1
|
Lesch KP, Gorbunov N. Antisocial personality disorder:Failure to balance excitation/inhibition? Neuropharmacology 2025; 268:110321. [PMID: 39855295 DOI: 10.1016/j.neuropharm.2025.110321] [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: 08/17/2024] [Revised: 01/17/2025] [Accepted: 01/20/2025] [Indexed: 01/27/2025]
Abstract
While healthy brain function relies on a dynamic but tightly regulated interaction between excitation (E) and inhibition (I), a spectrum of social cognition disorders, including antisocial behavior and antisocial personality disorder (ASPD), frequently ensuing from irregular neurodevelopment, may be associated with E/I imbalance and concomitant alterations in neural connectivity. Technological advances in the evaluation of structural and functional E/I balance proxies in clinical settings and in human cell culture models provide a general basis for identification of biomarkers providing a powerful concept for prevention and intervention across different dimensions of mental health and disease. In this perspective we outline a framework for research to characterize neurodevelopmental pathways to antisocial behavior and ASPD driven by (epi)genetic factors across life, and to identify molecular targets for preventing the detrimental effects of cognitive dysfunction and maladaptive social behavior, considering psychosocial experience; to validate signatures of E/I imbalance and altered myelination proxies as biomarkers of pathogenic neural circuitry mechanisms to determine etiological processes in the transition from mental health to antisocial behavior and ASPD and in the switch from prevention to treatment; to develop a neurobiologically-grounded integrative model of antisocial behavior and ASPD resultant of disrupted E/I balance, allowing to establish objective diagnoses and monitoring tools, to personalize prevention and therapeutic decisions, to predict treatment response, and thus counteract relapse; and finally, to promote transformation of dimensional disorder taxonomy and to enhance societal awareness and reception of the neurobiological basis of antisocial behavior and ASPD.
Collapse
Affiliation(s)
- Klaus-Peter Lesch
- Division of Molecular Psychiatry, Center of Mental Health, University Hospital Würzburg, Würzburg, Germany; Department of Child- and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital Würzburg, Würzburg, Germany; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands.
| | - Nikita Gorbunov
- Division of Molecular Psychiatry, Center of Mental Health, University Hospital Würzburg, Würzburg, Germany; Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
2
|
Parmar P, Spahic H, Lechner C, St Pierre M, Carlin K, Nugent M, Chavez-Valdez R. Neonatal hypoxia-ischemia alters the events governing the hippocampal critical period of postnatal synaptic plasticity leading to deficits in working memory in mice. Neurobiol Dis 2024; 202:106722. [PMID: 39486775 PMCID: PMC11646096 DOI: 10.1016/j.nbd.2024.106722] [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: 05/16/2024] [Revised: 10/04/2024] [Accepted: 10/29/2024] [Indexed: 11/04/2024] Open
Abstract
Postnatal critical periods of synaptic plasticity (CPsp) are characterized by profound neural network refinement, which is shaped by synaptic activity and sculpted by maturation of the GABAergic network. Even after therapeutic hypothermia (TH), neonatal hypoxia-ischemia (HI) impairs two triggers for the initiation of the CPsp in the hippocampus: i) PSA-NCAM developmental decline and ii) parvalbumin (PV) + interneuron (IN) maturation. Thus, we investigated whether neonatal HI despite TH disturbs other events governing the onset, consolidation and closure of the postnatal CPsp in the hippocampus. We induced cerebral HI in P10 C57BL6 mice with right carotid ligation and 45 m of hypoxia (FiO2 = 0.08), followed by normothermia (36 °C, NT) or TH (31 °C) for 4 h with anesthesia-exposed shams as controls. ELISA, immunoblotting and immunohistochemistry were performed at 24 h (P11), 5 days (P15), 8 days (P18) and 30 days (P40) after HI injury. We specifically assessed: i) BDNF levels and TrkB activation, controlling the CPsp, ii) Otx2 and NPTX2 immunoreactivity (IR), engaging CPsp onset and iii) NogoR1, Lynx1 IR, PNN formation and myelination (MBP) mediating CPsp closure. Pups aged to P40 also received a battery of tests assessing working memory. Here, we documented deficits in hippocampal BDNF levels and TrkB activation at P18 in response to neonatal HI even with TH. Neonatal HI impaired in the CA1 the developmental increase in PV, Otx2, and NPTX2 between P11 and P18, the colocalization of Otx2 and PV at P18 and P40, the accumulation of NPTX2 in PV+ dendrites at P18 and P40, and the expression of NogoR at P40. Furthermore, neonatal HI decreased BDNF and impaired PNN development and myelination (MBP) at P40. Most of these abnormalities were insensitive to TH and correlated with memory deficits. Neonatal HI appears to disrupt many of the molecular and structural events initiating and consolidating the postnatal hippocampal CPsp, perhaps due to the early and delayed deficits in TrkB activation leading to memory deficits.
Collapse
Affiliation(s)
- Pritika Parmar
- Department of Neuroscience, The Zanvyl Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Harisa Spahic
- Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Charles Lechner
- Department of Neuroscience, The Zanvyl Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Mark St Pierre
- Department of Neuroscience, The Zanvyl Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD, USA
| | | | - Michael Nugent
- Department of Neuroscience, The Zanvyl Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Raul Chavez-Valdez
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD, USA; Neuroscience Intensive Care Nursery Program, Johns Hopkins University- School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
3
|
Pinky PD, Bloemer J, Smith WD, Du Y, Heslin RT, Setti SE, Pfitzer JC, Chowdhury K, Hong H, Bhattacharya S, Dhanasekaran M, Dityatev A, Reed MN, Suppiramaniam V. Prenatal Cannabinoid Exposure Elicits Memory Deficits Associated with Reduced PSA-NCAM Expression, Altered Glutamatergic Signaling, and Adaptations in Hippocampal Synaptic Plasticity. Cells 2023; 12:2525. [PMID: 37947603 PMCID: PMC10648717 DOI: 10.3390/cells12212525] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 10/02/2023] [Accepted: 10/06/2023] [Indexed: 11/12/2023] Open
Abstract
Cannabis is now one of the most commonly used illicit substances among pregnant women. This is particularly concerning since developmental exposure to cannabinoids can elicit enduring neurofunctional and cognitive alterations. This study investigates the mechanisms of learning and memory deficits resulting from prenatal cannabinoid exposure (PCE) in adolescent offspring. The synthetic cannabinoid agonist WIN55,212-2 was administered to pregnant rats, and a series of behavioral, electrophysiological, and immunochemical studies were performed to identify potential mechanisms of memory deficits in the adolescent offspring. Hippocampal-dependent memory deficits in adolescent PCE animals were associated with decreased long-term potentiation (LTP) and enhanced long-term depression (LTD) at hippocampal Schaffer collateral-CA1 synapses, as well as an imbalance between GluN2A- and GluN2B-mediated signaling. Moreover, PCE reduced gene and protein expression of neural cell adhesion molecule (NCAM) and polysialylated-NCAM (PSA-NCAM), which are critical for GluN2A and GluN2B signaling balance. Administration of exogenous PSA abrogated the LTP deficits observed in PCE animals, suggesting PSA mediated alterations in GluN2A- and GluN2B- signaling pathways may be responsible for the impaired hippocampal synaptic plasticity resulting from PCE. These findings enhance our current understanding of how PCE affects memory and how this process can be manipulated for future therapeutic purposes.
Collapse
Affiliation(s)
- Priyanka D. Pinky
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA 92697, USA
| | - Jenna Bloemer
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA
- Department of Pharmaceutical and Biomedical Sciences, Touro College of Pharmacy, New York, NY 10036, USA
| | - Warren D. Smith
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA
| | - Yifeng Du
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA
| | - Ryan T. Heslin
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA
| | - Sharay E. Setti
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA
| | - Jeremiah C. Pfitzer
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA
| | - Kawsar Chowdhury
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA
| | - Hao Hong
- Key Laboratory of Neuropsychiatric Diseases, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Subhrajit Bhattacharya
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA
- Center for Neuroscience Initiative, Auburn University, Auburn, AL 36849, USA
- Keck Graduate Institute, School of Pharmacy and Health Sciences, Claremont Colleges, Claremont, CA 91711, USA
| | - Muralikrishnan Dhanasekaran
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), 37075 Magdeburg, Germany
| | - Alexander Dityatev
- Center for Neuroscience Initiative, Auburn University, Auburn, AL 36849, USA
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), 37075 Magdeburg, Germany
- Medical Faculty, Otto-von-Guericke University, 39106 Magdeburg, Germany
| | - Miranda N. Reed
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA
- Center for Neuroscience Initiative, Auburn University, Auburn, AL 36849, USA
| | - Vishnu Suppiramaniam
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA
- Center for Neuroscience Initiative, Auburn University, Auburn, AL 36849, USA
| |
Collapse
|
4
|
Andres-Alonso M, Borgmeyer M, Mirzapourdelavar H, Lormann J, Klein K, Schweizer M, Hoffmeister-Ullerich S, Oelschlegel AM, Dityatev A, Kreutz MR. Golgi satellites are essential for polysialylation of NCAM and expression of LTP at distal synapses. Cell Rep 2023; 42:112692. [PMID: 37355986 DOI: 10.1016/j.celrep.2023.112692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 04/28/2023] [Accepted: 06/08/2023] [Indexed: 06/27/2023] Open
Abstract
The complex cytoarchitecture of neurons poses significant challenges for the maturation of synaptic membrane proteins. It is currently unclear whether locally secreted synaptic proteins bypass the Golgi or whether they traffic through Golgi satellites (GSs). Here, we create a transgenic GS reporter mouse line and show that GSs are widely distributed along dendrites and are capable of mature glycosylation, in particular sialylation. We find that polysialylation of locally secreted NCAM takes place at GSs. Accordingly, in mice lacking a component of trans-Golgi network-to-plasma membrane trafficking, we find fewer GSs and significantly reduced PSA-NCAM levels in distal dendrites of CA1 neurons that receive input from the temporoammonic pathway. Induction of long-term potentiation at those, but not more proximal, synapses is severely impaired. We conclude that GSs serve the need for local mature glycosylation of synaptic membrane proteins in distal dendrites and thereby contribute to rapid changes in synaptic strength.
Collapse
Affiliation(s)
- Maria Andres-Alonso
- Leibniz Group "Dendritic Organelles and Synaptic Function," Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; RG Neuroplasticity, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany.
| | - Maximilian Borgmeyer
- Leibniz Group "Dendritic Organelles and Synaptic Function," Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; RG Neuroplasticity, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany
| | | | - Jakob Lormann
- Leibniz Group "Dendritic Organelles and Synaptic Function," Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; RG Neuroplasticity, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany
| | - Kim Klein
- Leibniz Group "Dendritic Organelles and Synaptic Function," Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Michaela Schweizer
- Core Facility Morphology und Electron Microscopy, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Sabine Hoffmeister-Ullerich
- Core Facility Bioanalytik, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Anja M Oelschlegel
- RG Neuroplasticity, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany
| | - Alexander Dityatev
- German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany; Center for Behavioral Brain Sciences, Otto von Guericke University, 39120 Magdeburg, Germany; Medical Faculty, Otto von Guericke University, 39120 Magdeburg, Germany
| | - Michael R Kreutz
- Leibniz Group "Dendritic Organelles and Synaptic Function," Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; RG Neuroplasticity, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany; German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany; Center for Behavioral Brain Sciences, Otto von Guericke University, 39120 Magdeburg, Germany.
| |
Collapse
|
5
|
Varbanov H, Jia S, Kochlamazashvili G, Bhattacharya S, Buabeid MA, El Tabbal M, Hayani H, Stoyanov S, Sun W, Thiesler H, Röckle I, Hildebrandt H, Senkov O, Suppiramaniam V, Gerardy-Schahn R, Dityatev A. Rescue of synaptic and cognitive functions in polysialic acid-deficient mice and dementia models by short polysialic acid fragments. Neurobiol Dis 2023; 180:106079. [PMID: 36918046 DOI: 10.1016/j.nbd.2023.106079] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 03/04/2023] [Accepted: 03/08/2023] [Indexed: 03/14/2023] Open
Abstract
Dysregulated cortical expression of the neural cell adhesion molecule (NCAM) and deficits of its associated polysialic acid (polySia) have been found in Alzheimer's disease and schizophrenia. However, the functional role of polySia in cortical synaptic plasticity remains poorly understood. Here, we show that acute enzymatic removal of polySia in medial prefrontal cortex (mPFC) slices leads to increased transmission mediated by the GluN1/GluN2B subtype of N-methyl-d-aspartate receptors (NMDARs), increased NMDAR-mediated extrasynaptic tonic currents, and impaired long-term potentiation (LTP). The latter could be fully rescued by pharmacological suppression of GluN1/GluN2B receptors, or by application of short soluble polySia fragments that inhibited opening of GluN1/GluN2B channels. These treatments and augmentation of synaptic NMDARs with the glycine transporter type 1 (GlyT1) inhibitor sarcosine also restored LTP in mice deficient in polysialyltransferase ST8SIA4. Furthermore, the impaired performance of polySia-deficient mice and two models of Alzheimer's disease in the mPFC-dependent cognitive tasks could be rescued by intranasal administration of polySia fragments. Our data demonstrate the essential role of polySia-NCAM in the balancing of signaling through synaptic/extrasynaptic NMDARs in mPFC and highlight the therapeutic potential of short polySia fragments to restrain GluN1/GluN2B-mediated signaling.
