1
|
Graus F, Sabater L, Gaig C, Gelpi E, Iranzo A, Dalmau JO, Santamaria J. Anti-IgLON5 Disease 10 Years Later: What We Know and What We Do Not Know. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2025; 12:e200353. [PMID: 39705634 DOI: 10.1212/nxi.0000000000200353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 10/24/2024] [Indexed: 12/22/2024]
Abstract
Anti-IgLON5 disease was identified 10 years ago, thanks to the discovery of IgLON5 antibodies and the joint effort of specialists in sleep medicine, neuroimmunology, and neuropathology. Without this collaboration, it would have been impossible to untangle fundamental aspects of this disease. After the seminal description in 2014, today there is growing evidence that most patients present a chronic progressive course with gait instability, abnormal movements, bulbar dysfunction, and a sleep disorder characterized by nonrapid eye movement and REM parasomnias, and obstructive sleep apnea with stridor. Unlike other autoimmune encephalitides, the response to immunotherapy is suboptimal. Neuropathologic studies in patients with a prolonged clinical course showed a novel 3-repeat and 4-repeat neuronal tauopathy mainly involving the hypothalamus and tegmentum of the brainstem. The absence of tau deposits in the brain of patients who died early, the demonstration that IgLON5 antibodies cause an irreversible decrease in cell-surface levels of IgLON5, and a disorganization of the neuronal cytoskeleton suggest that the disease is primarily autoimmune and the tauopathy a secondary event. After a decade, we now know the disease much better, but important issues still need to be addressed. We have to gather more information on the natural course of the disease, develop better treatments, and identify robust predictors of outcome. More basic research is needed on the physiology of IgLON5, how antibodies disrupt its function, and the downstream effects leading to neurodegeneration. Finally, better designed passive transfer and active immunization models are needed to confirm the pathogenic effect of IgLON5 antibodies.
Collapse
Affiliation(s)
- Francesc Graus
- Neuroimmunology Program, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Lidia Sabater
- Neuroimmunology Program, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Carles Gaig
- Service of Neurology, Hospital Clinic, Barcelona, Spain
- Multidisciplinary Sleep Disorders Unit, Hospital Clinic, Barcelona, Spain
| | - Ellen Gelpi
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health Vienna, Medical University of Vienna, Austria
| | - Alex Iranzo
- Service of Neurology, Hospital Clinic, Barcelona, Spain
- Multidisciplinary Sleep Disorders Unit, Hospital Clinic, Barcelona, Spain
| | - Josep O Dalmau
- Neuroimmunology Program, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Caixa Research Intitute (CRI), Barcelona, Spain; and
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Joan Santamaria
- Service of Neurology, Hospital Clinic, Barcelona, Spain
- Multidisciplinary Sleep Disorders Unit, Hospital Clinic, Barcelona, Spain
| |
Collapse
|
2
|
Su F, Pfundstein G, Sah S, Zhang S, Keable R, Hagan DW, Sharpe LJ, Clemens KJ, Begg D, Phelps EA, Brown AJ, Leshchyns'ka I, Sytnyk V. Neuronal growth regulator 1 (NEGR1) promotes the synaptic targeting of glutamic acid decarboxylase 65 (GAD65). J Neurochem 2025; 169:e16279. [PMID: 39676071 DOI: 10.1111/jnc.16279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 11/15/2024] [Accepted: 11/20/2024] [Indexed: 12/17/2024]
Abstract
Neuronal growth regulator 1 (NEGR1) is a synaptic plasma membrane localized cell adhesion molecule implicated in a wide spectrum of psychiatric disorders. By RNAseq analysis of the transcriptomic changes in the brain of NEGR1-deficient mice, we found that NEGR1 deficiency affects the expression of the Gad2 gene. We show that glutamic acid decarboxylase 65 (GAD65), the Gad2 - encoded enzyme synthesizing the inhibitory neurotransmitter GABA on synaptic vesicles, accumulates non-synaptically in brains of NEGR1-deficient mice. The density of non-synaptic GAD65 accumulations is also increased in NEGR1 deficient cultured hypothalamic neurons, and this effect is rescued by re-expression of NEGR1. By using a novel biosensor of the plasma membrane attachment of GAD65, we demonstrate that GAD65 attaches to the plasma membrane. NEGR1 promotes palmitoylation-dependent clearance of GAD65 from the plasma membrane and targeting of GAD65 to plasma membrane-derived endocytic vesicles. In NEGR1 deficient cultured hypothalamic neurons, the synaptic and extrasynaptic levels of the plasma membrane attached GAD65 are increased, and the synaptic levels of GABA are reduced. NEGR1-deficient mice are characterized by reduced body weight, lower GABAergic synapse densities in the arcuate nucleus, and blunted responsiveness to the reinforcing effects of food rewards. Our results indicate that abnormalities in synaptic GABA synthesis can contribute to brain disorders associated with abnormal expression of NEGR1 in humans.
Collapse
Affiliation(s)
- Feifei Su
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, New South Wales, Australia
| | - Grant Pfundstein
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, New South Wales, Australia
| | - Saroj Sah
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, New South Wales, Australia
| | - Shuyue Zhang
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, New South Wales, Australia
| | - Ryan Keable
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, New South Wales, Australia
| | - D Walker Hagan
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, USA
| | - Laura J Sharpe
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, New South Wales, Australia
| | - Kelly J Clemens
- School of Psychology, The University of New South Wales, Sydney, New South Wales, Australia
| | - Denovan Begg
- School of Psychology, The University of New South Wales, Sydney, New South Wales, Australia
| | - Edward A Phelps
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, USA
| | - Andrew J Brown
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, New South Wales, Australia
| | - Iryna Leshchyns'ka
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, New South Wales, Australia
| | - Vladimir Sytnyk
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
3
|
Zhang Q, Fujita M. Why nature evolved GPI-anchored proteins: unique structure characteristics enable versatile cell surface functions. Glycobiology 2024; 34:cwae089. [PMID: 39530348 PMCID: PMC11632373 DOI: 10.1093/glycob/cwae089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/02/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024] Open
Abstract
It remains a mystery why nature evolved the unique structural characteristics of GPI-anchored proteins (GPI-APs) and continues to sustain the complex, energy-intensive process of synthesizing these proteins. GPI-APs, despite their small size, rely on the coordinated activity of nearly 30 genes for their synthesis and remodeling, raising important evolutionary questions. The biological advantages of GPI-APs lie in their ability to rapidly redistribute across the cell membrane, localize within lipid rafts, utilize unique intracellular trafficking pathways, and function as both membrane-bound and soluble proteins. These properties allow GPI-APs to participate in diverse cellular processes such as synaptic plasticity, immune regulation, and signal transduction, highlighting their indispensable roles. Additionally, the shedding capability of GPI-APs extends their functional reach, adding further versatility to their biological roles. This review not only summarizes these key insights but also explores the broader implications of GPI-APs in cell signaling and disease. By understanding the evolutionary necessity of GPI-APs, we can better appreciate their complexity and potential as therapeutic targets.
Collapse
Affiliation(s)
- Qi Zhang
- Laboratory of Social Neural Networks, Faculty of Human Sciences, University of Tsukuba, 1-1-1Tennodai, Tsukuba 305-8577, Japan
| | - Morihisa Fujita
- Institute for Glyco-core Research (iGCORE), Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| |
Collapse
|
4
|
Kozlova I, Sytnyk V. Cell Adhesion Molecules as Modulators of the Epidermal Growth Factor Receptor. Cells 2024; 13:1919. [PMID: 39594667 PMCID: PMC11592701 DOI: 10.3390/cells13221919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Cell adhesion molecules (CAMs) are cell surface glycoproteins mediating interactions of cells with other cells and the extracellular matrix. By mediating the adhesion and modulating activity of other plasma membrane proteins, CAMs are involved in regulating a multitude of cellular processes, including growth, proliferation, migration, and survival of cells. In this review, we present evidence showing that various CAMs interact with the epidermal growth factor receptor (EGFR), a receptor tyrosine kinase inducing pro-proliferative and anti-apoptotic intracellular signaling in response to binding to several soluble ligands, including the epidermal growth factor. We discuss that CAMs are involved in regulating EGFR signaling by either potentiating or inhibiting the soluble ligand-dependent activation of EGFR. In addition, CAMs induce soluble ligand-independent forms of EGFR activity and regulate the levels of EGFR and its ligand-induced degradation. The CAM-dependent modulation of EGFR activity plays a key role in regulating the growth, proliferation, and survival of cells. Future research is needed to determine whether these processes can be targeted in both normal and cancerous cells by regulating interactions of EGFR with various CAMs.
