransgenic host embryos. Morpholino knockdown of Sema4E caused facial motor axons to defasciculate and follow aberrant pathways. These results show that Sema4E is repulsive for facial and gill motor axons and functions as a barrier for these axons within the pharyngeal arches.
Collapse
|
46
|
Holtmaat AJGD, De Winter F, De Wit J, Gorter JA, da Silva FHL, Verhaagen J. Semaphorins: contributors to structural stability of hippocampal networks? PROGRESS IN BRAIN RESEARCH 2002; 138:17-38. [PMID: 12432760 DOI: 10.1016/s0079-6123(02)38068-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Affiliation(s)
- Anthony J G D Holtmaat
- Netherlands Institute for Brain Research, Meibergdreef 33, 1105 AZ Amsterdam, The Netherlands.
| | | | | | | | | | | |
Collapse
|
47
|
Godenschwege TA, Hu H, Shan-Crofts X, Goodman CS, Murphey RK. Bi-directional signaling by Semaphorin 1a during central synapse formation in Drosophila. Nat Neurosci 2002; 5:1294-301. [PMID: 12436113 DOI: 10.1038/nn976] [Citation(s) in RCA: 105] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2002] [Accepted: 10/22/2002] [Indexed: 11/09/2022]
Abstract
Semaphorins have been intensively studied for their role in dendritic and axonal pathfinding, but less is known about their potential role in synapse formation. In the adult giant fiber (GF) system of fruit flies (Drosophila melanogaster), we show that transmembrane Semaphorin 1a (Sema-1a) is involved in synapse formation in addition to its role in guidance during pathfinding. Cell-autonomous rescue experiments showed that Sema-1a is involved in assembly of a central synapse and that it is required both pre- and postsynaptically. We also found that pre- but not postsynaptic gain-of-function Sema-1a was able to disrupt the GF-motor neuron synapse and that the phenotype depended on a proline-rich intracellular domain that contains a putative Enabled binding site. We suggest that transmembrane Sema-1a is part of a bi-directional signaling system that leads to the formation of the GF synapse and possibly acts as both a ligand and a receptor.
Collapse
Affiliation(s)
- Tanja A Godenschwege
- University of Massachusetts, Department of Biology, Morrill Science Center, Amherst, Massachusetts 01003, USA
| | | | | | | | | |
Collapse
|
48
|
Qu X, Wei H, Zhai Y, Que H, Chen Q, Tang F, Wu Y, Xing G, Zhu Y, Liu S, Fan M, He F. Identification, characterization, and functional study of the two novel human members of the semaphorin gene family. J Biol Chem 2002; 277:35574-85. [PMID: 12110693 DOI: 10.1074/jbc.m206451200] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
We cloned two novel human transmembrane semaphorins, (HSA)SEMA6C and (HSA)SEMA6D, that belong to the class VI subgroup of the semaphorin family. The genes for SEMA6C and SEMA6D are mapped on chromosome 1q12-21.1 and 15q21.1, respectively. Among the adult tissues, SEMA6C is expressed only in skeletal muscle, whereas SEMA6D is expressed abundantly in kidney, brain, and placenta and moderately in the heart and skeletal muscles. During murine development, neither SEMA6C nor SEMA6D was expressed in embryonic day 10.5 (E10.5) embryos, but both were highly expressed in the areas of the lateral ventricle, the striatum, the wall of the midbrain, the pons/midbrain junction, and the choroid plexus of E13 embryos. Were neurons, neither axons nor astrocytes, highly expressed both semaphorins. Three isoforms of SEMA6C and five isoforms of SEMA6D derived from alternative splicing were identified, and their expression was regulated in a tissue- and development-dependent manner. Deletion analysis indicated that a sema domain and a PSI domain are integrally necessary for correct post-translation modification and subcellular localization. The extracellular domain of SEMA6C inhibited axonal extension of nerve growth factor-differentiated PC12 cells and induced the growth cone collapse of chicken dorsal root ganglion, rat hippocampal neurons, and rat cortical neurons in a dose-responsive manner. SEMA6D acted like SEMA6C except it had no significant effect on the growth cones of rat cortical neurons.