Collapse
Affiliation(s)
- Hristo Varbanov
- German Center for Neurodegenerative Diseases (DZNE), Leipziger Str. 44, 39120 Magdeburg, Germany; Institute of Neurophysiology, Hannover Medical School, OE 4230, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Shaobo Jia
- German Center for Neurodegenerative Diseases (DZNE), Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Gaga Kochlamazashvili
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, 16163 Genova, Italy; Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Roessle-Straße 10, 13125 Berlin, Germany
| | - Subhrajit Bhattacharya
- School of Pharmaceutical and Health Sciences, Keck Graduate Institute, Claremont Colleges, Claremont, CA 91711, USA
| | - Manal Ali Buabeid
- Department of Drug Discovery and Development, School of Pharmacy, Auburn University, Auburn, AL 36849, USA
| | - Mohamed El Tabbal
- German Center for Neurodegenerative Diseases (DZNE), Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Hussam Hayani
- German Center for Neurodegenerative Diseases (DZNE), Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Stoyan Stoyanov
- German Center for Neurodegenerative Diseases (DZNE), Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Weilun Sun
- German Center for Neurodegenerative Diseases (DZNE), Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Hauke Thiesler
- Institute for Clinical Biochemistry, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Iris Röckle
- Institute for Clinical Biochemistry, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Herbert Hildebrandt
- Institute for Clinical Biochemistry, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany; Center for Systems Neuroscience Hannover (ZSN), Bünteweg 2, 30559 Hannover, Germany
| | - Oleg Senkov
- German Center for Neurodegenerative Diseases (DZNE), Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Vishnu Suppiramaniam
- Department of Drug Discovery and Development, School of Pharmacy, Auburn University, Auburn, AL 36849, USA; College of Science and Mathematics, Kennesaw State University, GA 30144, USA
| | - Rita Gerardy-Schahn
- Institute for Clinical Biochemistry, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Alexander Dityatev
- German Center for Neurodegenerative Diseases (DZNE), Leipziger Str. 44, 39120 Magdeburg, Germany; Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, 16163 Genova, Italy; Medical Faculty, Otto-von-Guericke-University, Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany; Center for Behavioral Brain Sciences (CBBS), Otto von Guericke University Magdeburg, Universitätsplatz 2, 39106 Magdeburg, Germany.
| |
Collapse
|
6
|
Highet B, Wiseman JA, Mein H, Parker R, Ryan B, Turner CP, Jing Y, Singh-Bains MK, Liu P, Dragunow M, Faull RLM, Murray HC, Curtis MA. PSA-NCAM Regulatory Gene Expression Changes in the Alzheimer's Disease Entorhinal Cortex Revealed with Multiplexed in situ Hybridization. J Alzheimers Dis 2023; 92:371-390. [PMID: 36744342 DOI: 10.3233/jad-220986] [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: 02/05/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is the most common form of dementia and is characterized by a substantial reduction of neuroplasticity. Our previous work demonstrated that neurons involved in memory function may lose plasticity because of decreased protein levels of polysialylated neural cell adhesion molecule (PSA-NCAM) in the entorhinal cortex (EC) of the human AD brain, but the cause of this decrease is unclear. OBJECTIVE To investigate genes involved in PSA-NCAM regulation which may underlie its decrease in the AD EC. METHODS We subjected neurologically normal and AD human EC sections to multiplexed fluorescent in situ hybridization and immunohistochemistry to investigate genes involved in PSA-NCAM regulation. Gene expression changes were sought to be validated in both human tissue and a mouse model of AD. RESULTS In the AD EC, a cell population expressing a high level of CALB2 mRNA and a cell population expressing a high level of PST mRNA were both decreased. CALB2 mRNA and protein were not decreased globally, indicating that the decrease in CALB2 was specific to a sub-population of cells. A significant decrease in PST mRNA expression was observed with single-plex in situ hybridization in middle temporal gyrus tissue microarray cores from AD patients, which negatively correlated with tau pathology, hinting at global loss in PST expression across the AD brain. No significant differences in PSA-NCAM or PST protein expression were observed in the MAPT P301S mouse brain at 9 months of age. CONCLUSION We conclude that PSA-NCAM dysregulation may cause subsequent loss of structural plasticity in AD, and this may result from a loss of PST mRNA expression. Due PSTs involvement in structural plasticity, intervention for AD may be possible by targeting this disrupted plasticity pathway.
Collapse
Affiliation(s)
- Blake Highet
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag, Auckland, New Zealand
| | - James A Wiseman
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag, Auckland, New Zealand
| | - Hannah Mein
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Remai Parker
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag, Auckland, New Zealand
| | - Brigid Ryan
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag, Auckland, New Zealand
| | - Clinton P Turner
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag, Auckland, New Zealand.,Department of Anatomical Pathology, LabPlus, Auckland City Hospital, New Zealand
| | - Yu Jing
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Malvindar K Singh-Bains
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag, Auckland, New Zealand
| | - Ping Liu
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Mike Dragunow
- Department of Pharmacology and Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag, Auckland, New Zealand
| | - Richard L M Faull
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag, Auckland, New Zealand
| | - Helen C Murray
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag, Auckland, New Zealand
| | - Maurice A Curtis
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag, Auckland, New Zealand
| |
Collapse
|
7
|
Fernández-Suárez D, Krapacher FA, Pietrajtis K, Andersson A, Kisiswa L, Carrier-Ruiz A, Diana MA, Ibáñez CF. Adult medial habenula neurons require GDNF receptor GFRα1 for synaptic stability and function. PLoS Biol 2021; 19:e3001350. [PMID: 34748545 PMCID: PMC8601618 DOI: 10.1371/journal.pbio.3001350] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 11/18/2021] [Accepted: 10/05/2021] [Indexed: 11/18/2022] Open
Abstract
The medial habenula (mHb) is an understudied small brain nucleus linking forebrain and midbrain structures controlling anxiety and fear behaviors. The mechanisms that maintain the structural and functional integrity of mHb neurons and their synapses remain unknown. Using spatiotemporally controlled Cre-mediated recombination in adult mice, we found that the glial cell-derived neurotrophic factor receptor alpha 1 (GFRα1) is required in adult mHb neurons for synaptic stability and function. mHb neurons express some of the highest levels of GFRα1 in the mouse brain, and acute ablation of GFRα1 results in loss of septohabenular and habenulointerpeduncular glutamatergic synapses, with the remaining synapses displaying reduced numbers of presynaptic vesicles. Chemo- and optogenetic studies in mice lacking GFRα1 revealed impaired circuit connectivity, reduced AMPA receptor postsynaptic currents, and abnormally low rectification index (R.I.) of AMPARs, suggesting reduced Ca2+ permeability. Further biochemical and proximity ligation assay (PLA) studies defined the presence of GluA1/GluA2 (Ca2+ impermeable) as well as GluA1/GluA4 (Ca2+ permeable) AMPAR complexes in mHb neurons, as well as clear differences in the levels and association of AMPAR subunits with mHb neurons lacking GFRα1. Finally, acute loss of GFRα1 in adult mHb neurons reduced anxiety-like behavior and potentiated context-based fear responses, phenocopying the effects of lesions to septal projections to the mHb. These results uncover an unexpected function for GFRα1 in the maintenance and function of adult glutamatergic synapses and reveal a potential new mechanism for regulating synaptic plasticity in the septohabenulointerpeduncular pathway and attuning of anxiety and fear behaviors.
Collapse
Affiliation(s)
- Diana Fernández-Suárez
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
- Department of Physiology and Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | | | - Katarzyna Pietrajtis
- Sorbonne Université, CNRS, INSERM, Neurosciences Paris Seine–Institut de Biologie Paris Seine (NPS-IBPS), Paris, France
| | - Annika Andersson
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Lilian Kisiswa
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | | | - Marco A. Diana
- Sorbonne Université, CNRS, INSERM, Neurosciences Paris Seine–Institut de Biologie Paris Seine (NPS-IBPS), Paris, France
| | - Carlos F. Ibáñez
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
- Department of Physiology and Life Sciences Institute, National University of Singapore, Singapore, Singapore
- Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University School of Life Sciences and Chinese Institute for Brain Research, Beijing, China
| |
Collapse
|
8
|
Schmitt-Ulms G, Mehrabian M, Williams D, Ehsani S. The IDIP framework for assessing protein function and its application to the prion protein. Biol Rev Camb Philos Soc 2021; 96:1907-1932. [PMID: 33960099 DOI: 10.1111/brv.12731] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 04/22/2021] [Accepted: 04/26/2021] [Indexed: 01/06/2023]
Abstract
The quest to determine the function of a protein can represent a profound challenge. Although this task is the mandate of countless research groups, a general framework for how it can be approached is conspicuously lacking. Moreover, even expectations for when the function of a protein can be considered to be 'known' are not well defined. In this review, we begin by introducing concepts pertinent to the challenge of protein function assignments. We then propose a framework for inferring a protein's function from four data categories: 'inheritance', 'distribution', 'interactions' and 'phenotypes' (IDIP). We document that the functions of proteins emerge at the intersection of inferences drawn from these data categories and emphasise the benefit of considering them in an evolutionary context. We then apply this approach to the cellular prion protein (PrPC ), well known for its central role in prion diseases, whose function continues to be considered elusive by many investigators. We document that available data converge on the conclusion that the function of the prion protein is to control a critical post-translational modification of the neural cell adhesion molecule in the context of epithelial-to-mesenchymal transition and related plasticity programmes. Finally, we argue that this proposed function of PrPC has already passed the test of time and is concordant with the IDIP framework in a way that other functions considered for this protein fail to achieve. We anticipate that the IDIP framework and the concepts analysed herein will aid the investigation of other proteins whose primary functional assignments have thus far been intractable.
Collapse
Affiliation(s)
- Gerold Schmitt-Ulms
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, M5T 0S8, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | | | - Declan Williams
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, M5T 0S8, Canada
| | - Sepehr Ehsani
- Theoretical and Philosophical Biology, Department of Philosophy, University College London, Bloomsbury, London, WC1E 6BT, U.K.,Ronin Institute for Independent Scholarship, Montclair, NJ, 07043, U.S.A
| |
Collapse
|
9
|
Chavez-Valdez R, Lechner C, Emerson P, Northington FJ, Martin LJ. Accumulation of PSA-NCAM marks nascent neurodegeneration in the dorsal hippocampus after neonatal hypoxic-ischemic brain injury in mice. J Cereb Blood Flow Metab 2021; 41:1039-1057. [PMID: 32703109 PMCID: PMC8054724 DOI: 10.1177/0271678x20942707] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Neonatal hypoxia-ischemia (nHI) disrupts hippocampal GABAergic development leading to memory deficits in mice. Polysialic-acid neural-cell adhesion molecule (PSA-NCAM) developmentally declines to trigger GABAergic maturation. We hypothesized that nHI changes PSA-NCAM abundance and cellular distribution, impairing GABAergic development, and marking nascent neurodegeneration. Cell degeneration, atrophy, and PSA-NCAM immunoreactivity (IR) were measured in CA1 of nHI-injured C57BL6 mice related to: (i) cellular subtype markers; (ii) GAD65/67 and synatophysin (SYP), pre-synaptic markers; (iii) phospho-Ser396Tau, cytoskeletal marker; and (iv) GAP43, axonalregeneration marker. PSA-NCAM IR was minimal in CA1 of shams at P11. After nHI, PSA-NCAM IR was increased in injured pyramidal cells (PCs), minimal in parvalbumin (PV)+INs, and absent in glia. PSA-NCAM IR correlated with injury severity and became prominent in perikaryal cytoplasm at P18. GAD65/67 and SYP IRs only weakly related to PSA-NCAM after nHI. Injured phospho-Ser396Tau+ PCs and PV+INs variably co-expressed PSA-NCAM at P40. While PCs with cytoplasmic marginalized PSA-NCAM had increased perisomatic GAP43, those with perikaryal cytoplasmic PSA-NCAM had minimal GAP43. PSA-NCAM increased in serum of nHI-injured mice. Increased PSA-NCAM is likely a generic acute response to nHI brain injury. PSA-NCAM aberrant cellular localization may aggravate neuronal degeneration. The significance of PSA-NCAM as a biomarker of recovery from nHI and nascent neurodegeneration needs further study.