Collapse
Affiliation(s)
| | - Vladimir Sytnyk
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW 2052, Australia;
| |
Collapse
|
5
|
Kurihara Y, Kawaguchi Y, Ohta Y, Kawasaki N, Fujita Y, Takei K. Nogo Receptor Antagonist LOTUS Promotes Neurite Outgrowth through Its Interaction with Teneurin-4. Cells 2024; 13:1369. [PMID: 39195260 DOI: 10.3390/cells13161369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/02/2024] [Accepted: 08/13/2024] [Indexed: 08/29/2024] Open
Abstract
Neurite outgrowth is a crucial process for organizing neuronal circuits in neuronal development and regeneration after injury. Regenerative failure in the adult mammalian central nervous system (CNS) is attributed to axonal growth inhibitors such as the Nogo protein that commonly binds to Nogo receptor-1 (NgR1). We previously reported that lateral olfactory tract usher substance (LOTUS) functions as an endogenous antagonist for NgR1 in forming neuronal circuits in the developing brain and improving axonal regeneration in the adult injured CNS. However, another molecular and cellular function of LOTUS remains unknown. In this study, we found that cultured retinal explant neurons extend their neurites on the LOTUS-coating substrate. This action was also observed in cultured retinal explant neurons derived from Ngr1-deficient mouse embryos, indicating that the promoting action of LOTUS on neurite outgrowth may be mediated by unidentified LOTUS-binding protein(s). We therefore screened the binding partner(s) of LOTUS by using a liquid chromatography-tandem mass spectrometry (LC-MS/MS). LC-MS/MS analysis and pull-down assay showed that LOTUS interacts with Teneurin-4 (Ten-4), a cell adhesion molecule. RNAi knockdown of Ten-4 inhibited neurite outgrowth on the LOTUS substrate in retinoic acid (RA)-treated Neuro2A cells. Furthermore, a soluble form of Ten-4 attenuates the promoting action on neurite outgrowth in cultured retinal explant neurons on the LOTUS substrate. These results suggest that LOTUS promotes neurite outgrowth by interacting with Ten-4. Our findings may provide a new molecular mechanism of LOTUS to contribute to neuronal circuit formation in development and to enhance axonal regeneration after CNS injury.
Collapse
Affiliation(s)
- Yuji Kurihara
- Molecular Medical Bioscience Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, Yokohama 230-0045, Japan
- Department of Anatomy & Developmental Biology, Faculty of Medicine, Shimane University, Izumo 693-8501, Japan
| | - Yuki Kawaguchi
- Molecular Medical Bioscience Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, Yokohama 230-0045, Japan
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama 230-0045, Japan
| | - Yuki Ohta
- Laboratory of Biopharmaceutical and Regenerative Sciences, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, Yokohama 230-0045, Japan
| | - Nana Kawasaki
- Laboratory of Biopharmaceutical and Regenerative Sciences, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, Yokohama 230-0045, Japan
| | - Yuki Fujita
- Department of Anatomy & Developmental Biology, Faculty of Medicine, Shimane University, Izumo 693-8501, Japan
| | - Kohtaro Takei
- Molecular Medical Bioscience Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, Yokohama 230-0045, Japan
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama 230-0045, Japan
| |
Collapse
|
6
|
Li S, Tang Q, Jiang Y, Chen X. Inherited glycosylphosphatidylinositol deficiency: a review from molecular and clinical perspectives. Acta Biochim Biophys Sin (Shanghai) 2024; 56:1234-1243. [PMID: 39081219 PMCID: PMC11466713 DOI: 10.3724/abbs.2024128] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 07/16/2024] [Indexed: 09/04/2024] Open
Abstract
Glycosylphosphatidylinositol (GPI) is a highly conserved post-translational modification in eukaryotes, which is essential for anchoring various proteins to the cell surface. Dysfunction of GPI biogenesis leads to human diseases, such as inherited GPI deficiency (IGD) caused by germline mutations in GPI-related genes. With accumulating reports on individuals with IGD, there has been increasing interest and studies on disease mechanism, diagnosis, and therapy. This review outlines the biosynthetic pathway of GPI-anchored proteins (GPI-APs) and summarizes clinical IGD cases from a molecular perspective. We also review current diagnostic and therapeutic approaches for IGD. Finally, we discuss future research directions to facilitate the understanding and treatment of GPI-related disorders.
Collapse
Affiliation(s)
- Shan Li
- Children’s Medical CenterPeking University First HospitalBeijing100034China
| | - Qi Tang
- College of Chemistry and Molecular EngineeringPeking UniversityBeijing100871China
- Beijing National Laboratory for Molecular SciencesPeking UniversityBeijing100871China
| | - Yuwu Jiang
- Children’s Medical CenterPeking University First HospitalBeijing100034China
| | - Xing Chen
- College of Chemistry and Molecular EngineeringPeking UniversityBeijing100871China
- Beijing National Laboratory for Molecular SciencesPeking UniversityBeijing100871China
- Peking-Tsinghua Center for Life SciencesPeking UniversityBeijing100871China
- Synthetic and Functional Biomolecules CenterPeking UniversityBeijing100871China
- Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of EducationPeking UniversityBeijing100871China
| |
Collapse
|
7
|
Salluzzo M, Vianello C, Abdullatef S, Rimondini R, Piccoli G, Carboni L. The Role of IgLON Cell Adhesion Molecules in Neurodegenerative Diseases. Genes (Basel) 2023; 14:1886. [PMID: 37895235 PMCID: PMC10606101 DOI: 10.3390/genes14101886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 09/26/2023] [Accepted: 09/27/2023] [Indexed: 10/29/2023] Open
Abstract
In the brain, cell adhesion molecules (CAMs) are critical for neurite outgrowth, axonal fasciculation, neuronal survival and migration, and synapse formation and maintenance. Among CAMs, the IgLON family comprises five members: Opioid Binding Protein/Cell Adhesion Molecule Like (OPCML or OBCAM), Limbic System Associated Membrane Protein (LSAMP), neurotrimin (NTM), Neuronal Growth Regulator 1 (NEGR1), and IgLON5. IgLONs exhibit three N-terminal C2 immunoglobulin domains; several glycosylation sites; and a glycosylphosphatidylinositol anchoring to the membrane. Interactions as homo- or heterodimers in cis and in trans, as well as binding to other molecules, appear critical for their functions. Shedding by metalloproteases generates soluble factors interacting with cellular receptors and activating signal transduction. The aim of this review was to analyse the available data implicating a role for IgLONs in neuropsychiatric disorders. Starting from the identification of a pathological role for antibodies against IgLON5 in an autoimmune neurodegenerative disease with a poorly understood mechanism of action, accumulating evidence links IgLONs to neuropsychiatric disorders, albeit with still undefined mechanisms which will require future thorough investigations.
Collapse
Affiliation(s)
- Marco Salluzzo
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy;
| | - Clara Vianello
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy; (C.V.); (R.R.)
| | - Sandra Abdullatef
- Department of Cellular, Computational and Integrative Biology, University of Trento, 38123 Trento, Italy; (S.A.); (G.P.)
| | - Roberto Rimondini
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy; (C.V.); (R.R.)
| | - Giovanni Piccoli
- Department of Cellular, Computational and Integrative Biology, University of Trento, 38123 Trento, Italy; (S.A.); (G.P.)