Collapse
Affiliation(s)
- Xianghu Qu
- Department of Genomics and Proteomics, Beijing Institute of Radiation Medicine, Chinese National Human Genome Center at Beijing
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Hirotani M, Ohoka Y, Yamamoto T, Nirasawa H, Furuyama T, Kogo M, Matsuya T, Inagaki S. Interaction of plexin-B1 with PDZ domain-containing Rho guanine nucleotide exchange factors. Biochem Biophys Res Commun 2002; 297:32-7. [PMID: 12220504 DOI: 10.1016/s0006-291x(02)02122-8] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The Rho family GTPase has been implicated in plexin-B1, a receptor for Semaphorin 4D (Sema4D), mediating signal transduction. Rho may also play a function in this signaling pathway as well as Rac, but the mechanisms for Rho regulation are poorly understood. In this study, we have identified two kinds of PDZ domain-containing Rho-specific guanine nucleotide exchange factors (RhoGEFs) as proteins interacting with plexin-B1 cytoplasmic domain. These PDZ domain-containing RhoGEFs showed significant homology to human KIAA0380 (PDZ-RhoGEF) and LARG (KIAA0382), respectively. Both KIAA0380 and LARG could bind plexin-B1 and a deletion mutant analysis of plexin-B1, KIAA0380 and LARG revealed that KIAA0380 and LARG bound plexin-B1 cytoplasmic tail through their PDZ domains. The tissue distribution analysis indicated that plexin-B1 was co-localized with KIAA0380 and LARG in various tissues. Immunocytochemical analysis showed that LARG was recruited to plasma membrane by plexin-B1. These results suggest that PDZ domain-containing RhoGEFs play a role in Sema4D-plexin-B1 mediating signal transduction.
Collapse
Affiliation(s)
- Mutsumi Hirotani
- Group of Neurobiology, School of Allied Health Sciences, Osaka University Faculty of Medicine, Yamadaoka 1-7, Suita-shi, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Beumer K, Matthies HJG, Bradshaw A, Broadie K. Integrins regulate DLG/FAS2 via a CaM kinase II-dependent pathway to mediate synapse elaboration and stabilization during postembryonic development. Development 2002; 129:3381-91. [PMID: 12091308 DOI: 10.1242/dev.129.14.3381] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Calcium/calmodulin dependent kinase II (CaMKII), PDZ-domain scaffolding protein Discs-large (DLG), immunoglobin superfamily cell adhesion molecule Fasciclin 2 (FAS2) and the position specific (PS) integrin receptors, including βPS and its alpha partners (αPS1, αPS2, αPS3/αVolado), are all known to regulate the postembryonic development of synaptic terminal arborization at the Drosophila neuromuscular junction (NMJ). Recent work has shown that DLG and FAS2 function together to modulate activity-dependent synaptic development and that this role is regulated by activation of CaMKII. We show that PS integrins function upstream of CaMKII in the development of synaptic architecture at the NMJ. βPS integrin physically associates with the synaptic complex anchored by the DLG scaffolding protein, which contains CaMKII and FAS2. We demonstrate an alteration of the FAS2 molecular cascade in integrin regulatory mutants, as a result of CaMKII/integrin interactions. Regulatory βPS integrin mutations increase the expression and synaptic localization of FAS2. Synaptic structural defects in βPS integrin mutants are rescued by transgenic overexpression of CaMKII (proximal in pathway) or genetic reduction of FAS2 (distal in pathway). These studies demonstrate that βPS integrins act through CaMKII activation to control the localization of synaptic proteins involved in the development of NMJ synaptic morphology.
Collapse
Affiliation(s)
- Kelly Beumer
- Department of Biology, University of Utah, Salt Lake City, UT 84112-0840, USA
| | | | | | | |
Collapse
|