Collapse
Affiliation(s)
- Raul Chavez-Valdez
- Division of Neonatology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Charles Lechner
- Division of Neonatology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Paul Emerson
- Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA
| | - Frances J Northington
- Division of Neonatology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lee J Martin
- Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA.,Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
10
|
Yang Y, Murai R, Takahashi Y, Mori A, Hane M, Kitajima K, Sato C. Comparative Studies of Polysialic Acids Derived from Five Different Vertebrate Brains. Int J Mol Sci 2020; 21:ijms21228593. [PMID: 33202622 PMCID: PMC7696247 DOI: 10.3390/ijms21228593] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 10/30/2020] [Accepted: 11/09/2020] [Indexed: 02/06/2023] Open
Abstract
Polysialic acid (polySia/PSA) is a linear homopolymer of sialic acid (Sia) that primarily modifies the neural cell adhesion molecule (NCAM) in mammalian brains. PolySia-NCAM not only displays an anti-adhesive function due to the hydration effect, but also possesses a molecule-retaining function via a direct binding to neurologically active molecules. The quality and quantity of polySia determine the function of polySia-NCAM and are considered to be profoundly related to the maintenance of normal brain functions. In this study, to compare the structures of polySia-NCAM in brains of five different vertebrates (mammals, birds, reptiles, amphibians, and fish), we adopted newly developed combinational methods for the analyses. The results revealed that the structural features of polySia considerably varied among different species. Interestingly, mice, as a mammal, possess eminently distinct types of polySia, in both quality and quantity, compared with those possessed by other animals. Thus, the mouse polySia is of larger quantities, of longer and more diverse chain lengths, and of a larger molecular size with higher negative charge, compared with polySia of other species. These properties might enable more advanced brain function. Additionally, it is suggested that the polySia/Sia ratio, which likely reflects the complexity of brain function, can be used as a new promising index to evaluate the intelligence of different vertebrate brains.
Collapse
Affiliation(s)
- Yi Yang
- Bioscience and Biotechnology Center, Nagoya University, Chikusa, Nagoya 464-8601, Japan; (Y.Y.); (R.M.); (Y.T.); (A.M.); (M.H.); (K.K.)
- Graduate School of Bioagricultural Science, Nagoya University, Chikusa, Nagoya 464-8601, Japan
| | - Ryo Murai
- Bioscience and Biotechnology Center, Nagoya University, Chikusa, Nagoya 464-8601, Japan; (Y.Y.); (R.M.); (Y.T.); (A.M.); (M.H.); (K.K.)
- Graduate School of Bioagricultural Science, Nagoya University, Chikusa, Nagoya 464-8601, Japan
| | - Yuka Takahashi
- Bioscience and Biotechnology Center, Nagoya University, Chikusa, Nagoya 464-8601, Japan; (Y.Y.); (R.M.); (Y.T.); (A.M.); (M.H.); (K.K.)
- Graduate School of Bioagricultural Science, Nagoya University, Chikusa, Nagoya 464-8601, Japan
| | - Airi Mori
- Bioscience and Biotechnology Center, Nagoya University, Chikusa, Nagoya 464-8601, Japan; (Y.Y.); (R.M.); (Y.T.); (A.M.); (M.H.); (K.K.)
- Graduate School of Bioagricultural Science, Nagoya University, Chikusa, Nagoya 464-8601, Japan
| | - Masaya Hane
- Bioscience and Biotechnology Center, Nagoya University, Chikusa, Nagoya 464-8601, Japan; (Y.Y.); (R.M.); (Y.T.); (A.M.); (M.H.); (K.K.)
- Graduate School of Bioagricultural Science, Nagoya University, Chikusa, Nagoya 464-8601, Japan
- Institute for Glyco-core Research (iGCORE), Nagoya University, Chikusa, Nagoya 464-8601, Japan
| | - Ken Kitajima
- Bioscience and Biotechnology Center, Nagoya University, Chikusa, Nagoya 464-8601, Japan; (Y.Y.); (R.M.); (Y.T.); (A.M.); (M.H.); (K.K.)
- Graduate School of Bioagricultural Science, Nagoya University, Chikusa, Nagoya 464-8601, Japan
- Institute for Glyco-core Research (iGCORE), Nagoya University, Chikusa, Nagoya 464-8601, Japan
| | - Chihiro Sato
- Bioscience and Biotechnology Center, Nagoya University, Chikusa, Nagoya 464-8601, Japan; (Y.Y.); (R.M.); (Y.T.); (A.M.); (M.H.); (K.K.)
- Graduate School of Bioagricultural Science, Nagoya University, Chikusa, Nagoya 464-8601, Japan
- Institute for Glyco-core Research (iGCORE), Nagoya University, Chikusa, Nagoya 464-8601, Japan
- Correspondence: ; Tel.: +81-52-789-4129
| |
Collapse
|
11
|
Puigdellívol M, Allendorf DH, Brown GC. Sialylation and Galectin-3 in Microglia-Mediated Neuroinflammation and Neurodegeneration. Front Cell Neurosci 2020; 14:162. [PMID: 32581723 PMCID: PMC7296093 DOI: 10.3389/fncel.2020.00162] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 05/15/2020] [Indexed: 12/31/2022] Open
Abstract
Microglia are brain macrophages that mediate neuroinflammation and contribute to and protect against neurodegeneration. The terminal sugar residue of all glycoproteins and glycolipids on the surface of mammalian cells is normally sialic acid, and addition of this negatively charged residue is known as “sialylation,” whereas removal by sialidases is known as “desialylation.” High sialylation of the neuronal cell surface inhibits microglial phagocytosis of such neurons, via: (i) activating sialic acid receptors (Siglecs) on microglia that inhibit phagocytosis and (ii) inhibiting binding of opsonins C1q, C3, and galectin-3. Microglial sialylation inhibits inflammatory activation of microglia via: (i) activating Siglec receptors CD22 and CD33 on microglia that inhibit phagocytosis and (ii) inhibiting Toll-like receptor 4 (TLR4), complement receptor 3 (CR3), and other microglial receptors. When activated, microglia release a sialidase activity that desialylates both microglia and neurons, activating the microglia and rendering the neurons susceptible to phagocytosis. Activated microglia also release galectin-3 (Gal-3), which: (i) further activates microglia via binding to TLR4 and TREM2, (ii) binds to desialylated neurons opsonizing them for phagocytosis via Mer tyrosine kinase, and (iii) promotes Aβ aggregation and toxicity in vivo. Gal-3 and desialylation may increase in a variety of brain pathologies. Thus, Gal-3 and sialidases are potential treatment targets to prevent neuroinflammation and neurodegeneration.
Collapse
Affiliation(s)
- Mar Puigdellívol
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - David H Allendorf
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Guy C Brown
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
12
|
Myrtus communis subsp. communis improved cognitive functions in ovariectomized diabetic rats. Gene 2020; 744:144616. [PMID: 32222531 DOI: 10.1016/j.gene.2020.144616] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 02/25/2020] [Accepted: 03/23/2020] [Indexed: 12/15/2022]
Abstract
AIM The purpose of this study was to investigate the possible effects of Myrtus communis subsp. communis (MC) on cognitive impairment in ovariectomized diabetic rats. MATERIAL AND METHOD Female Sprague-Dawley rats were divided into 5 groups consisting of 15 rats each; Control (C), Diabetes (D), Ovariectomy and diabetes (OVX + D), Ovariectomy, diabetes and donepezil (OVX + D + Don), Ovariectomy, diabetes and Myrtus communis subsp. communis (OVX + D + MC). Blood glucose measurements were made at the beginning and end of the experiments. The animals underwent the novel object recognition test (NORT) and their performance was evaluated. In hippocampal tissues; amyloid beta (Aβ) and neprilysin levels, acetylcholinesterase (AChE), and choline acetyltransferase (ChAT) activities, polysialylated neural cell adhesion molecule (PSA-NCAM), α7 subunit of neuronal nicotinic acetylcholine receptor (α7-nAChR) and brain derived neurotrophic factor (BDNF) gene expressions were examined. RESULTS Animals with ovariectomy and diabetes showed increased levels of blood glucose, AChE activity and Aβ levels, and decreased neprilysin levels, ChAT activity, α7-nAChR, PSA-NCAM and BDNF gene expressions in parallel with a decrease in NORT performance score. On the other hand, in the MC-treated OVX + D group, there was a significant decrease observed in blood glucose levels and AChE activities while there was improvement in NORT performances and an increase in hippocampal ChAT activity, neprilysin levels, α7-nAChR, PSA-NCAM and BDNF expressions. CONCLUSION These results suggest that MC extract could improve cognitive and neuronal functions with its anticholinesterase and antihyperglycemic properties.
Collapse
|
13
|
Wang C, Inselman A, Liu S, Liu F. Potential mechanisms for phencyclidine/ketamine-induced brain structural alterations and behavioral consequences. Neurotoxicology 2019; 76:213-219. [PMID: 31812709 DOI: 10.1016/j.neuro.2019.12.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 11/24/2019] [Accepted: 12/04/2019] [Indexed: 01/30/2023]
Abstract
Evidence of structural abnormalities in the nervous system of recreational drug [e.g., phencyclidine (PCP) or ketamine] users and/or preclinical animal research models suggests interference with the activity of multiple neurotransmitters, particularly glutamate neurotransmission. The damage to the central nervous system (CNS) may include neuronal loss, synaptic changes, disturbed neural network formation and reduced projections to subcortical fields. Notably, the reduced projections may considerably compromise the establishment of the subcortical areas, such as the nucleus accumbens located in the basal forebrain. With its abundant dopaminergic innervation, the nucleus accumbens is believed to be directly associated with addictive behaviors and mental disorders. This review seeks to delineate the relationship between PCP/ketamine-induced loss of cortical neurons and the reduced level of polysialic acid neural cell adhesion molecule (PSA-NCAM) in the striatum, and the likely changes in striatal synaptogenesis during development. The basic mechanism of how PSA-NCAM cell surface expression may be regulated will also be discussed, as well as the hypothesis that PSA-NCAM activity is critical to the regulation of synaptic protein expression. Overall, the present review will address the general hypothesis that damage/interruption of cortico-striatal communication and subcortical synaptogenesis could underlie the erratic/sensitization or addictive states produced by chronic or prolonged PCP/ketamine usage.
Collapse
Affiliation(s)
- Cheng Wang
- Division of Neurotoxicology, National Center for Toxicological Research/U.S. Food & Drug Administration, Jefferson, AR, United States.
| | - Amy Inselman
- Division of Systems Biology, National Center for Toxicological Research/U.S. Food & Drug Administration, Jefferson, AR, United States
| | - Shuliang Liu
- Division of Neurotoxicology, National Center for Toxicological Research/U.S. Food & Drug Administration, Jefferson, AR, United States
| | - Fang Liu
- Division of Neurotoxicology, National Center for Toxicological Research/U.S. Food & Drug Administration, Jefferson, AR, United States.
| |
Collapse
|
14
|
Zhang XL, Xu FX, Han XY. siRNA-mediated NCAM1 gene silencing suppresses oxidative stress in pre-eclampsia by inhibiting the p38MAPK signaling pathway. J Cell Biochem 2019; 120:18608-18617. [PMID: 31353686 DOI: 10.1002/jcb.28778] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 03/21/2019] [Accepted: 04/01/2019] [Indexed: 02/03/2023]
Abstract
Pre-eclampsia (PE), whose pathophysiology and etiology remain undefined, represents a leading consequence of fetal and maternal mortality and morbidity. Oxidative stress (OS) is recognized to involve in this disorder. In this study, we hypothesized that neural cell adhesion molecule 1 (NCAM1) gene silencing would suppress the OS in the pregnancy complicated by PE. Initially, clinical samples were collected for determination of NCAM1 expression in placental tissues and levels of OS products in blood. To assess the regulatory mechanism of NCAM1 knockdown on OS, we used small interfering RNA (siRNA) to silence NCAM1 expression in human umbilical vein endothelial cells (HUVECs). Next, cells were treated with or without hypoxia/reoxygenation to observe the level changes of OS products and p38 mitogen-activated protein kinase (p38MAPK) pathway-related genes. Finally, an evaluation of HUVEC migration and invasion abilities was conducted by wound-healing and transwell assays. Placenta of pregnancy with PE presented significantly increased NCAM1 expression in comparison to placenta of normal pregnancy. Meanwhile, enhanced OS in blood of pregnant women with PE was observed relative to women with normal pregnancy. siRNA-mediated knockdown of NCAM1 gene could inhibit the p38MAPK signaling pathway, repress OS, and promote cell migration and invasion in HUVECs, indicating that NCAM1 inhibition could reduce the influence of PE. Importantly, blocking the p38MAPK signaling pathway reversed the inhibitory role of NCAM1 gene silencing on PE. Collectively, this study defines potential role of NCAM1 gene silencing as a therapeutic target in PE through inhibiting OS and enhancing HUVEC migration and invasion by disrupting the p38MAPK signaling pathway.