| | - Lucia Carboni
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy;
| |
Collapse
|
8
|
Gu Z, Matsuura K, Letelier A, Basista M, Craig C, Imai F, Yoshida Y. Axon Fasciculation, Mediated by Transmembrane Semaphorins, Is Critical for the Establishment of Segmental Specificity of Corticospinal Circuits. J Neurosci 2023; 43:5753-5768. [PMID: 37344234 PMCID: PMC10423052 DOI: 10.1523/jneurosci.0073-22.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 04/21/2023] [Accepted: 04/27/2023] [Indexed: 06/23/2023] Open
Abstract
Axon fasciculation is thought to be a critical step in neural circuit formation and function. Recent studies have revealed various molecular mechanisms that underlie axon fasciculation; however, the impacts of axon fasciculation, and its corollary, defasciculation, on neural circuit wiring remain unclear. Corticospinal (CS) neurons in the sensorimotor cortex project axons to the spinal cord to control skilled movements. In rodents, the axons remain tightly fasciculated in the brain and traverse the dorsal funiculus of the spinal cord. Here we show that plexinA1 (PlexA1) and plexinA3 (PlexA3) receptors are expressed by CS neurons, whereas their ligands, semaphorin-5A (Sema5A) and semaphorin-5B (Sema5B) are expressed in the medulla at the decussation site of CS axons to inhibit premature defasciculation of these axons. In the absence of Sema5A/5B-PlexA1/A3 signaling, some CS axons are prematurely defasciculated in the medulla of the brainstem, and those defasciculated CS axons aberrantly transverse in the spinal gray matter instead of the spinal dorsal funiculus. In the absence of Sema5A/Sema5B-PlexA1/A3 signaling, CS axons, which would normally innervate the lumbar spinal cord, are unbundled in the spinal gray matter, and prematurely innervate the cervical gray matter with reduced innervation of the lumbar gray matter. In both Sema5A/5B and PlexA1/A3 mutant mice (both sexes), stimulation of the hindlimb motor cortex aberrantly evokes robust forelimb muscle activation. Finally, Sema5A/5B and PlexA1/A3 mutant mice show deficits in skilled movements. These results suggest that proper fasciculation of CS axons is required for appropriate neural circuit wiring and ultimately affect the ability to perform skilled movements.SIGNIFICANCE STATEMENT Axon fasciculation is believed to be essential for neural circuit formation and function. However, whether and how defects in axon fasciculation affect the formation and function of neural circuits remain unclear. Here we examine whether the transmembrane proteins semaphorin-5A (Sema5A) and semaphorin-5B (Sema5B), and their receptors, plexinA1 (PlexA1) and plexinA3 (PlexA3) play roles in the development of corticospinal circuits. We find that Sema5A/Sema5B and PlexA1/A3 are required for proper axon fasciculation of corticospinal neurons. Furthermore, Sema5A/5B and PlexA1/A3 mutant mice show marked deficits in skilled motor behaviors. Therefore, these results strongly suggest that proper corticospinal axon fasciculation is required for the appropriate formation and functioning of corticospinal circuits in mice.
Collapse
Affiliation(s)
- Zirong Gu
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
| | - Ken Matsuura
- Neural Circuit Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, 904-0495, Japan
| | | | - Mark Basista
- Burke Neurological Institute, White Plains, New York 10605
- Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065
| | - Corey Craig
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
| | - Fumiyasu Imai
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
- Burke Neurological Institute, White Plains, New York 10605
- Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065
| | - Yutaka Yoshida
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
- Burke Neurological Institute, White Plains, New York 10605
- Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065
- Neural Circuit Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, 904-0495, Japan
| |
Collapse
|
9
|
Tang X, Tena J, Di Lucente J, Maezawa I, Harvey DJ, Jin LW, Lebrilla CB, Zivkovic AM. Transcriptomic and glycomic analyses highlight pathway-specific glycosylation alterations unique to Alzheimer's disease. Sci Rep 2023; 13:7816. [PMID: 37188790 PMCID: PMC10185676 DOI: 10.1038/s41598-023-34787-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 05/08/2023] [Indexed: 05/17/2023] Open
Abstract
Glycosylation has been found to be altered in the brains of individuals with Alzheimer's disease (AD). However, it is unknown which specific glycosylation-related pathways are altered in AD dementia. Using publicly available RNA-seq datasets covering seven brain regions and including 1724 samples, we identified glycosylation-related genes ubiquitously changed in individuals with AD. Several differentially expressed glycosyltransferases found by RNA-seq were confirmed by qPCR in a different set of human medial temporal cortex (MTC) samples (n = 20 AD vs. 20 controls). N-glycan-related changes predicted by expression changes in these glycosyltransferases were confirmed by mass spectrometry (MS)-based N-glycan analysis in the MTC (n = 9 AD vs. 6 controls). About 80% of glycosylation-related genes were differentially expressed in at least one brain region of AD participants (adjusted p-values < 0.05). Upregulation of MGAT1 and B4GALT1 involved in complex N-linked glycan formation and galactosylation, respectively, were reflected by increased concentrations of corresponding N-glycans. Isozyme-specific changes were observed in expression of the polypeptide N-acetylgalactosaminyltransferase (GALNT) family and the alpha-N-acetylgalactosaminide alpha-2,6-sialyltransferase (ST6GALNAC) family of enzymes. Several glycolipid-specific genes (UGT8, PIGM) were upregulated. The critical transcription factors regulating the expression of N-glycosylation and elongation genes were predicted and found to include STAT1 and HSF5. The miRNA predicted to be involved in regulating N-glycosylation and elongation glycosyltransferases were has-miR-1-3p and has-miR-16-5p, respectively. Our findings provide an overview of glycosylation pathways affected by AD and potential regulators of glycosyltransferase expression that deserve further validation and suggest that glycosylation changes occurring in the brains of AD dementia individuals are highly pathway-specific and unique to AD.
Collapse
Affiliation(s)
- Xinyu Tang
- Department of Nutrition, University of California, Davis, One Shields Ave, Davis, CA, 95616, USA
| | - Jennyfer Tena
- Department of Chemistry, University of California, Davis, Davis, CA, USA
| | - Jacopo Di Lucente
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California, Davis, Sacramento, CA, USA
- UC Davis MIND Institute, Sacramento, CA, USA
| | - Izumi Maezawa
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California, Davis, Sacramento, CA, USA
- UC Davis MIND Institute, Sacramento, CA, USA
| | - Danielle J Harvey
- Division of Biostatistics, Department of Public Health Sciences, School of Medicine, University of California, Davis, Davis, CA, USA
| | - Lee-Way Jin
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California, Davis, Sacramento, CA, USA
- UC Davis MIND Institute, Sacramento, CA, USA
| | - Carlito B Lebrilla
- Department of Chemistry, University of California, Davis, Davis, CA, USA
| | - Angela M Zivkovic
- Department of Nutrition, University of California, Davis, One Shields Ave, Davis, CA, 95616, USA.
| |
Collapse
|
10
|
Tonner H, Hunn S, Auler N, Schmelter C, Pfeiffer N, Grus FH. Dynamin-like Protein 1 (DNML1) as a Molecular Target for Antibody-Based Immunotherapy to Treat Glaucoma. Int J Mol Sci 2022; 23:ijms232113618. [PMID: 36362420 PMCID: PMC9654827 DOI: 10.3390/ijms232113618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 09/23/2022] [Accepted: 10/29/2022] [Indexed: 11/09/2022] Open
Abstract
Slow and progressive loss of retinal ganglion cells (RGCs) is the main characteristic of glaucoma, the second leading cause of blindness worldwide. Previous studies have shown that impaired mitochondrial dynamics could facilitate retinal neurodegeneration. Mitochondrial dynamics are regulated directly (fission) or more indirectly (fusion) by dynamin-like protein 1 (DNML1). Therefore, DNM1L might be a promising target for an antibody-based approach to treat glaucoma. The consequences of targeting endogenous DNM1L by antibodies in a glaucoma animal model have not been investigated yet. Here, we show that the intravitreal application of an anti-DNM1L antibody showed protective effects regarding the survival of RGCs and their axons in the retinal nerve fiber layer (RNFL). Antibody treatment also improved retinal functionality, as observed by electroretinography (Ganzfeld ERG). Western blot analysis revealed altered DNM1L phosphorylation and altered expression of proteins related to apoptosis suggesting a decreased apoptosis rate. Mass spectrometry analysis revealed 28 up-regulated and 21 down-regulated proteins (p < 0.05) in both experimental groups. Protein pathway analysis showed that many proteins interacted directly with the target protein DNM1L and could be classified into three main protein clusters: Vesicle traffic-associated (NSF, SNCA, ARF1), mitochondrion-associated (HSP9A, SLC25A5/ANT2, GLUD1) and cytoskeleton-associated (MAP1A) signaling pathway. Our results demonstrate that DNM1L is a promising target for an antibody-based approach to glaucoma therapy.
Collapse
Affiliation(s)
| | | | | | | | | | - Franz H. Grus
- Correspondence: ; Tel.: +49-6131-17-3328; Fax: +49-6131-4970563
| |
Collapse
|
11
|
Poondla N, Sheykhhasan M, Akbari M, Samadi P, Kalhor N, Manoochehri H. The Promise of CAR T-Cell Therapy for the treatment of cancer stem cells: A Short Review. Curr Stem Cell Res Ther 2022; 17:400-406. [PMID: 35176990 DOI: 10.2174/1574888x17666220217101817] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/27/2021] [Accepted: 11/15/2021] [Indexed: 11/22/2022]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is a type of sophisticated tailored immunotherapy used to treat a variety of tumors. Immunotherapy works by utilizing the body's own immune system to discover and destroy malignant cells. In CAR-T therapy, a patient's own immune cells are genetically engineered to recognize and attack cancer. Treatments employing CAR T-cells are currently showing promising therapeutic results in patients with hematologic malignancies, and their safety and feasibility in solid tumors has been verified. In this review, we will discuss in detail the likelihood that CAR T-cells inhibit cancer stem cells (CSCs) by selectively targeting their cell surface markers will ultimately improve the therapeutic response for patients with various forms of cancer. This review addresses the major components of cancer stem cell (CSC)-targeted CAR T-cells against malignancies, from bench to bedside.
Collapse
Affiliation(s)
- Naresh Poondla
- Richmond University Medical Center, 355, Bard Avenue, Staten Island, New York 10310, United States
| | - Mohsen Sheykhhasan
- Hamadan University of Medical Sciences, Research Center for Molecular Medicine; Academic Center for Education, Culture and Research (ACECR), Qom Branch, Qom, Iran
| | - Mohammad Akbari
- General Physician, Department of Medical School, Faculty of Medical Sciences, Islamic Azad University, Tonekabon Branch, Mazandaran, Iran
| | - Pouria Samadi
- Hamadan University of Medical Sciences, Research Center for Molecular Medicine
| | - Naser Kalhor
- Academic Center for Education, Culture and Research (ACECR), Qom Branch, Qom, Iran
| | - Hamed Manoochehri
- Hamadan University of Medical Sciences, Research Center for Molecular Medicine, Iran
| |
Collapse
|
12
|
Hirata T, Kobayashi A, Furuse T, Yamada I, Tamura M, Tomita H, Tokoro Y, Ninomiya A, Fujihara Y, Ikawa M, Maeda Y, Murakami Y, Kizuka Y, Kinoshita T. Loss of the N-acetylgalactosamine side chain of the GPI-anchor impairs bone formation and brain functions and accelerates the prion disease pathology. J Biol Chem 2022; 298:101720. [PMID: 35151686 PMCID: PMC8913354 DOI: 10.1016/j.jbc.2022.101720] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 01/28/2022] [Accepted: 01/29/2022] [Indexed: 02/07/2023] Open
Abstract
Glycosylphosphatidylinositol (GPI) is a posttranslational glycolipid modification of proteins that anchors proteins in lipid rafts on the cell surface. Although some GPI-anchored proteins (GPI-APs), including the prion protein PrPC, have a glycan side chain composed of N-acetylgalactosamine (GalNAc)−galactose−sialic acid on the core structure of GPI glycolipid, in vivo functions of this GPI-GalNAc side chain are largely unresolved. Here, we investigated the physiological and pathological roles of the GPI-GalNAc side chain in vivo by knocking out its initiation enzyme, PGAP4, in mice. We show that Pgap4 mRNA is highly expressed in the brain, particularly in neurons, and mass spectrometry analysis confirmed the loss of the GalNAc side chain in PrPC GPI in PGAP4-KO mouse brains. Furthermore, PGAP4-KO mice exhibited various phenotypes, including an elevated blood alkaline phosphatase level, impaired bone formation, decreased locomotor activity, and impaired memory, despite normal expression levels and lipid raft association of various GPI-APs. Thus, we conclude that the GPI-GalNAc side chain is required for in vivo functions of GPI-APs in mammals, especially in bone and the brain. Moreover, PGAP4-KO mice were more vulnerable to prion diseases and died earlier after intracerebral inoculation of the pathogenic prion strains than wildtype mice, highlighting the protective roles of the GalNAc side chain against prion diseases.
Collapse
Affiliation(s)
- Tetsuya Hirata
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, Japan
| | - Atsushi Kobayashi
- Laboratory of Comparative Pathology, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Tamio Furuse
- Technology and Development Team for Mouse Phenotype Analysis, RIKEN BioResource Research Center, Tsukuba, Ibaraki, Japan
| | - Ikuko Yamada
- Technology and Development Team for Mouse Phenotype Analysis, RIKEN BioResource Research Center, Tsukuba, Ibaraki, Japan
| | - Masaru Tamura
- Technology and Development Team for Mouse Phenotype Analysis, RIKEN BioResource Research Center, Tsukuba, Ibaraki, Japan
| | - Hiroyuki Tomita
- Department of Tumor Pathology, Graduate School of Medicine, Gifu University, Gifu, Japan
| | - Yuko Tokoro
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, Japan
| | - Akinori Ninomiya
- Core Instrumentation Facility, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Yoshitaka Fujihara
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Masahito Ikawa
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Yusuke Maeda
- Yabumoto Department of Intractable Disease Research, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan; WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Yoshiko Murakami
- Yabumoto Department of Intractable Disease Research, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan; WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Yasuhiko Kizuka
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, Japan.
| | - Taroh Kinoshita
- Yabumoto Department of Intractable Disease Research, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan; WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan.
| |
Collapse
|
13
|
Licheri V, Brigman JL. Altering Cell-Cell Interaction in Prenatal Alcohol Exposure Models: Insight on Cell-Adhesion Molecules During Brain Development. Front Mol Neurosci 2022; 14:753537. [PMID: 34975396 PMCID: PMC8715949 DOI: 10.3389/fnmol.2021.753537] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 11/11/2021] [Indexed: 11/17/2022] Open
Abstract
Alcohol exposure during pregnancy disrupts the development of the brain and produces long lasting behavioral and cognitive impairments collectively known as Fetal Alcohol Spectrum Disorders (FASDs). FASDs are characterized by alterations in learning, working memory, social behavior and executive function. A large body of literature using preclinical prenatal alcohol exposure models reports alcohol-induced changes in architecture and activity in specific brain regions affecting cognition. While multiple putative mechanisms of alcohol’s long-lasting effects on morphology and behavior have been investigated, an area that has received less attention is the effect of alcohol on cell adhesion molecules (CAMs). The embryo/fetal development represents a crucial period for Central Nervous System (CNS) development during which the cell-cell interaction plays an important role. CAMs play a critical role in neuronal migration and differentiation, synaptic organization and function which may be disrupted by alcohol. In this review, we summarize the physiological structure and role of CAMs involved in brain development, review the current literature on prenatal alcohol exposure effects on CAM function in different experimental models and pinpoint areas needed for future study to better understand how CAMs may mediate the morphological, sensory and behavioral outcomes in FASDs.
Collapse
Affiliation(s)
- Valentina Licheri
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Jonathan L Brigman
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, United States.,New Mexico Alcohol Research Center, UNM Health Sciences Center, Albuquerque, NM, United States
| |
Collapse
|
14
|
Grüner J, Stengel H, Werner C, Appeltshauser L, Sommer C, Villmann C, Doppler K. Anti-contactin-1 Antibodies Affect Surface Expression and Sodium Currents in Dorsal Root Ganglia. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2021; 8:8/5/e1056. [PMID: 34429341 PMCID: PMC8407150 DOI: 10.1212/nxi.0000000000001056] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 05/17/2021] [Indexed: 01/13/2023]
Abstract
Background and Objectives As autoantibodies to contactin-1 from patients with chronic inflammatory demyelinating polyradiculoneuropathy not only bind to the paranodes where they are supposed to cause conduction failure but also bind to other neuronal cell types, we aimed to investigate the effect of anti–contactin-1 autoantibodies on contactin-1 surface expression in cerebellar granule neurons, dorsal root ganglion neurons, and contactin-1–transfected human embryonic kidney 293 cells. Methods Immunocytochemistry including structured illumination microscopy and immunoblotting was used to determine expression levels of contactin-1 and/or sodium channels after long-term exposure to autoantibodies from 3 seropositive patients. For functional analysis of sodium channels, whole-cell recordings of sodium currents were performed on dorsal root ganglion neurons incubated with anti–contactin-1 autoantibodies. Results We found a reduction in contactin-1 expression levels on dorsal root ganglion neurons, cerebellar granule neurons, and contactin-1–transfected human embryonic kidney 293 cells and decreased dorsal root ganglion sodium currents after long-term exposure to anti–contactin-1 autoantibodies. Sodium channel density did not decrease. Discussion Our results demonstrate a direct effect of anti–contactin-1 autoantibodies on the surface expression of contactin-1 and sodium currents in dorsal root ganglion neurons. This may be the pathophysiologic correlate of sensory ataxia reported in these patients.