Collapse
Affiliation(s)
- Xiao-Lin Zhang
- Department of Obstetrics, Zoucheng Hospital District of Affiliated Hospital of Jining Medical University, Zoucheng, People's Republic of China
| | - Feng-Xian Xu
- Department of Obstetrics, Zoucheng Hospital District of Affiliated Hospital of Jining Medical University, Zoucheng, People's Republic of China
| | - Xiao-Yun Han
- Department of Obstetrics, Affiliated Hospital of Jining Medical University, Jining, People's Republic of China
| |
Collapse
|
15
|
Kim P, Scott MR, Meador-Woodruff JH. Abnormal ER quality control of neural GPI-anchored proteins via dysfunction in ER export processing in the frontal cortex of elderly subjects with schizophrenia. Transl Psychiatry 2019; 9:6. [PMID: 30664618 PMCID: PMC6341114 DOI: 10.1038/s41398-018-0359-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 10/09/2018] [Accepted: 12/09/2018] [Indexed: 01/22/2023] Open
Abstract
Abnormalities of posttranslational protein modifications (PTMs) have recently been implicated in the pathophysiology of schizophrenia. Glycosylphosphatidylinositols (GPIs) are a class of complex glycolipids, which anchor surface proteins and glycoproteins to the cell membrane. GPI attachment to proteins represents one of the most common PTMs and GPI-associated proteins (GPI-APs) facilitate many cell surface processes, including synapse development and maintenance. Mutations in the GPI processing pathway are associated with intellectual disability, emphasizing the potential role of GPI-APs in cognition and schizophrenia-associated cognitive dysfunction. As initial endoplasmic reticulum (ER)-associated protein processing is essential for GPI-AP function, we measured protein expression of molecules involved in attachment (GPAA1), modification (PGAP1), and ER export (Tmp21) of GPI-APs, in homogenates and in an ER enriched fraction derived from dorsolateral prefrontal cortex (DLPFC) of 15 matched pairs of schizophrenia and comparison subjects. In total homogenate we found a significant decrease in transmembrane protein 21 (Tmp21) and in the ER-enriched fraction we found reduced expression of post-GPI attachment protein (PGAP1). PGAP1 modifies GPI-anchors through inositol deacylation, allowing it to be recognized by Tmp21. Tmp21 is a component of the p24 complex that recognizes GPI-anchored proteins, senses the status of the GPI-anchor, and regulates incorporation into COPII vesicles for export to the Golgi apparatus. Together, these proteins are the molecular mechanisms underlying GPI-AP quality control and ER export. To investigate the potential consequences of a deficit in export and/or quality control, we measured cell membrane-associated expression of known GPI-APs that have been previously implicated in schizophrenia, including GPC1, NCAM, MDGA2, and EPHA1, using Triton X-114 phase separation. Additionally, we tested the sensitivity of those candidate proteins to phosphatidylinositol-specific phospholipase C (PI-PLC), an enzyme that cleaves GPI from GPI-APs. While we did not observe a difference in the amount of these GPI-APs in Triton X-114 phase separated membrane fractions, we found decreased NCAM and GPC1 within the PI-PLC sensitive fraction. These findings suggest dysregulation of ER-associated GPI-AP protein processing, with impacts on post-translational modifications of proteins previously implicated in schizophrenia such as NCAM and GPC1. These findings provide evidence for a deficit in ER protein processing pathways in this illness.
Collapse
Affiliation(s)
- Pitna Kim
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| | - Madeline R Scott
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - James H Meador-Woodruff
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| |
Collapse
|
16
|
Demina EP, Pierre WC, Nguyen ALA, Londono I, Reiz B, Zou C, Chakraberty R, Cairo CW, Pshezhetsky AV, Lodygensky GA. Persistent reduction in sialylation of cerebral glycoproteins following postnatal inflammatory exposure. J Neuroinflammation 2018; 15:336. [PMID: 30518374 PMCID: PMC6282350 DOI: 10.1186/s12974-018-1367-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 11/14/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The extension of sepsis encompassing the preterm newborn's brain is often overlooked due to technical challenges in this highly vulnerable population, yet it leads to substantial long-term neurodevelopmental disabilities. In this study, we demonstrate how neonatal neuroinflammation following postnatal E. coli lipopolysaccharide (LPS) exposure in rat pups results in persistent reduction in sialylation of cerebral glycoproteins. METHODS Male Sprague-Dawley rat pups at postnatal day 3 (P3) were injected in the corpus callosum with saline or LPS. Twenty-four hours (P4) or 21 days (P24) following injection, brains were extracted and analyzed for neuraminidase activity and expression as well as for sialylation of cerebral glycoproteins and glycolipids. RESULTS At both P4 and P24, we detected a significant increase of the acidic neuraminidase activity in LPS-exposed rats. It correlated with significantly increased neuraminidase 1 (Neu1) mRNA in LPS-treated brains at P4 and with neuraminidases 1 and 4 at P24 suggesting that these enzymes were responsible for the rise of neuraminidase activity. At both P4 and P24, sialylation of N-glycans on brain glycoproteins decreased according to both mass-spectrometry analysis and lectin blotting, but the ganglioside composition remained intact. Finally, at P24, analysis of brain tissues by immunohistochemistry showed that neurons in the upper layers (II-III) of somatosensory cortex had a reduced surface content of polysialic acid. CONCLUSIONS Together, our data demonstrate that neonatal LPS exposure results in specific and sustained induction of Neu1 and Neu4, causing long-lasting negative changes in sialylation of glycoproteins on brain cells. Considering the important roles played by sialoglycoproteins in CNS function, we speculate that observed re-programming of the brain sialome constitutes an important part of pathophysiological consequences in perinatal infectious exposure.
Collapse
Affiliation(s)
- Ekaterina P Demina
- Department of Paediatrics, Sainte-Justine Hospital Research Center, Université de Montréal, Montreal, H3T 1C5, QC, Canada
| | - Wyston C Pierre
- Department of Paediatrics, Sainte-Justine Hospital Research Center, Université de Montréal, Montreal, H3T 1C5, QC, Canada
| | - Annie L A Nguyen
- Department of Paediatrics, Sainte-Justine Hospital Research Center, Université de Montréal, Montreal, H3T 1C5, QC, Canada
| | - Irene Londono
- Department of Paediatrics, Sainte-Justine Hospital Research Center, Université de Montréal, Montreal, H3T 1C5, QC, Canada
| | - Bela Reiz
- Alberta Glycomics Centre and Department of Chemistry, University of Alberta, Edmonton, T6G 2G2, AB, Canada
| | - Chunxia Zou
- Alberta Glycomics Centre and Department of Chemistry, University of Alberta, Edmonton, T6G 2G2, AB, Canada
| | - Radhika Chakraberty
- Alberta Glycomics Centre and Department of Chemistry, University of Alberta, Edmonton, T6G 2G2, AB, Canada
| | - Christopher W Cairo
- Alberta Glycomics Centre and Department of Chemistry, University of Alberta, Edmonton, T6G 2G2, AB, Canada
| | - Alexey V Pshezhetsky
- Department of Paediatrics, Sainte-Justine Hospital Research Center, Université de Montréal, Montreal, H3T 1C5, QC, Canada. .,Department of Anatomy and Cell Biology, McGill University, Montreal, H3A0C7, QC, Canada. .,Centre de recherche, CHU Sainte-Justine, 3175 Côte-Sainte-Catherine, Montreal, QC, H3T 1C5, Canada.
| | - Gregory A Lodygensky
- Department of Paediatrics, Sainte-Justine Hospital Research Center, Université de Montréal, Montreal, H3T 1C5, QC, Canada. .,Department of Pharmacology and Physiology, Université de Montréal, Montreal, H3T 1J4, QC, Canada. .,Montreal Heart Institute, Montreal, H1T 1C8, QC, Canada. .,Centre de recherche, CHU Sainte-Justine, 3175 Côte-Sainte-Catherine, Montreal, QC, H3T 1C5, Canada.
| |
Collapse
|
17
|
Cirielli V, Cima L, Chindemi C, Danzi O, Ghimenton C, Eccher A, Mauriello S, Bortolotti F, De Leo D, Brunelli M, Tagliaro F. Cortical Expression of the Polysialylated Isoform of the Neural Cell Adhesion Molecule on Brain Tissue to Recognize Drug-Related Death: An Exploratory Analysis. Am J Forensic Med Pathol 2018; 39:8-13. [PMID: 29293100 DOI: 10.1097/paf.0000000000000366] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The polysialylated isoform of the neural cell adhesion molecule (PSA-NCAM) has been shown to be a key player in neuroplastic changes and is expressed in various disorders. We investigated the PSA-NCAM expression on brain cortical tissue in a cohort of drug-related deaths. Brains from 25 drug abusers and 10 control subjects were removed at autopsy, and 2 samples of the right parietal lobe of each case were obtained. The polysialylated isoform of NCAM was evaluated on formalin-fixed and paraffin-embedded tissues. Eleven patients were polydrug abusers; 14 used a single substance. The mechanisms of death were acute respiratory failure (n = 19), cardiorespiratory failure (n = 4), acute heart failure (n = 1), and brain injury (n = 1). Toxicological analyses of blood were available for all cases, and urine and bile analyses for 19 of 25 cases. The polysialylated isoform of NCAM immunoexpression in the neuronal soma and dendritic spines was observed in 18 (72%) of 25 drug abusers and in 2 (20%) of 10 control subjects. Drug abusers were statistically more positive for PSA-NCAM than control subjects (P = 0.0082). The expression of PSA-NCAM in the parietal cortex could be an indicator of brain damage due to drug abuse, and its availability could allow the forensic pathologists to develop rapid and low-cost additional or alternative method to improve detection of drug-related deaths.
Collapse
Affiliation(s)
- Vito Cirielli
- From the *Forensic Pathology Unit, Department of Diagnostics and Public Health, and †Anatomic Pathology Unit, Department of Diagnostics and Public Health, University and Hospital Trust of Verona; and ‡Department of Neurological, Biomedical and Movement Sciences, University of Verona, Verona; and §Institute of Legal Medicine, Department of Public Health Sciences, University of Roma Tor Vergata, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Hillen AEJ, Burbach JPH, Hol EM. Cell adhesion and matricellular support by astrocytes of the tripartite synapse. Prog Neurobiol 2018; 165-167:66-86. [PMID: 29444459 DOI: 10.1016/j.pneurobio.2018.02.002] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 07/25/2017] [Accepted: 02/07/2018] [Indexed: 12/18/2022]
Abstract
Astrocytes contribute to the formation, function, and plasticity of synapses. Their processes enwrap the neuronal components of the tripartite synapse, and due to this close interaction they are perfectly positioned to modulate neuronal communication. The interaction between astrocytes and synapses is facilitated by cell adhesion molecules and matricellular proteins, which have been implicated in the formation and functioning of tripartite synapses. The importance of such neuron-astrocyte integration at the synapse is underscored by the emerging role of astrocyte dysfunction in synaptic pathologies such as autism and schizophrenia. Here we review astrocyte-expressed cell adhesion molecules and matricellular molecules that play a role in integration of neurons and astrocytes within the tripartite synapse.