Collapse
Affiliation(s)
- Julia Grüner
- From the Department of Neurology (J.G., H.S., L.A., C.S., K.D.), University Hospital Würzburg, Germany; Department of Biotechnology and Biophysics (C.W.), Julius-Maximilians-University of Würzburg; and Institute of Clinical Neurobiology (C.V.), University Hospital, Julius-Maximilians-University of Würzburg, Germany
| | - Helena Stengel
- From the Department of Neurology (J.G., H.S., L.A., C.S., K.D.), University Hospital Würzburg, Germany; Department of Biotechnology and Biophysics (C.W.), Julius-Maximilians-University of Würzburg; and Institute of Clinical Neurobiology (C.V.), University Hospital, Julius-Maximilians-University of Würzburg, Germany
| | - Christian Werner
- From the Department of Neurology (J.G., H.S., L.A., C.S., K.D.), University Hospital Würzburg, Germany; Department of Biotechnology and Biophysics (C.W.), Julius-Maximilians-University of Würzburg; and Institute of Clinical Neurobiology (C.V.), University Hospital, Julius-Maximilians-University of Würzburg, Germany
| | - Luise Appeltshauser
- From the Department of Neurology (J.G., H.S., L.A., C.S., K.D.), University Hospital Würzburg, Germany; Department of Biotechnology and Biophysics (C.W.), Julius-Maximilians-University of Würzburg; and Institute of Clinical Neurobiology (C.V.), University Hospital, Julius-Maximilians-University of Würzburg, Germany
| | - Claudia Sommer
- From the Department of Neurology (J.G., H.S., L.A., C.S., K.D.), University Hospital Würzburg, Germany; Department of Biotechnology and Biophysics (C.W.), Julius-Maximilians-University of Würzburg; and Institute of Clinical Neurobiology (C.V.), University Hospital, Julius-Maximilians-University of Würzburg, Germany
| | - Carmen Villmann
- From the Department of Neurology (J.G., H.S., L.A., C.S., K.D.), University Hospital Würzburg, Germany; Department of Biotechnology and Biophysics (C.W.), Julius-Maximilians-University of Würzburg; and Institute of Clinical Neurobiology (C.V.), University Hospital, Julius-Maximilians-University of Würzburg, Germany
| | - Kathrin Doppler
- From the Department of Neurology (J.G., H.S., L.A., C.S., K.D.), University Hospital Würzburg, Germany; Department of Biotechnology and Biophysics (C.W.), Julius-Maximilians-University of Würzburg; and Institute of Clinical Neurobiology (C.V.), University Hospital, Julius-Maximilians-University of Würzburg, Germany.
| |
Collapse
|
15
|
Kandasamy LC, Tsukamoto M, Banov V, Tsetsegee S, Nagasawa Y, Kato M, Matsumoto N, Takeda J, Itohara S, Ogawa S, Young LJ, Zhang Q. Limb-clasping, cognitive deficit and increased vulnerability to kainic acid-induced seizures in neuronal glycosylphosphatidylinositol deficiency mouse models. Hum Mol Genet 2021; 30:758-770. [PMID: 33607654 PMCID: PMC8161520 DOI: 10.1093/hmg/ddab052] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 02/04/2021] [Accepted: 02/11/2021] [Indexed: 11/26/2022] Open
Abstract
Posttranslational modification of a protein with glycosylphosphatidylinositol (GPI) is a conserved mechanism exists in all eukaryotes. Thus far, >150 human GPI-anchored proteins have been discovered and ~30 enzymes have been reported to be involved in the biosynthesis and maturation of mammalian GPI. Phosphatidylinositol glycan biosynthesis class A protein (PIGA) catalyzes the very first step of GPI anchor biosynthesis. Patients carrying a mutation of the PIGA gene usually suffer from inherited glycosylphosphatidylinositol deficiency (IGD) with intractable epilepsy and intellectual developmental disorder. We generated three mouse models with PIGA deficits specifically in telencephalon excitatory neurons (Ex-M-cko), inhibitory neurons (In-M-cko) or thalamic neurons (Th-H-cko), respectively. Both Ex-M-cko and In-M-cko mice showed impaired long-term fear memory and were more susceptible to kainic acid-induced seizures. In addition, In-M-cko demonstrated a severe limb-clasping phenotype. Hippocampal synapse changes were observed in Ex-M-cko mice. Our Piga conditional knockout mouse models provide powerful tools to understand the cell-type specific mechanisms underlying inherited GPI deficiency and to test different therapeutic modalities.
Collapse
Affiliation(s)
- Lenin C Kandasamy
- Laboratory of Social Neural Networks, Center for Social Neural Networks, University of Tsukuba, Tsukuba 305-8577, Japan
| | - Mina Tsukamoto
- Laboratory of Social Neural Networks, Center for Social Neural Networks, University of Tsukuba, Tsukuba 305-8577, Japan
| | - Vitaliy Banov
- Laboratory for Behavioral Genetics, CBS, RIKEN, Wako 351-0198, Japan.,Institute of Neuroinformatics, University of Zürich, ETH Zürich, Zürich 8057, Switzerland
| | - Sambuu Tsetsegee
- Laboratory of Social Neural Networks, Center for Social Neural Networks, University of Tsukuba, Tsukuba 305-8577, Japan
| | - Yutaro Nagasawa
- Laboratory of Social Neural Networks, Center for Social Neural Networks, University of Tsukuba, Tsukuba 305-8577, Japan
| | - Mitsuhiro Kato
- Department of Pediatrics, Showa University School of Medicine, Tokyo 142-8555, Japan
| | - Naomichi Matsumoto
- Department of Human Genetics, Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan
| | - Junji Takeda
- Yabumoto Department of Intractable Disease Research, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | | | - Sonoko Ogawa
- Laboratory of Behavioral Neuroendocrinology, Faculty of Human Sciences, University of Tsukuba, Tsukuba 305-8577, Japan
| | - Larry J Young
- Faculty of Human Sciences, Center for Social Neural Networks, University of Tsukuba, Tsukuba 305-8577, Japan.,Center for Translational Social Neuroscience, Department of Psychiatry and Behavioral Sciences, Yerkes National Primate Research Center, Emory University, Atlanta GA 30329, USA
| | - Qi Zhang
- Laboratory of Social Neural Networks, Center for Social Neural Networks, University of Tsukuba, Tsukuba 305-8577, Japan.,Laboratory for Behavioral Genetics, CBS, RIKEN, Wako 351-0198, Japan.,Faculty of Human Sciences, Center for Social Neural Networks, University of Tsukuba, Tsukuba 305-8577, Japan
| |
Collapse
|
16
|
Lim JH, Beg MMA, Ahmad K, Shaikh S, Ahmad SS, Chun HJ, Choi D, Lee WJ, Jin JO, Kim J, Jan AT, Lee EJ, Choi I. IgLON5 Regulates the Adhesion and Differentiation of Myoblasts. Cells 2021; 10:417. [PMID: 33671182 PMCID: PMC7922608 DOI: 10.3390/cells10020417] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 02/06/2021] [Accepted: 02/15/2021] [Indexed: 02/07/2023] Open
Abstract
IgLON5 is a cell adhesion protein belonging to the immunoglobulin superfamily and has important cellular functions. The objective of this study was to determine the role played by IgLON5 during myogenesis. We found IgLON5 expression progressively increased in C2C12 myoblasts during transition from the adhesion to differentiation stage. IgLON5 knockdown (IgLON5kd) cells exhibited reduced cell adhesion, myotube formation, and maturation and reduced expressions of different types of genes, including those coding for extracellular matrix (ECM) components (COL1a1, FMOD, DPT, THBS1), cell membrane proteins (ITM2a, CDH15), and cytoskeletal protein (WASP). Furthermore, decreased IgLON5 expression in FMODkd, DPTkd, COL1a1kd, and ITM2akd cells suggested that IgLON5 and these genes mutually control gene expression during myogenesis. IgLON5 immunoneutralization resulted in significant reduction in the protein level of myogenic markers (MYOD, MYOG, MYL2). IgLON5 expression was higher in the CTX-treated gastrocnemius mice muscles (day 7), which confirmed increase expression of IgLON5 during muscle. Collectively, these results suggest IgLON5 plays an important role in myogenesis, muscle regeneration, and that proteins in ECM and myoblast membranes form an interactive network that establishes an essential microenvironment that ensures muscle stem cell survival.
Collapse
Affiliation(s)
- Jeong Ho Lim
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea; (J.H.L.); (M.M.A.B.); (S.S.); (S.S.A.); (H.J.C.); (D.C.); (J.-O.J.); (J.K.)
| | - Mirza Masroor Ali Beg
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea; (J.H.L.); (M.M.A.B.); (S.S.); (S.S.A.); (H.J.C.); (D.C.); (J.-O.J.); (J.K.)
| | - Khurshid Ahmad
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Korea;
| | - Sibhghatulla Shaikh
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea; (J.H.L.); (M.M.A.B.); (S.S.); (S.S.A.); (H.J.C.); (D.C.); (J.-O.J.); (J.K.)
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Korea;
| | - Syed Sayeed Ahmad
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea; (J.H.L.); (M.M.A.B.); (S.S.); (S.S.A.); (H.J.C.); (D.C.); (J.-O.J.); (J.K.)
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Korea;
| | - Hee Jin Chun
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea; (J.H.L.); (M.M.A.B.); (S.S.); (S.S.A.); (H.J.C.); (D.C.); (J.-O.J.); (J.K.)
| | - Dukhwan Choi
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea; (J.H.L.); (M.M.A.B.); (S.S.); (S.S.A.); (H.J.C.); (D.C.); (J.-O.J.); (J.K.)
| | - Woo-Jong Lee
- Biomedical Manufacturing Technology Center, Korea Institute of Industrial Technology, Yeongcheon 38822, Korea;
| | - Jun-O Jin
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea; (J.H.L.); (M.M.A.B.); (S.S.); (S.S.A.); (H.J.C.); (D.C.); (J.-O.J.); (J.K.)
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Korea;
| | - Jihoe Kim
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea; (J.H.L.); (M.M.A.B.); (S.S.); (S.S.A.); (H.J.C.); (D.C.); (J.-O.J.); (J.K.)
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Korea;
| | - Arif Tasleem Jan
- School of Biosciences and Biotechnology, Baba Ghulam Shah Badshah University, Rajouri 185234, India;
| | - Eun Ju Lee
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea; (J.H.L.); (M.M.A.B.); (S.S.); (S.S.A.); (H.J.C.); (D.C.); (J.-O.J.); (J.K.)
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Korea;
| | - Inho Choi
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea; (J.H.L.); (M.M.A.B.); (S.S.); (S.S.A.); (H.J.C.); (D.C.); (J.-O.J.); (J.K.)
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Korea;
| |
Collapse
|
17
|
Kozlova I, Sah S, Keable R, Leshchyns'ka I, Janitz M, Sytnyk V. Cell Adhesion Molecules and Protein Synthesis Regulation in Neurons. Front Mol Neurosci 2020; 13:592126. [PMID: 33281551 PMCID: PMC7689008 DOI: 10.3389/fnmol.2020.592126] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 10/16/2020] [Indexed: 12/18/2022] Open
Abstract
Cell adhesion molecules (CAMs) mediate interactions of neurons with the extracellular environment by forming adhesive bonds with CAMs on adjacent membranes or via binding to proteins of the extracellular matrix. Binding of CAMs to their extracellular ligands results in the activation of intracellular signaling cascades, leading to changes in neuronal structure and the molecular composition and function of neuronal contacts. Ultimately, many of these changes depend on the synthesis of new proteins. In this review, we summarize the evidence showing that CAMs regulate protein synthesis by modulating the activity of transcription factors, gene expression, protein translation, and the structure and distribution of organelles involved in protein synthesis and transport.
Collapse
Affiliation(s)
- Irina Kozlova
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW, Australia
| | - Saroj Sah
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW, Australia
| | - Ryan Keable
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW, Australia
| | - Iryna Leshchyns'ka
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW, Australia
| | - Michael Janitz
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW, Australia
| | - Vladimir Sytnyk
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
18
|
Landa J, Gaig C, Plagumà J, Saiz A, Antonell A, Sanchez-Valle R, Dalmau J, Graus F, Sabater L. Effects of IgLON5 Antibodies on Neuronal Cytoskeleton: A Link between Autoimmunity and Neurodegeneration. Ann Neurol 2020; 88:1023-1027. [PMID: 32740999 DOI: 10.1002/ana.25857] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 07/28/2020] [Accepted: 07/29/2020] [Indexed: 12/27/2022]
Abstract
Anti-IgLON5 disease is a neurological disorder characterized by autoantibodies against IgLON5 and pathological evidence of neuronal-specific tau accumulation. Here, we report that patients' IgLON5 IgG, but not other cell-surface antibodies, disrupt the cytoskeletal organization in cultured rat hippocampal neurons, resulting in dystrophic neurites and axonal swelling. Adsorption of IgLON5 IgG with HEK293 cells expressing IgLON5 abrogated the indicated cytoskeletal changes. These findings, along with an increase of levels of neurofilaments in patients' cerebrospinal fluid, suggest that IgLON5 IgG, unlike other cell-surface antibodies, disrupts neuronal cytoskeleton maintenance, providing a link between autoimmunity and neurodegeneration. ANN NEUROL 2020;88:1023-1027.
Collapse
Affiliation(s)
- Jon Landa
- Neuroimmunology Program, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Carles Gaig
- Neuroimmunology Program, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain.,Service of Neurology, Hospital Clinic de Barcelona, Barcelona, Spain
| | - Jesús Plagumà
- Neuroimmunology Program, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Albert Saiz
- Neuroimmunology Program, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain.,Service of Neurology, Hospital Clinic de Barcelona, Barcelona, Spain
| | - Ana Antonell
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Department, Hospital Clinic, IDIBAPS, Universitat de Barcelona, Barcelona, Spain
| | - Raquel Sanchez-Valle
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Department, Hospital Clinic, IDIBAPS, Universitat de Barcelona, Barcelona, Spain
| | - Josep Dalmau
- Neuroimmunology Program, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain.,Service of Neurology, Hospital Clinic de Barcelona, Barcelona, Spain.,Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Francesc Graus
- Neuroimmunology Program, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Lidia Sabater
- Neuroimmunology Program, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
19
|
Zhang L, Tian W, Zhou B. Polymorphisms in Neuronal Growth Regulator 1 and Otoancorin Alternate the Susceptibility to Lung Cancer in Chinese Nonsmoking Females. DNA Cell Biol 2020; 39:1657-1663. [PMID: 32552051 DOI: 10.1089/dna.2020.5654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Cell adhesion molecules (CAMs) play crucial roles in the genesis and progress of tumor. We investigated the effects of single nucleotide polymorphisms (SNPs) of CAMs, neuronal growth regulator 1 (NEGR1), and Otoancorin (OTOA) on lung cancer susceptibility in Chinese nonsmoking females. Logistic regression and Cox regression analyses were conducted to investigate the effects of SNPs and environmental factors. For rs3102911, genotype TT carriers decreased the risk of lung cancer with an odds ratio (OR) of 0.635. AA genotypes of rs741718 increased the risk of lung cancer with an OR of 3.527. In stratified analysis, genotype AA carriers of rs741718 had a high susceptibility to lung adenocarcinoma compared with GG and AG genotypes. Analyses of association between SNPs and clinical characteristics revealed that rs3102911 as a protective factor and rs741718 as a risk factor influenced the lung cancer occurrence and progression in nonsmoking females.
Collapse
Affiliation(s)
- Ludan Zhang
- Department of Clinical Epidemiology, First Affiliated Hospital, China Medical University, Shenyang, China.,Department of Clinical Medicine, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Wen Tian
- Department of Clinical Epidemiology, First Affiliated Hospital, China Medical University, Shenyang, China.,Department of Epidemiology, School of Public Health, China Medical University, Shenyang, China
| | - Baosen Zhou
- Department of Clinical Epidemiology, First Affiliated Hospital, China Medical University, Shenyang, China.,Department of Epidemiology, School of Public Health, China Medical University, Shenyang, China
| |
Collapse
|
20
|
Noh K, Park JC, Han JS, Lee SJ. From Bound Cells Comes a Sound Mind: The Role of Neuronal Growth Regulator 1 in Psychiatric Disorders. Exp Neurobiol 2020; 29:1-10. [PMID: 32122104 PMCID: PMC7075657 DOI: 10.5607/en.2020.29.1.1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 02/23/2020] [Accepted: 02/25/2020] [Indexed: 12/21/2022] Open
Abstract
Cell-to-cell adhesion is important for maintenance of brain structure and function. Abnormal neuronal cell adhesion and loss of its connectivity are considered a main cause of psychiatric disorders such as major depressive disorder (MDD). Various cell adhesion molecules (CAMs) are involved in neuronal cell adhesions and thereby affect brain functions such as learning and memory, cognitive functions, and psychiatric functions. Compared with other CAMs, neuronal growth regulator 1 (Negr1) has a distinct functioning mechanism in terms of its cross-talk with cytokine receptor signaling. Negr1 is a member of the immunoglobulin LON (IgLON) family of proteins and is involved in neuronal outgrowth, dendritic arborization, and synapse formation. In humans, Negr1 is a risk gene for obesity based on a genome-wide association study. More recently, accumulating evidence supports that it also plays a critical role in psychiatric disorders. In this review, we discuss the recent findings on the role of Negr1 in MDD, focusing on its regulatory mechanism. We also provide evidence of putative involvement of Negr1 in other psychiatric disorders based on the novel behavioral phenotypes of Negr1 knockout mice.