Collapse
Affiliation(s)
- Anne E J Hillen
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CG Utrecht, The Netherlands; Department of Pediatrics/Child Neurology, VU University Medical Center, 1081 HV Amsterdam, The Netherlands
| | - J Peter H Burbach
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CG Utrecht, The Netherlands
| | - Elly M Hol
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CG Utrecht, The Netherlands; Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, 1098 XH Amsterdam, The Netherlands; Department of Neuroimmunology, Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences, 1105 BA Amsterdam, The Netherlands.
| |
Collapse
|
19
|
Kos MZ, Carless MA, Peralta J, Curran JE, Quillen EE, Almeida M, Blackburn A, Blondell L, Roalf DR, Pogue-Geile MF, Gur RC, Göring HHH, Nimgaonkar VL, Gur RE, Almasy L. Exome sequences of multiplex, multigenerational families reveal schizophrenia risk loci with potential implications for neurocognitive performance. Am J Med Genet B Neuropsychiatr Genet 2017; 174:817-827. [PMID: 28902459 PMCID: PMC5760172 DOI: 10.1002/ajmg.b.32597] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 08/16/2017] [Indexed: 12/28/2022]
Abstract
Schizophrenia is a serious mental illness, involving disruptions in thought and behavior, with a worldwide prevalence of about one percent. Although highly heritable, much of the genetic liability of schizophrenia is yet to be explained. We searched for susceptibility loci in multiplex, multigenerational families affected by schizophrenia, targeting protein-altering variation with in silico predicted functional effects. Exome sequencing was performed on 136 samples from eight European-American families, including 23 individuals diagnosed with schizophrenia or schizoaffective disorder. In total, 11,878 non-synonymous variants from 6,396 genes were tested for their association with schizophrenia spectrum disorders. Pathway enrichment analyses were conducted on gene-based test results, protein-protein interaction (PPI) networks, and epistatic effects. Using a significance threshold of FDR < 0.1, association was detected for rs10941112 (p = 2.1 × 10-5 ; q-value = 0.073) in AMACR, a gene involved in fatty acid metabolism and previously implicated in schizophrenia, with significant cis effects on gene expression (p = 5.5 × 10-4 ), including brain tissue data from the Genotype-Tissue Expression project (minimum p = 6.0 × 10-5 ). A second SNP, rs10378 located in TMEM176A, also shows risk effects in the exome data (p = 2.8 × 10-5 ; q-value = 0.073). PPIs among our top gene-based association results (p < 0.05; n = 359 genes) reveal significant enrichment of genes involved in NCAM-mediated neurite outgrowth (p = 3.0 × 10-5 ), while exome-wide SNP-SNP interaction effects for rs10941112 and rs10378 indicate a potential role for kinase-mediated signaling involved in memory and learning. In conclusion, these association results implicate AMACR and TMEM176A in schizophrenia risk, whose effects may be modulated by genes involved in synaptic plasticity and neurocognitive performance.
Collapse
Affiliation(s)
- Mark Z. Kos
- South Texas Diabetes and Obesity Institute, The University of Texas
Rio Grande Valley, San Antonio and Brownsville, TX, USA
| | - Melanie A. Carless
- Department of Genetics, Texas Biomedical Research Institute, San
Antonio, TX, USA
| | - Juan Peralta
- South Texas Diabetes and Obesity Institute, The University of Texas
Rio Grande Valley, San Antonio and Brownsville, TX, USA
| | - Joanne E. Curran
- South Texas Diabetes and Obesity Institute, The University of Texas
Rio Grande Valley, San Antonio and Brownsville, TX, USA
| | - Ellen E. Quillen
- Department of Genetics, Texas Biomedical Research Institute, San
Antonio, TX, USA
| | - Marcio Almeida
- South Texas Diabetes and Obesity Institute, The University of Texas
Rio Grande Valley, San Antonio and Brownsville, TX, USA
| | - August Blackburn
- South Texas Diabetes and Obesity Institute, The University of Texas
Rio Grande Valley, San Antonio and Brownsville, TX, USA
| | - Lucy Blondell
- South Texas Diabetes and Obesity Institute, The University of Texas
Rio Grande Valley, San Antonio and Brownsville, TX, USA
| | - David R. Roalf
- Department of Psychiatry, University of Pennsylvania School of
Medicine, Philadelphia, PA, USA
| | | | - Ruben C. Gur
- Department of Psychiatry, University of Pennsylvania School of
Medicine, Philadelphia, PA, USA
| | - Harald H. H. Göring
- South Texas Diabetes and Obesity Institute, The University of Texas
Rio Grande Valley, San Antonio and Brownsville, TX, USA
| | | | - Raquel E. Gur
- Department of Psychiatry, University of Pennsylvania School of
Medicine, Philadelphia, PA, USA
| | - Laura Almasy
- Departments of Genetics, University of Pennsylvania School of
Medicine, Philadelphia, PA, USA
| |
Collapse
|
20
|
Westphal N, Theis T, Loers G, Schachner M, Kleene R. Nuclear fragments of the neural cell adhesion molecule NCAM with or without polysialic acid differentially regulate gene expression. Sci Rep 2017; 7:13631. [PMID: 29051583 PMCID: PMC5648764 DOI: 10.1038/s41598-017-14056-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 10/04/2017] [Indexed: 02/05/2023] Open
Abstract
The neural cell adhesion molecule (NCAM) is the major carrier of polysialic acid (PSA) which modulates NCAM functions of neural cells at the cell surface. In previous studies, we have shown that stimulation of cultured neurons with surrogate NCAM ligands leads to the generation and nuclear import of PSA-lacking and -carrying NCAM fragments. Here, we show that the nuclear import of the PSA-carrying NCAM fragment is mediated by positive cofactor 4 and cofilin, which we identified as novel PSA-binding proteins. In the nucleus, the PSA-carrying NCAM fragment interacts via PSA with PC4 and cofilin, which are involved in RNA polymerase II-dependent transcription. Microarray analysis revealed that the nuclear PSA-carrying and -lacking NCAM fragments affect expression of different genes. By qPCR and immunoblot analysis we verified that the nuclear PSA-carrying NCAM fragment increases mRNA and protein expression of nuclear receptor subfamily 2 group F member 6, whereas the PSA-lacking NCAM fragment increases mRNA and protein expression of low density lipoprotein receptor-related protein 2 and α-synuclein. Differential gene expression evoked by nuclear NCAM fragments without and with PSA indicates that PSA-carrying and -lacking NCAM play different functional roles in the nervous system.
Collapse
Affiliation(s)
- Nina Westphal
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, 20251, Hamburg, Germany
| | - Thomas Theis
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, 20251, Hamburg, Germany
| | - Gabriele Loers
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, 20251, Hamburg, Germany
| | - Melitta Schachner
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, China.
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ, 08854, USA.
| | - Ralf Kleene
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, 20251, Hamburg, Germany
| |
Collapse
|
21
|
Loers G, Astafiev S, Hapiak Y, Saini V, Mishra B, Gul S, Kaur G, Schachner M, Theis T. The polysialic acid mimetics idarubicin and irinotecan stimulate neuronal survival and neurite outgrowth and signal via protein kinase C. J Neurochem 2017; 142:392-406. [PMID: 28542923 PMCID: PMC5539918 DOI: 10.1111/jnc.14076] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 05/11/2017] [Accepted: 05/15/2017] [Indexed: 02/05/2023]
Abstract
Polysialic acid (PSA) is a large, negatively charged, linear homopolymer of alpha2-8-linked sialic acid residues. It is generated by two polysialyltransferases and attached to N- and/or O-linked glycans, and its main carrier is the neural cell adhesion molecule (NCAM). PSA controls the development and regeneration of the nervous system by enhancing cell migration, axon pathfinding, synaptic targeting, synaptic plasticity, by regulating the differentiation of progenitor cells and by modulating cell-cell and cell-matrix adhesions. In the adult, PSA plays a role in the immune system, and PSA mimetics promote functional recovery after nervous system injury. In search for novel small molecule mimetics of PSA that are applicable for therapy, we identified idarubicin, an antineoplastic anthracycline, and irinotecan, an antineoplastic agent of the topoisomerase I inhibitor class, as PSA mimetics using a competition enzyme-linked immunosorbent assay. Idarubicin and irinotecan compete with the PSA-mimicking peptide and colominic acid, the bacterial analog of PSA, for binding to the PSA-specific monoclonal antibody 735. Idarubicin and irinotecan stimulate neurite outgrowth and survival of cultured cerebellar neurons after oxidative stress via protein kinase C and Erk1/2 in a similar manner as colominic acid, whereas Fyn, casein kinase II and the phosphatase and tensin homolog are only involved in idarubicin and irinotecan-stimulated neurite outgrowth. These novel results show that the structure and function of PSA can be mimicked by the small organic compounds irinotecan and idarubicin which trigger the same signaling cascades as PSA, thus introducing the possibility of retargeting these drugs to treat nervous system injuries.
Collapse
Affiliation(s)
- Gabriele Loers
- Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, D-20251 Hamburg, Germany
| | - Steven Astafiev
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854, USA
| | - Yuliya Hapiak
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854, USA
| | - Vedangana Saini
- Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, D-20251 Hamburg, Germany
- Department of Biotechnology, Guru Nanak Dev University, GT Road, 143005 Amritsar, India
| | - Bibhudatta Mishra
- Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, D-20251 Hamburg, Germany
| | - Sheraz Gul
- Fraunhofer Institute for Molecular Biology and Applied Ecology Screening Port (Fraunhofer-IME SP), Schnackenburgalle114, D-22525 Hamburg, Germany
| | - Gurcharan Kaur
- Department of Biotechnology, Guru Nanak Dev University, GT Road, 143005 Amritsar, India
| | - Melitta Schachner
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854, USA
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong 515041, China
- To whom correspondence should be addressed: Melitta Schachner, Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854, USA; phone: +1-732-445-1780; fax: +1-732-445-2063; ; or Melitta Schachner, Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong 515041, China; phone: + 86 754 8890 0276; fax: + 86 754 8890 0236;
| | - Thomas Theis
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854, USA
| |
Collapse
|
22
|
Monzo HJ, Coppieters N, Park TIH, Dieriks BV, Faull RLM, Dragunow M, Curtis MA. Insulin promotes cell migration by regulating PSA-NCAM. Exp Cell Res 2017; 355:26-39. [PMID: 28341445 DOI: 10.1016/j.yexcr.2017.03.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 03/02/2017] [Accepted: 03/14/2017] [Indexed: 12/21/2022]
Abstract
Cellular interactions with the extracellular environment are modulated by cell surface polysialic acid (PSA) carried by the neural cell adhesion molecule (NCAM). PSA-NCAM is involved in cellular processes such as differentiation, plasticity, and migration, and is elevated in Alzheimer's disease as well as in metastatic tumour cells. Our previous work demonstrated that insulin enhances the abundance of cell surface PSA by inhibiting PSA-NCAM endocytosis. In the present study we have identified a mechanism for insulin-dependent inhibition of PSA-NCAM turnover affecting cell migration. Insulin enhanced the phosphorylation of the focal adhesion kinase leading to dissociation of αv-integrin/PSA-NCAM clusters, and promoted cell migration. Our results show that αv-integrin plays a key role in the PSA-NCAM turnover process. αv-integrin knockdown stopped PSA-NCAM from being endocytosed, and αv-integrin/PSA-NCAM clusters co-labelled intracellularly with Rab5, altogether indicating a role for αv-integrin as a carrier for PSA-NCAM during internalisation. Furthermore, inhibition of p-FAK caused dissociation of αv-integrin/PSA-NCAM clusters and counteracted the insulin-induced accumulation of PSA at the cell surface and cell migration was impaired. Our data reveal a functional association between the insulin/p-FAK-dependent regulation of PSA-NCAM turnover and cell migration through the extracellular matrix. Most importantly, they identify a novel mechanism for insulin-stimulated cell migration.
Collapse
Affiliation(s)
- Hector J Monzo
- Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag, 92019, Auckland, New Zealand; Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag, 92019, Auckland, New Zealand
| | - Natacha Coppieters
- Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag, 92019, Auckland, New Zealand; Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag, 92019, Auckland, New Zealand
| | - Thomas I H Park
- Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag, 92019, Auckland, New Zealand; Department of Pharmacology, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag, 92019, Auckland, New Zealand
| | - Birger V Dieriks
- Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag, 92019, Auckland, New Zealand; Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag, 92019, Auckland, New Zealand
| | - Richard L M Faull
- Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag, 92019, Auckland, New Zealand; Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag, 92019, Auckland, New Zealand
| | - Mike Dragunow
- Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag, 92019, Auckland, New Zealand; Department of Pharmacology, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag, 92019, Auckland, New Zealand
| | - Maurice A Curtis
- Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag, 92019, Auckland, New Zealand; Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag, 92019, Auckland, New Zealand.
| |
Collapse
|
23
|
Minami A, Meguro Y, Ishibashi S, Ishii A, Shiratori M, Sai S, Horii Y, Shimizu H, Fukumoto H, Shimba S, Taguchi R, Takahashi T, Otsubo T, Ikeda K, Suzuki T. Rapid regulation of sialidase activity in response to neural activity and sialic acid removal during memory processing in rat hippocampus. J Biol Chem 2017; 292:5645-5654. [PMID: 28213516 DOI: 10.1074/jbc.m116.764357] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 02/07/2017] [Indexed: 12/12/2022] Open
Abstract
Sialidase cleaves sialic acids on the extracellular cell surface as well as inside the cell and is necessary for normal long-term potentiation (LTP) at mossy fiber-CA3 pyramidal cell synapses and for hippocampus-dependent spatial memory. Here, we investigated in detail the role of sialidase in memory processing. Sialidase activity measured with 4-methylumbelliferyl-α-d-N-acetylneuraminic acid (4MU-Neu5Ac) or 5-bromo-4-chloroindol-3-yl-α-d-N-acetylneuraminic acid (X-Neu5Ac) and Fast Red Violet LB was increased by high-K+-induced membrane depolarization. Sialidase activity was also increased by chemical LTP induction with forskolin and activation of BDNF signaling, non-NMDA receptors, or NMDA receptors. The increase in sialidase activity with neural excitation appears to be caused not by secreted sialidase or by an increase in sialidase expression but by a change in the subcellular localization of sialidase. Astrocytes as well as neurons are also involved in the neural activity-dependent increase in sialidase activity. Sialidase activity visualized with a benzothiazolylphenol-based sialic acid derivative (BTP3-Neu5Ac), a highly sensitive histochemical imaging probe for sialidase activity, at the CA3 stratum lucidum of rat acute hippocampal slices was immediately increased in response to LTP-inducible high-frequency stimulation on a time scale of seconds. To obtain direct evidence for sialic acid removal on the extracellular cell surface during neural excitation, the extracellular free sialic acid level in the hippocampus was monitored using in vivo microdialysis. The free sialic acid level was increased by high-K+-induced membrane depolarization. Desialylation also occurred during hippocampus-dependent memory formation in a contextual fear-conditioning paradigm. Our results show that neural activity-dependent desialylation by sialidase may be involved in hippocampal memory processing.
Collapse
Affiliation(s)
- Akira Minami
- From the Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan and
| | - Yuko Meguro
- From the Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan and
| | - Sayaka Ishibashi
- From the Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan and
| | - Ami Ishii
- From the Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan and
| | - Mako Shiratori
- From the Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan and
| | - Saki Sai
- From the Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan and
| | - Yuuki Horii
- From the Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan and
| | - Hirotaka Shimizu
- From the Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan and
| | - Hokuto Fukumoto
- From the Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan and
| | - Sumika Shimba
- From the Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan and
| | - Risa Taguchi
- From the Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan and
| | - Tadanobu Takahashi
- From the Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan and
| | - Tadamune Otsubo
- Department of Organic Chemistry, School of Pharmaceutical Sciences, Hiroshima International University, 5-1-1 Hirokoshingai, Kure-shi, Hiroshima 737-0112, Japan
| | - Kiyoshi Ikeda
- Department of Organic Chemistry, School of Pharmaceutical Sciences, Hiroshima International University, 5-1-1 Hirokoshingai, Kure-shi, Hiroshima 737-0112, Japan
| | - Takashi Suzuki
- From the Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan and
| |
Collapse
|
24
|
Mehrabian M, Hildebrandt H, Schmitt-Ulms G. NCAM1 Polysialylation: The Prion Protein's Elusive Reason for Being? ASN Neuro 2016; 8:8/6/1759091416679074. [PMID: 27879349 PMCID: PMC5122176 DOI: 10.1177/1759091416679074] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 09/08/2016] [Accepted: 10/02/2016] [Indexed: 01/06/2023] Open
Abstract
Much confusion surrounds the physiological function of the cellular prion protein (PrPC). It is, however, anticipated that knowledge of its function will shed light on its contribution to neurodegenerative diseases and suggest ways to interfere with the cellular toxicity central to them. Consequently, efforts to elucidate its function have been all but exhaustive. Building on earlier work that uncovered the evolutionary descent of the prion founder gene from an ancestral ZIP zinc transporter, we recently investigated a possible role of PrPC in a morphogenetic program referred to as epithelial-to-mesenchymal transition (EMT). By capitalizing on PrPC knockout cell clones in a mammalian cell model of EMT and using a comparative proteomics discovery strategy, neural cell adhesion molecule-1 emerged as a protein whose upregulation during EMT was perturbed in PrPC knockout cells. Follow-up work led us to observe that PrPC regulates the polysialylation of the neural cell adhesion molecule NCAM1 in cells undergoing morphogenetic reprogramming. In addition to governing cellular migration, polysialylation modulates several other cellular plasticity programs PrPC has been phenotypically linked to. These include neurogenesis in the subventricular zone, controlled mossy fiber sprouting and trimming in the hippocampal formation, hematopoietic stem cell renewal, myelin repair and maintenance, integrity of the circadian rhythm, and glutamatergic signaling. This review revisits this body of literature and attempts to present it in light of this novel contextual framework. When approached in this manner, a coherent model of PrPC acting as a regulator of polysialylation during specific cell and tissue morphogenesis events comes into focus.
Collapse
Affiliation(s)
- Mohadeseh Mehrabian
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Herbert Hildebrandt
- Institute for Cellular Chemistry, Hannover Medical School, Hannover, Germany
| | - Gerold Schmitt-Ulms
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada .,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
25
|
Regulation of extrasynaptic signaling by polysialylated NCAM: Impact for synaptic plasticity and cognitive functions. Mol Cell Neurosci 2016; 81:12-21. [PMID: 27865768 DOI: 10.1016/j.mcn.2016.11.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 11/10/2016] [Accepted: 11/11/2016] [Indexed: 01/24/2023] Open
Abstract
The activation of synaptic N-methyl-d-aspartate-receptors (NMDARs) is crucial for induction of synaptic plasticity and supports cell survival, whereas activation of extrasynaptic NMDARs inhibits long-term potentiation and triggers neurodegeneration. A soluble polysialylated form of the neural cell adhesion molecule (polySia-NCAM) suppresses signaling through peri-/extrasynaptic GluN2B-containing NMDARs. Genetic or enzymatic manipulations blocking this mechanism result in impaired synaptic plasticity and learning, which could be repaired by reintroduction of polySia, or inhibition of either GluN1/GluN2B receptors or downstream signaling through RasGRF1 and p38 MAP kinase. Ectodomain shedding of NCAM, and hence generation of soluble NCAM, is controlled by metalloproteases of a disintegrin and metalloprotease (ADAM) family. As polySia-NCAM is predominantly associated with GABAergic interneurons in the prefrontal cortex, it is noteworthy that EphrinA5/EphA3-induced ADAM10 activity promotes polySia-NCAM shedding in these neurons. Thus, in addition to the well-known regulation of synaptic NMDARs by the secreted molecule Reelin, shed polySia-NCAM may restrain activation of extrasynaptic NMDARs. These data support a concept that GABAergic interneuron-derived extracellular proteins control the balance in synaptic/extrasynaptic NMDAR-mediated signaling in principal cells. Strikingly, dysregulation of Reelin or polySia expression is linked to schizophrenia. Thus, targeting of the GABAergic interneuron-principle cell communication and restoring the balance in synaptic/extrasynaptic NMDARs represent promising strategies for treatment of psychiatric diseases.
Collapse
|
26
|
Minami A, Saito M, Mamada S, Ieno D, Hikita T, Takahashi T, Otsubo T, Ikeda K, Suzuki T. Role of Sialidase in Long-Term Potentiation at Mossy Fiber-CA3 Synapses and Hippocampus-Dependent Spatial Memory. PLoS One 2016; 11:e0165257. [PMID: 27783694 PMCID: PMC5081204 DOI: 10.1371/journal.pone.0165257] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 10/07/2016] [Indexed: 12/22/2022] Open
Abstract
Sialic acid bound to glycans in glycolipids and glycoproteins is essential for synaptic plasticity and memory. Sialidase (EC 3.2.1.18), which has 4 isozymes including Neu1, Neu2, Neu3 and Neu4, regulates the sialylation level of glycans by removing sialic acid from sialylglycoconjugate. In the present study, we investigated the distribution of sialidase activity in rat hippocampus and the role of sialidase in hippocampal memory processing. We previously developed a highly sensitive histochemical imaging probe for sialidase activity, BTP3-Neu5Ac. BTP3-Neu5Ac was cleaved efficiently by rat Neu2 and Neu4 at pH 7.3 and by Neu1 and Neu3 at pH 4.6. When a rat hippocampal acute slice was stained with BTP3-Neu5Ac at pH 7.3, mossy fiber terminal fields showed relatively intense sialidase activity. Thus, the role of sialidase in the synaptic plasticity was investigated at mossy fiber terminal fields. The long-term potentiation (LTP) at mossy fiber-CA3 pyramidal cell synapses was impaired by 2,3-dehydro-2-deoxy-N-acetylneuraminic acid (DANA), a sialidase inhibitor. DANA also failed to decrease paired-pulse facilitation after LTP induction. We also investigated the role of sialidase in hippocampus-dependent spatial memory by using the Morris water maze. The escape latency time to reach the platform was prolonged by DANA injection into the hippocampal CA3 region or by knockdown of Neu4 without affecting motility. The results show that the regulation of sialyl signaling by Neu4 is involved in hippocampal memory processing.
Collapse
Affiliation(s)
- Akira Minami
- Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52–1 Yada, Suruga-ku, Shizuoka 422–8526, Japan
| | - Masakazu Saito
- Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52–1 Yada, Suruga-ku, Shizuoka 422–8526, Japan
| | - Shou Mamada
- Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52–1 Yada, Suruga-ku, Shizuoka 422–8526, Japan
| | - Daisuke Ieno
- Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52–1 Yada, Suruga-ku, Shizuoka 422–8526, Japan
| | - Tomoya Hikita
- Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52–1 Yada, Suruga-ku, Shizuoka 422–8526, Japan
| | - Tadanobu Takahashi
- Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52–1 Yada, Suruga-ku, Shizuoka 422–8526, Japan
| | - Tadamune Otsubo
- Department of Organic Chemistry, School of Pharmaceutical Sciences, Hiroshima International University, 5-1-1, Hirokoshingai, Kure-shi, Hiroshima 737–0112, Japan
| | - Kiyoshi Ikeda
- Department of Organic Chemistry, School of Pharmaceutical Sciences, Hiroshima International University, 5-1-1, Hirokoshingai, Kure-shi, Hiroshima 737–0112, Japan
| | - Takashi Suzuki
- Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52–1 Yada, Suruga-ku, Shizuoka 422–8526, Japan
- * E-mail:
| |
Collapse
|
27
|
Frei JA, Stoeckli ET. SynCAMs - From axon guidance to neurodevelopmental disorders. Mol Cell Neurosci 2016; 81:41-48. [PMID: 27594578 DOI: 10.1016/j.mcn.2016.08.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 08/28/2016] [Accepted: 08/31/2016] [Indexed: 12/22/2022] Open
Abstract
Many cell adhesion molecules are located at synapses but only few of them can be considered synaptic cell adhesion molecules in the strict sense. Besides the Neurexins and Neuroligins, the LRRTMs (leucine rich repeat transmembrane proteins) and the SynCAMs/CADMs can induce synapse formation when expressed in non-neuronal cells and therefore are true synaptic cell adhesion molecules. SynCAMs (synaptic cell adhesion molecules) are a subfamily of the immunoglobulin superfamily of cell adhesion molecules. As suggested by their name, they were first identified as cell adhesion molecules at the synapse which were sufficient to trigger synapse formation. They also contribute to myelination by mediating axon-glia cell contacts. More recently, their role in earlier stages of neural circuit formation was demonstrated, as they also guide axons both in the peripheral and in the central nervous system. Mutations in SynCAM genes were found in patients diagnosed with autism spectrum disorders. The diverse functions of SynCAMs during development suggest that neurodevelopmental disorders are not only due to defects in synaptic plasticity. Rather, early steps of neural circuit formation are likely to contribute.