Collapse
Affiliation(s)
- Kyungchul Noh
- Department of Physiology and Neuroscience, Dental Research Institute, Seoul National University School of Dentistry, Seoul 08826, Korea
| | - Jung-Cheol Park
- Department of Biological Science, Konkuk University, Seoul 05029, Korea
| | - Jung-Soo Han
- Department of Biological Science, Konkuk University, Seoul 05029, Korea
| | - Sung Joong Lee
- Department of Physiology and Neuroscience, Dental Research Institute, Seoul National University School of Dentistry, Seoul 08826, Korea
| |
Collapse
|
21
|
Wang X, Xu G, Yang N, Yan Y, Wu G, Sun C. Differential proteomic analysis revealed crucial egg white proteins for hatchability of chickens. Poult Sci 2019; 98:7076-7089. [PMID: 31424521 PMCID: PMC8913984 DOI: 10.3382/ps/pez459] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 07/26/2019] [Indexed: 11/20/2022] Open
Abstract
For healthy development, an avian embryo needs the nutritional and functional molecules maternally deposited in avian eggs. Egg white not only provides nutritional components but also exhibits functional properties, such as defenses against microbial invasion. However, the roles of the more detailed messages in embryo development remain unclear. In this study, a tandem mass tag labeling quantitation approach was used to innovatively identify the differential proteins in the egg whites of fresh eggs produced by hens with divergent high/low hatchability and in the egg whites of embryonated eggs with healthy and dead embryos. A total of 378 proteins were quantified in egg white, which is the most complete proteome identified for egg white to date, and up to 102 differential proteins were identified. GO enrichment, pathway, and hierarchical clustering analysis revealed some of the differential proteins that are the main participants in several biological processes, including blood coagulation, intermediate filament, antibacterial activity, and neurodevelopment. A list of 11 putative protein biomarkers, such as keratin (KRT19, KRT12, KRT15, and KRT6A), which is involved in cell architecture, and fibrinogen (fibrinogen alpha chain, fibrinogen beta chain, and fibrinogen gamma chain), which is related to blood coagulation, were ultimately screened. The current study screened egg white proteins that can predict low hatchability and embryonic death and deciphered the role of these proteins in embryonic development, which is meaningful for the comprehensive understanding of embryonic growth.
Collapse
Affiliation(s)
- Xiqiong Wang
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Guiyun Xu
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Ning Yang
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yiyuan Yan
- Beijing Engineering Research Center of Layer, Beijing 101206, China
| | - Guiqin Wu
- Beijing Engineering Research Center of Layer, Beijing 101206, China
| | - Congjiao Sun
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| |
Collapse
|
22
|
Shida M, Mikami T, Tamura JI, Kitagawa H. Chondroitin sulfate-D promotes neurite outgrowth by acting as an extracellular ligand for neuronal integrin αVβ3. Biochim Biophys Acta Gen Subj 2019; 1863:1319-1331. [PMID: 31181256 DOI: 10.1016/j.bbagen.2019.06.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 05/20/2019] [Accepted: 06/05/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Chondroitin sulfate (CS) chains are prominent extra/pericellular matrix components in the central nervous system (CNS) and can exert positive or negative regulatory effects on neurite outgrowth, depending on the CS structure and the amount. Despite the remarkable abilities of highly sulfated forms of CS chains to enhance neurite outgrowth, the neuronal recognition systems for such promotional CS chains, including CS-D polysaccharide, remain to be fully elucidated. METHODS We explored the molecular basis of the CS-D-mediated neurite extension using primary hippocampal neurons cultured on substrate precoated with CS-D polysaccharides, and evaluated functional involvement of a distinct integrin heterodimer as a novel neuronal CS receptor for CS-D. RESULTS We identified an extracellular matrix receptor, integrin αVβ3, as a functional receptor for CS-D. CS-D, but not CS-C (a precursor form of CS-D) showed significant binding affinity toward recombinant integrin αVβ3 heterodimer and activated intracellular signaling(s) involving focal adhesion kinase (FAK) and Src/Fyn kinase. Functional blockade of the respective players for integrin signaling abrogated the promotional effects of CS-D. We also found the existence of CS-D-induced integrin activation system in neuronal stem/progenitor cell population. CONCLUSIONS The neuronal cell surface integrin αVβ3 can function as a CS receptor for a highly sulfated CS subtype, CS-D. GENERAL SIGNIFICANCE Our findings are the first to demonstrate that CS-dependent neurite outgrowth promotion is exerted via direct activation of specific integrin heterodimers on neuronal cell surfaces, providing new insights into understanding the CS-sensing machineries that regulate CNS development and regeneration.
Collapse
Affiliation(s)
- Miharu Shida
- Laboratory of Biochemistry, Kobe Pharmaceutical University, Higashinada-ku, Kobe 658-8558, Japan
| | - Tadahisa Mikami
- Laboratory of Biochemistry, Kobe Pharmaceutical University, Higashinada-ku, Kobe 658-8558, Japan
| | - Jun-Ichi Tamura
- Department of Life and Environmental Agricultural Sciences, Faculty of Agriculture, Tottori University, Tottori 680-8551, Japan
| | - Hiroshi Kitagawa
- Laboratory of Biochemistry, Kobe Pharmaceutical University, Higashinada-ku, Kobe 658-8558, Japan.
| |
Collapse
|
23
|
Cheng S, Park Y, Kurleto JD, Jeon M, Zinn K, Thornton JW, Özkan E. Family of neural wiring receptors in bilaterians defined by phylogenetic, biochemical, and structural evidence. Proc Natl Acad Sci U S A 2019; 116:9837-9842. [PMID: 31043568 PMCID: PMC6525511 DOI: 10.1073/pnas.1818631116] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The evolution of complex nervous systems was accompanied by the expansion of numerous protein families, including cell-adhesion molecules, surface receptors, and their ligands. These proteins mediate axonal guidance, synapse targeting, and other neuronal wiring-related functions. Recently, 32 interacting cell surface proteins belonging to two newly defined families of the Ig superfamily (IgSF) in fruit flies were discovered to label different subsets of neurons in the brain and ventral nerve cord. They have been shown to be involved in synaptic targeting and morphogenesis, retrograde signaling, and neuronal survival. Here, we show that these proteins, Dprs and DIPs, are members of a widely distributed family of two- and three-Ig domain molecules with neuronal wiring functions, which we refer to as Wirins. Beginning from a single ancestral Wirin gene in the last common ancestor of Bilateria, numerous gene duplications produced the heterophilic Dprs and DIPs in protostomes, along with two other subfamilies that diversified independently across protostome phyla. In deuterostomes, the ancestral Wirin evolved into the IgLON subfamily of neuronal receptors. We show that IgLONs interact with each other and that their complexes can be broken by mutations designed using homology models based on Dpr and DIP structures. The nematode orthologs ZIG-8 and RIG-5 also form heterophilic and homophilic complexes, and crystal structures reveal numerous apparently ancestral features shared with Dpr-DIP complexes. The evolutionary, biochemical, and structural relationships we demonstrate here provide insights into neural development and the rise of the metazoan nervous system.
Collapse
Affiliation(s)
- Shouqiang Cheng
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL 60637
| | - Yeonwoo Park
- Committee on Genetics, Genomics and Systems Biology, The University of Chicago, Chicago, IL 60637
| | - Justyna D Kurleto
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL 60637
- Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland
| | - Mili Jeon
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
| | - Kai Zinn
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
| | - Joseph W Thornton
- Committee on Genetics, Genomics and Systems Biology, The University of Chicago, Chicago, IL 60637
- Department of Human Genetics, The University of Chicago, Chicago, IL 60637
- Department of Ecology and Evolution, The University of Chicago, Chicago, IL 60637
| | - Engin Özkan
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL 60637;
| |
Collapse
|
24
|
Ranaivoson FM, Turk LS, Ozgul S, Kakehi S, von Daake S, Lopez N, Trobiani L, De Jaco A, Denissova N, Demeler B, Özkan E, Montelione GT, Comoletti D. A Proteomic Screen of Neuronal Cell-Surface Molecules Reveals IgLONs as Structurally Conserved Interaction Modules at the Synapse. Structure 2019; 27:893-906.e9. [PMID: 30956130 DOI: 10.1016/j.str.2019.03.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 01/10/2019] [Accepted: 03/07/2019] [Indexed: 12/21/2022]
Abstract
In the developing brain, cell-surface proteins play crucial roles, but their protein-protein interaction network remains largely unknown. A proteomic screen identified 200 interactions, 89 of which were not previously published. Among these interactions, we find that the IgLONs, a family of five cell-surface neuronal proteins implicated in various human disorders, interact as homo- and heterodimers. We reveal their interaction patterns and report the dimeric crystal structures of Neurotrimin (NTRI), IgLON5, and the neuronal growth regulator 1 (NEGR1)/IgLON5 complex. We show that IgLONs maintain an extended conformation and that their dimerization occurs through the first Ig domain of each monomer and is Ca2+ independent. Cell aggregation shows that NTRI and NEGR1 homo- and heterodimerize in trans. Taken together, we report 89 unpublished cell-surface ligand-receptor pairs and describe structural models of trans interactions of IgLONs, showing that their structures are compatible with a model of interaction across the synaptic cleft.