Collapse
Affiliation(s)
- Jeannine A Frei
- Hussman Institute for Autism, 801 W Baltimore Street, Baltimore, MD 20201, United States
| | - Esther T Stoeckli
- Dept of Molecular Life Sciences and Neuroscience Center Zurich, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland.
| |
Collapse
|
28
|
Szewczyk LM, Brozko N, Nagalski A, Röckle I, Werneburg S, Hildebrandt H, Wisniewska MB, Kuznicki J. ST8SIA2 promotes oligodendrocyte differentiation and the integrity of myelin and axons. Glia 2016; 65:34-49. [PMID: 27534376 PMCID: PMC5129544 DOI: 10.1002/glia.23048] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 07/18/2016] [Accepted: 08/01/2016] [Indexed: 12/19/2022]
Abstract
ST8SIA2 is a polysialyltransferase that attaches polysialic acid to the glycoproteins NCAM1 and CADM1. Polysialylation is involved in brain development and plasticity. ST8SIA2 is a schizophrenia candidate gene, and St8sia2−/− mice exhibit schizophrenia‐like behavior. We sought to identify new pathological consequences of ST8SIA2 deficiency. Our proteomic analysis suggested myelin impairment in St8sia2−/− mice. Histological and immune staining together with Western blot revealed that the onset of myelination was not delayed in St8sia2−/− mice, but the content of myelin was lower. Ultrastructure analysis of the corpus callosum showed thinner myelin sheaths, smaller and irregularly shaped axons, and white matter lesions in adult St8sia2−/− mice. Then we evaluated oligodendrocyte differentiation in vivo and in vitro. Fewer OLIG2+ cells in the cortex and corpus callosum, together with the higher percentage of undifferentiated oligodenroglia in St8sia2−/− mice suggested an impairment in oligodendrocyte generation. Experiment on primary cultures of oligodendrocyte precursor cells (OPCs) confirmed a cell‐autonomous effect of ST8SIA2 in oligodendroglia, and demonstrated that OPC to oligodendrocyte transition is inhibited in St8sia2−/− mice. Concluding, ST8SIA2‐mediated polysialylation influences on oligodendrocyte differentiation, and oligodendrocyte deficits in St8sia2 mice are a possible cause of the demyelination and degeneration of axons, resembling nerve fiber alterations in schizophrenia. GLIA 2016;65:34–49
Collapse
Affiliation(s)
- Lukasz Mateusz Szewczyk
- Laboratory of Neurodegeneration, International Institute of Molecular and Cell Biology, ul. Ks. Trojdena 4, Warszawa, 02-109, Poland.,Laboratory of Molecular Neurobiology, Centre of New Technologies, University of Warsaw, ul. Banacha 2C, Warszawa, 02-097, Poland.,Postgraduate School of Molecular Medicine, ul. Zwirki i Wigury 61, Warszawa, 02-091, Poland
| | - Nikola Brozko
- Laboratory of Molecular Neurobiology, Centre of New Technologies, University of Warsaw, ul. Banacha 2C, Warszawa, 02-097, Poland.,Postgraduate School of Molecular Medicine, ul. Zwirki i Wigury 61, Warszawa, 02-091, Poland
| | - Andrzej Nagalski
- Laboratory of Neurodegeneration, International Institute of Molecular and Cell Biology, ul. Ks. Trojdena 4, Warszawa, 02-109, Poland
| | - Iris Röckle
- Institute for Cellular Chemistry, Hannover Medical School, Carl-Neuberg-Strasse 1, Hannover, 30625, Germany
| | - Sebastian Werneburg
- Institute for Cellular Chemistry, Hannover Medical School, Carl-Neuberg-Strasse 1, Hannover, 30625, Germany
| | - Herbert Hildebrandt
- Institute for Cellular Chemistry, Hannover Medical School, Carl-Neuberg-Strasse 1, Hannover, 30625, Germany
| | - Marta Barbara Wisniewska
- Laboratory of Neurodegeneration, International Institute of Molecular and Cell Biology, ul. Ks. Trojdena 4, Warszawa, 02-109, Poland.,Laboratory of Molecular Neurobiology, Centre of New Technologies, University of Warsaw, ul. Banacha 2C, Warszawa, 02-097, Poland
| | - Jacek Kuznicki
- Laboratory of Neurodegeneration, International Institute of Molecular and Cell Biology, ul. Ks. Trojdena 4, Warszawa, 02-109, Poland
| |
Collapse
|
29
|
Saini V, Loers G, Kaur G, Schachner M, Jakovcevski I. Impact of neural cell adhesion molecule deletion on regeneration after mouse spinal cord injury. Eur J Neurosci 2016; 44:1734-46. [PMID: 27178448 DOI: 10.1111/ejn.13271] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 04/28/2016] [Accepted: 05/09/2016] [Indexed: 02/05/2023]
Abstract
The neural cell adhesion molecule (NCAM) plays important functional roles in development of the nervous system. We investigated the influence of a constitutive ablation of NCAM on the outcome of spinal cord injury. Transgenic mice lacking NCAM (NCAM-/-) were subjected to severe compression injury of the lower thoracic spinal cord using wild-type (NCAM+/+) littermates as controls. According to the single-frame motion analysis, the NCAM-/- mice showed reduced locomotor recovery in comparison to control mice at 3 and 6 weeks after injury, indicating an overall positive impact of NCAM on recovery after injury. Also the Basso Mouse Scale score was lower in NCAM-/- mice at 3 weeks after injury, whereas at 6 weeks after injury the difference between genotypes was not statistically significant. Worse locomotor function was associated with decreased monoaminergic and cholinergic innervation of the spinal cord caudal to the injury site and decreased axonal regrowth/sprouting at the site of injury. Astrocytic scar formation at the injury site, as assessed by immunohistology for glial fibrillary acidic protein at and around the lesion site was increased in NCAM-/- compared with NCAM+/+ mice. Migration of cultured monolayer astrocytes from NCAM-/- mice was reduced as assayed by scratch wounding. Numbers of Iba-1 immunopositive microglia were not different between genotypes. We conclude that constitutive NCAM deletion in young adult mice reduces recovery after spinal cord injury, validating the hypothesized beneficial role of this molecule in recovery after injury.
Collapse
Affiliation(s)
- Vedangana Saini
- Zentrum für Molekulare Neurobiologie, Universitätskrankenhaus Hamburg-Eppendorf, Universität Hamburg, Hamburg, Germany
- Department of Biotechnology, Guru Nanak Dev University, Punjab, India
| | - Gabriele Loers
- Zentrum für Molekulare Neurobiologie, Universitätskrankenhaus Hamburg-Eppendorf, Universität Hamburg, Hamburg, Germany
| | - Gurcharan Kaur
- Department of Biotechnology, Guru Nanak Dev University, Punjab, India
| | - Melitta Schachner
- Zentrum für Molekulare Neurobiologie, Universitätskrankenhaus Hamburg-Eppendorf, Universität Hamburg, Hamburg, Germany
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, 515041, China
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, 08854, USA
| | - Igor Jakovcevski
- Institute for Molecular and Behavioral Neuroscience, University Hospital Cologne, Köln, Germany
- Experimental Neurophysiology, German Center for Neurodegenerative Diseases, Ludwig-Erhard-Allee 2, D-53175, Bonn, Germany
| |
Collapse
|
30
|
Kaplan BLF, Li J, LaPres JJ, Pruett SB, Karmaus PWF. Contributions of nonhematopoietic cells and mediators to immune responses: implications for immunotoxicology. Toxicol Sci 2016; 145:214-32. [PMID: 26008184 DOI: 10.1093/toxsci/kfv060] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Immunotoxicology assessments have historically focused on the effects that xenobiotics exhibit directly on immune cells. These studies are invaluable as they identify immune cell targets and help characterize mechanisms and/or adverse outcome pathways of xenobiotics within the immune system. However, leukocytes can receive environmental cues by cell-cell contact or via released mediators from cells of organs outside of the immune system. These organs include, but are not limited to, the mucosal areas such as the lung and the gut, the liver, and the central nervous system. Homeostatic perturbation in these organs induced directly by toxicants can initiate and alter the outcome of local and systemic immunity. This review will highlight some of the identified nonimmune influences on immune homeostasis and provide summaries of how immunotoxic mechanisms of selected xenobiotics involve nonimmune cells or mediators. Thus, this review will identify data gaps and provide possible alternative mechanisms by which xenobiotics alter immune function that could be considered during immunotoxicology safety assessment.
Collapse
Affiliation(s)
- Barbara L F Kaplan
- *Center for Environmental Health Sciences, Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi St, Mississippi 39762, Safety Assessment, Genentech, Inc. South San Francisco, California 94080, Department of Biochemistry and Molecular Biology, Center for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824 and Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105
| | - Jinze Li
- *Center for Environmental Health Sciences, Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi St, Mississippi 39762, Safety Assessment, Genentech, Inc. South San Francisco, California 94080, Department of Biochemistry and Molecular Biology, Center for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824 and Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105
| | - John J LaPres
- *Center for Environmental Health Sciences, Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi St, Mississippi 39762, Safety Assessment, Genentech, Inc. South San Francisco, California 94080, Department of Biochemistry and Molecular Biology, Center for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824 and Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105
| | - Stephen B Pruett
- *Center for Environmental Health Sciences, Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi St, Mississippi 39762, Safety Assessment, Genentech, Inc. South San Francisco, California 94080, Department of Biochemistry and Molecular Biology, Center for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824 and Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105
| | - Peer W F Karmaus
- *Center for Environmental Health Sciences, Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi St, Mississippi 39762, Safety Assessment, Genentech, Inc. South San Francisco, California 94080, Department of Biochemistry and Molecular Biology, Center for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824 and Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105
| |
Collapse
|
31
|
Mione J, Manrique C, Duhoo Y, Roman FS, Guiraudie-Capraz G. Expression of polysialyltransferases (STX and PST) in adult rat olfactory bulb after an olfactory associative discrimination task. Neurobiol Learn Mem 2016; 130:52-60. [PMID: 26844880 DOI: 10.1016/j.nlm.2016.01.011] [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: 10/26/2015] [Revised: 01/11/2016] [Accepted: 01/25/2016] [Indexed: 11/18/2022]
Abstract
Neuronal plasticity and neurogenesis occur in the adult hippocampus and in other brain structures such as the olfactory bulb and often involve the neural cell adhesion molecule NCAM. During an olfactory associative discrimination learning task, NCAM polysialylation triggers neuronal plasticity in the adult hippocampus. The PST enzyme likely modulates this polysialylation, but not STX, a second sialyltransferase. How the two polysialyltransferases are involved in the adult olfactory bulb remains unknown. We addressed this question by investigating the effect of olfactory associative learning on plasticity and neurogenesis. After a hippocampo-dependent olfactory associative task learning, we measured the expression of both PST and STX polysialyltransferases in the olfactory bulbs of adult rats using quantitative PCR. In parallel, immunohistochemistry was used to evaluate both NCAM polysialylation level and newly-born cells, with or without learning. After learning, no changes were observed neither in the expression level of PST and NCAM polysialylation, nor in STX gene expression level and newly-born cells number in the olfactory bulb.
Collapse
Affiliation(s)
- J Mione
- Aix Marseille Université, CNRS, NICN, UMR 7259, 13344 Marseille, France
| | - C Manrique
- Aix Marseille Université, CNRS, FR 3512, 13331 Marseille, France
| | - Y Duhoo
- Aix Marseille Université, CNRS, NICN, UMR 7259, 13344 Marseille, France
| | - F S Roman
- Aix Marseille Université, CNRS, NICN, UMR 7259, 13344 Marseille, France
| | - G Guiraudie-Capraz
- Aix Marseille Université, CNRS, NICN, UMR 7259, 13344 Marseille, France.
| |
Collapse
|
32
|
Loers G, Saini V, Mishra B, Gul S, Chaudhury S, Wallqvist A, Kaur G, Schachner M. Vinorelbine and epirubicin share common features with polysialic acid and modulate neuronal and glial functions. J Neurochem 2016; 136:48-62. [PMID: 26443186 PMCID: PMC4904230 DOI: 10.1111/jnc.13383] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 09/21/2015] [Accepted: 10/02/2015] [Indexed: 02/05/2023]
Abstract
Polysialic acid (PSA), a large, linear glycan composed of 8 to over 100 α2,8-linked sialic acid residues, modulates development of the nervous system by enhancing cell migration, axon pathfinding, and synaptic targeting and by regulating differentiation of progenitor cells. PSA also functions in developing and adult immune systems and is a signature of many cancers. In this study we identified vinorelbine, a semi-synthetic third generation vinca alkaloid, and epirubicin, an anthracycline and 4'-epimer of doxorubicin, as PSA mimetics. Similar to PSA, vinorelbine and epirubicin bind to the PSA-specific monoclonal antibody 735 and compete with the bacterial analog of PSA, colominic acid in binding to monoclonal antibody 735. Vinorelbine and epirubicin stimulate neurite outgrowth of cerebellar neurons via the neural cell adhesion molecule, via myristoylated alanine-rich C kinase substrate, and via fibroblast growth factor receptor, signaling through Erk pathways. Furthermore, the two compounds enhance process formation of Schwann cells and migration of cerebellar neurons in culture, and reduce migration of astrocytes after injury. These novel results show that the structure and function of PSA can be mimicked by the small organic compounds vinorelbine and epirubicin, thus raising the possibility to re-target drugs used in treatment of cancers to nervous system repair. Vinorelbine and epirubicin, identified as PSA mimetics, enhance, like PSA, neuronal migration, neuritogenesis, and formation of Schwann cell processes, and reduce astrocytic migration. Ablating NCAM, inhibiting fibroblast growth factor (FGFR) receptor, or adding the effector domain of myristoylated alanine-rich C kinase substrate (MARCKS) minimize the vinorelbine and epirubicin effects, indicating that they are true PSA mimetics triggering PSA-mediated functions.