Collapse
Affiliation(s)
| | - Liam S Turk
- Child Health Institute of New Jersey, New Brunswick, NJ 08901, USA
| | - Sinem Ozgul
- Child Health Institute of New Jersey, New Brunswick, NJ 08901, USA
| | - Sumie Kakehi
- Child Health Institute of New Jersey, New Brunswick, NJ 08901, USA
| | | | - Nicole Lopez
- Child Health Institute of New Jersey, New Brunswick, NJ 08901, USA
| | - Laura Trobiani
- Department of Biology and Biotechnology "Charles Darwin" and Pasteur Institute - Cenci Bolognetti Foundation, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Antonella De Jaco
- Department of Biology and Biotechnology "Charles Darwin" and Pasteur Institute - Cenci Bolognetti Foundation, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Natalia Denissova
- Department of Molecular Biology and Biochemistry and Center for Advanced Biotechnology and Medicine, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Borries Demeler
- Department of Chemistry and Biochemistry, University of Lethbridge, 4401 University Drive, Lethbridge, AB T1K 3M4, Canada
| | - Engin Özkan
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637, USA
| | - Gaetano T Montelione
- Department of Molecular Biology and Biochemistry and Center for Advanced Biotechnology and Medicine, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA; Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Davide Comoletti
- Child Health Institute of New Jersey, New Brunswick, NJ 08901, USA; Departments of Neuroscience and Cell Biology Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA; Department of Pediatrics, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA; School of Biological Sciences, Victoria University of Wellington, Wellington 6140, New Zealand.
| |
Collapse
|
25
|
Melrose J. Keratan sulfate (KS)-proteoglycans and neuronal regulation in health and disease: the importance of KS-glycodynamics and interactive capability with neuroregulatory ligands. J Neurochem 2019; 149:170-194. [PMID: 30578672 DOI: 10.1111/jnc.14652] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Revised: 11/26/2018] [Accepted: 12/13/2018] [Indexed: 12/18/2022]
Abstract
Compared to the other classes of glycosaminoglycans (GAGs), that is, chondroitin/dermatan sulfate, heparin/heparan sulfate and hyaluronan, keratan sulfate (KS), have the least known of its interactive properties. In the human body, the cornea and the brain are the two most abundant tissue sources of KS. Embryonic KS is synthesized as a linear poly-N-acetyllactosamine chain of d-galactose-GlcNAc repeat disaccharides which become progressively sulfated with development, sulfation of GlcNAc is more predominant than galactose. KS contains multi-sulfated high-charge density, monosulfated and non-sulfated poly-N-acetyllactosamine regions and thus is a heterogeneous molecule in terms of chain length and charge distribution. A recent proteomics study on corneal KS demonstrated its interactivity with members of the Slit-Robbo and Ephrin-Ephrin receptor families and proteins which regulate Rho GTPase signaling and actin polymerization/depolymerization in neural development and differentiation. KS decorates a number of peripheral nervous system/CNS proteoglycan (PG) core proteins. The astrocyte KS-PG abakan defines functional margins of the brain and is up-regulated following trauma. The chondroitin sulfate/KS PG aggrecan forms perineuronal nets which are dynamic neuroprotective structures with anti-oxidant properties and roles in neural differentiation, development and synaptic plasticity. Brain phosphacan a chondroitin sulfate, KS, HNK-1 PG have roles in neural development and repair. The intracellular microtubule and synaptic vesicle KS-PGs MAP1B and SV2 have roles in metabolite transport, storage, and export of neurotransmitters and cytoskeletal assembly. MAP1B has binding sites for tubulin and actin through which it promotes cytoskeletal development in growth cones and is highly expressed during neurite extension. The interactive capability of KS with neuroregulatory ligands indicate varied roles for KS-PGs in development and regenerative neural processes.
Collapse
Affiliation(s)
- James Melrose
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, St. Leonards, New South Wales, Australia.,Graduate School of Biomedical Engineering, University of New South Wales, Sydney, New South Wales, Australia.,Sydney Medical School, Northern Campus, Royal North Shore Hospital, The University of Sydney, New South Wales, Australia.,Faculty of Medicine and Health, Royal North Shore Hospital, The University of Sydney, St. Leonards, New South Wales, Australia
| |
Collapse
|
26
|
Singh K, Lilleväli K, Gilbert SF, Bregin A, Narvik J, Jayaram M, Rahi M, Innos J, Kaasik A, Vasar E, Philips MA. The combined impact of IgLON family proteins Lsamp and Neurotrimin on developing neurons and behavioral profiles in mouse. Brain Res Bull 2018; 140:5-18. [PMID: 29605488 DOI: 10.1016/j.brainresbull.2018.03.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 02/26/2018] [Accepted: 03/23/2018] [Indexed: 12/13/2022]
Abstract
Cell surface neural adhesion proteins are critical components in the complex orchestration of cell proliferation, apoptosis, and neuritogenesis essential for proper brain construction and behavior. We focused on the impact of two plasticity-associated IgLON family neural adhesion molecules, Neurotrimin (Ntm) and Limbic system associated membrane protein (Lsamp), on mouse behavior and its underlying neural development. Phenotyping neurons derived from the hippocampi of Lsamp-/-, Ntm-/- and Lsamp-/-Ntm-/- mice was performed in parallel with behavioral testing. While the anatomy of mutant brains revealed no gross changes, the Ntm-/- hippocampal neurons exhibited premature sprouting of neurites and manifested accelerated neurite elongation and branching. We propose that Ntm exerts an inhibitory impact on neurite outgrowth, whereas Lsamp appears to be an enhancer of the said process as premature neuritogenesis in Ntm-/- neurons is apparent only in the presence of Lsamp. We also show interplay between Lsamp and Ntm in regulating tissue homeostasis: the impact of Ntm on cellular proliferation was dependent on Lsamp, and Lsamp appeared to be a positive regulator of apoptosis in the presence of Ntm. Behavioral phenotyping indicated test-specific interactions between Lsamp and Ntm. The phenotypes of single mutant lines, such as reduced swimming speed in Morris water maze and increased activity in the elevated plus maze, were magnified in Lsamp-/-Ntm-/- mice. Altogether, evidence both from behavioral experiments and cultured hippocampal cells show combined and differential interactions between Ntm and Lsamp in the formation of hippocampal circuits and behavioral profiles. We demonstrate that mutual interactions between IgLON molecules regulate the initiation of neurite sprouting at very early ages, and even cell-autonomously, independent of their regulation of cell-cell adhesion.
Collapse
Affiliation(s)
- Katyayani Singh
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia; Centre of Excellence in Genomics and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia
| | - Kersti Lilleväli
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia; Centre of Excellence in Genomics and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia
| | - Scott F Gilbert
- Department of Biology, Swarthmore College, Swarthmore, PA, USA
| | - Aleksandr Bregin
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia; Centre of Excellence in Genomics and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia
| | - Jane Narvik
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia; Centre of Excellence in Genomics and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia
| | - Mohan Jayaram
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia; Centre of Excellence in Genomics and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia
| | - Märt Rahi
- Institute of Agricultural and Environmental Sciences, Estonian University of Life Sciences, Fr.R. Kreutzwaldi 5, 51014, Tartu, Estonia
| | - Jürgen Innos
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia; Centre of Excellence in Genomics and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia
| | - Allen Kaasik
- Department of Pharmacology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia
| | - Eero Vasar
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia; Centre of Excellence in Genomics and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia
| | - Mari-Anne Philips
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia; Centre of Excellence in Genomics and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia.
| |
Collapse
|