Collapse
Affiliation(s)
- Gabriele Loers
- Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, D-20251 Hamburg, Germany
| | - Vedangana Saini
- Department of Biotechnology, Guru Nanak Dev University, GT Road, 143005 Amritsar, India
| | - Bibhudatta Mishra
- Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, D-20251 Hamburg, Germany
| | - Sheraz Gul
- Fraunhofer Institute for Molecular Biology and Applied Ecology ScreeningPort (Fraunhofer-IME SP), Schnackenburgalle114, D-22525 Hamburg, Germany
| | - Sidhartha Chaudhury
- DoD Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, US Army Medical Research and Materiel Command, Fort Detrick, MD 21702 (USA)
| | - Anders Wallqvist
- DoD Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, US Army Medical Research and Materiel Command, Fort Detrick, MD 21702 (USA)
| | - Gurcharan Kaur
- Department of Biotechnology, Guru Nanak Dev University, GT Road, 143005 Amritsar, India
| | - Melitta Schachner
- Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, D-20251 Hamburg, Germany
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854, USA
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong 515041, China
| |
Collapse
|
33
|
A Dendritic Golgi Satellite between ERGIC and Retromer. Cell Rep 2016; 14:189-99. [DOI: 10.1016/j.celrep.2015.12.024] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 11/08/2015] [Accepted: 11/25/2015] [Indexed: 11/20/2022] Open
|
34
|
Colombo F, Meldolesi J. L1-CAM and N-CAM: From Adhesion Proteins to Pharmacological Targets. Trends Pharmacol Sci 2015; 36:769-781. [PMID: 26478212 DOI: 10.1016/j.tips.2015.08.004] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 07/31/2015] [Accepted: 08/04/2015] [Indexed: 12/14/2022]
Abstract
L1 cell adhesion molecule (L1-CAM) and neural cell adhesion molecule (N-CAM), key members of the immunoglobulin-like CAM (Ig-CAM) family, were first recognized to play critical roles in surface interactions of neurons, by binding with each other and with extracellular matrix (ECM) proteins. Subsequently, adhesion was recognized to include signaling due to both activation of β-integrin, with the generation of intracellular cascades, and integration with the surface cytoskeleton. The importance of the two Ig-CAMs was revealed by their activation of the tyrosine kinase receptors of fibroblast growth factor (FGF), epidermal growth factor (EGF), and nerve growth factor (NGF). Based on these complex signaling properties, L1-CAM and N-CAM have become of great potential pharmacological interest in neurons and cancers. Treatment of neurodegenerative disorders and cognitive deficits of neurons is aimed to increase the cell Ig-CAM tone, possibly provided by synthetic/mimetic peptides. In cancer cells, where Ig-CAMs are often overexpressed, the proteins are employed for prognosis. The approaches to therapy are based on protein downregulation, antibodies, and adoptive immunotherapy.
Collapse
Affiliation(s)
- Federico Colombo
- Vita-Salute San Raffaele University and S. Raffaele Scientific Institute, Via Olgettina 58, 20132 Milan, Italy
| | - Jacopo Meldolesi
- Vita-Salute San Raffaele University and S. Raffaele Scientific Institute, Via Olgettina 58, 20132 Milan, Italy.
| |
Collapse
|
35
|
Mehrabian M, Brethour D, Wang H, Xi Z, Rogaeva E, Schmitt-Ulms G. The Prion Protein Controls Polysialylation of Neural Cell Adhesion Molecule 1 during Cellular Morphogenesis. PLoS One 2015; 10:e0133741. [PMID: 26288071 PMCID: PMC4546001 DOI: 10.1371/journal.pone.0133741] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 06/19/2015] [Indexed: 01/06/2023] Open
Abstract
Despite its multi-faceted role in neurodegenerative diseases, the physiological function of the prion protein (PrP) has remained elusive. On the basis of its evolutionary relationship to ZIP metal ion transporters, we considered that PrP may contribute to the morphogenetic reprogramming of cells underlying epithelial-to-mesenchymal transitions (EMT). Consistent with this hypothesis, PrP transcription increased more than tenfold during EMT, and stable PrP-deficient cells failed to complete EMT in a mammalian cell model. A global comparative proteomics analysis identified the neural cell adhesion molecule 1 (NCAM1) as a candidate mediator of this impairment, which led to the observation that PrP-deficient cells fail to undergo NCAM1 polysialylation during EMT. Surprisingly, this defect was caused by a perturbed transcription of the polysialyltransferase ST8SIA2 gene. Proteomics data pointed toward β-catenin as a transcriptional regulator affected in PrP-deficient cells. Indeed, pharmacological blockade or siRNA-based knockdown of β-catenin mimicked PrP-deficiency in regards to NCAM1 polysialylation. Our data established the existence of a PrP-ST8SIA2-NCAM signaling loop, merged two mature fields of investigation and offer a simple model for explaining phenotypes linked to PrP.
Collapse
Affiliation(s)
- Mohadeseh Mehrabian
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Dylan Brethour
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Hansen Wang
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
| | - Zhengrui Xi
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
| | - Ekaterina Rogaeva
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Gerold Schmitt-Ulms
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
36
|
Abstract
This review presents principles of glycosylation, describes the relevant glycosylation pathways and their related disorders, and highlights some of the neurological aspects and issues that continue to challenge researchers. More than 100 rare human genetic disorders that result from deficiencies in the different glycosylation pathways are known today. Most of these disorders impact the central and/or peripheral nervous systems. Patients typically have developmental delays/intellectual disabilities, hypotonia, seizures, neuropathy, and metabolic abnormalities in multiple organ systems. Among these disorders there is great clinical diversity because all cell types differentially glycosylate proteins and lipids. The patients have hundreds of misglycosylated products, which afflict a myriad of processes, including cell signaling, cell-cell interaction, and cell migration. This vast complexity in glycan composition and function, along with the limited availability of analytic tools, has impeded the identification of key glycosylated molecules that cause pathologies. To date, few critical target proteins have been pinpointed.
Collapse
|
37
|
Taguchi R, Minami A, Matsuda Y, Takahashi T, Otsubo T, Ikeda K, Suzuki T. Preferential Accumulation of 14C-N-Glycolylneuraminic Acid over 14C-N-Acetylneuraminic Acid in the Rat Brain after Tail Vein Injection. PLoS One 2015; 10:e0131061. [PMID: 26098915 PMCID: PMC4476740 DOI: 10.1371/journal.pone.0131061] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 05/28/2015] [Indexed: 11/25/2022] Open
Abstract
The two main molecular species of sialic acid existing in nature are N-acetylneuraminic acid (Neu5Ac) and N-glycolylneuraminic acid (Neu5Gc). Neu5Ac is abundant in mammalian brains and plays crucial roles in many neural functions. In contrast, Neu5Gc is present only at a trace level in vertebrate brains. The brain-specific suppression of Neu5Gc synthesis, which is a common feature in mammals, suggests that Neu5Gc has toxicity against brain functions. However, in vivo kinetics of Neu5Gc in the whole body, especially in the brain, has not been studied in sufficient detail. To determine the in vivo kinetics of Neu5Gc, 14C-Neu5Gc was enzymatically synthesized and injected into rat tail veins. Although most of 14C-Neu5Gc was excreted in urine, a small amount of 14C-Neu5Gc was detected in the brain. Brain autoradiography indicated that 14C-Neu5Gc was accumulated predominantly in the hippocampus. 14C-Neu5Gc transferred into the brain was incorporated into gangliosides including GM1, GD1a, GD1b, GT1b and GQ1b. Reduction of 14C-Neu5Gc after intracerebroventricular infusion was slower than that of 14C-Neu5Ac in the brain and hippocampus. The results suggest that Neu5Gc is transferred from blood into the brain across the blood brain barrier and accumulates in the brain more preferentially than does Neu5Ac.
Collapse
Affiliation(s)
- Risa Taguchi
- Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Akira Minami
- Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Yukino Matsuda
- Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Tadanobu Takahashi
- Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Tadamune Otsubo
- Department of Organic Chemistry, School of Pharmaceutical Sciences, Hiroshima International University, Hiroshima, Japan
| | - Kiyoshi Ikeda
- Department of Organic Chemistry, School of Pharmaceutical Sciences, Hiroshima International University, Hiroshima, Japan
| | - Takashi Suzuki
- Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
- * E-mail:
| |
Collapse
|
38
|
Viswaprakash N, Vaithianathan T, Viswaprakash A, Judd R, Parameshwaran K, Suppiramaniam V. Insulin treatment restores glutamate (α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid) receptor function in the hippocampus of diabetic rats. J Neurosci Res 2015; 93:1442-50. [PMID: 25807926 DOI: 10.1002/jnr.23589] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2014] [Revised: 12/22/2014] [Accepted: 02/26/2015] [Indexed: 11/10/2022]
Abstract
Type 1 diabetes is associated with cognitive dysfunction. Cognitive processing, particularly memory acquisition, depends on the regulated enhancement of expression and function of glutamate receptor subtypes in the hippocampus. Impairment of memory was been detected in rodent models of type 1 diabetes induced by streptozotocin (STZ). This study examines the functional properties of synaptic α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors and the expression of synaptic molecules that regulate glutamatergic synaptic transmission in the hippocampus of STZ-diabetic rats. The AMPA receptor-mediated miniature excitatory postsynaptic currents (mEPSCs) and single-channel properties of synaptosomal AMPA receptors were examined after 4 weeks of diabetes induction. Results show that amplitude and frequency of mEPSCs recorded from CA1 pyramidal neurons were decreased in diabetic rats. In addition, the single-channel properties of synaptic AMPA receptors from diabetic rat hippocampi were different from those of controls. These impairments in synaptic currents gated by AMPA receptors were accompanied by decreased protein levels of AMPA receptor subunit GluR1, the presynaptic protein synaptophysin, and the postsynaptic anchor protein postsynaptic density protein 95 in the hippocampus of diabetic rats. Neural cell adhesion molecule (NCAM), an extracellular matrix molecule abundantly expressed in the brain, and the polysialic acid (PSA) attached to NCAM were also downregulated in the hippocampus of diabetic rats. Insulin treatment, when initiated at the onset of diabetes induction, reduced these effects. These findings suggest that STZ-induced diabetes may result in functional deteriorations in glutamatergic synapses in the hippocampus of rats and that these effects may be reduced by insulin treatment.
Collapse
Affiliation(s)
- Nilmini Viswaprakash
- Department of Biomedical Sciences, College of Veterinary Medicine, Nursing and Allied Health, Tuskegee University, Tuskegee, Alabama
| | - Thirumalini Vaithianathan
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, New York.,Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, Alabama
| | - Ajitan Viswaprakash
- Biology Department and Spine Rehabilitation Center, University of Alabama-Birmingham, Birmingham, Alabama
| | - Robert Judd
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Kodeeswaran Parameshwaran
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, Alabama.,Department of Biological and Environmental Sciences, Texas A&M University-Commerce, Commerce, Texas
| | - Vishnu Suppiramaniam
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, Alabama
| |
Collapse
|
39
|
Tissue-based metabolic labeling of polysialic acids in living primary hippocampal neurons. Proc Natl Acad Sci U S A 2015; 112:E241-8. [PMID: 25564666 DOI: 10.1073/pnas.1419683112] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The posttranslational modification of neural cell-adhesion molecule (NCAM) with polysialic acid (PSA) and the spatiotemporal distribution of PSA-NCAM play an important role in the neuronal development. In this work, we developed a tissue-based strategy for metabolically incorporating an unnatural monosaccharide, peracetylated N-azidoacetyl-D-mannosamine, in the sialic acid biochemical pathway to present N-azidoacetyl sialic acid to PSA-NCAM. Although significant neurotoxicity was observed in the conventional metabolic labeling that used the dissociated neuron cells, neurotoxicity disappeared in this modified strategy, allowing for investigation of the temporal and spatial distributions of PSA in the primary hippocampal neurons. PSA-NCAM was synthesized and recycled continuously during neuronal development, and the two-color labeling showed that newly synthesized PSA-NCAMs were transported and inserted mainly to the growing neurites and not significantly to the cell body. This report suggests a reliable and cytocompatible method for in vitro analysis of glycans complementary to the conventional cell-based metabolic labeling for chemical glycobiology.
Collapse
|
40
|
Tantra M, Kröcher T, Papiol S, Winkler D, Röckle I, Jatho J, Burkhardt H, Ronnenberg A, Gerardy-Schahn R, Ehrenreich H, Hildebrandt H. St8sia2 deficiency plus juvenile cannabis exposure in mice synergistically affect higher cognition in adulthood. Behav Brain Res 2014; 275:166-75. [DOI: 10.1016/j.bbr.2014.08.062] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2014] [Revised: 08/25/2014] [Accepted: 08/30/2014] [Indexed: 12/23/2022]
|
41
|
Saroja SR, Sase A, Kircher SG, Wan J, Berger J, Höger H, Pollak A, Lubec G. Hippocampal proteoglycans brevican and versican are linked to spatial memory of Sprague-Dawley rats in the morris water maze. J Neurochem 2014; 130:797-804. [DOI: 10.1111/jnc.12783] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 05/22/2014] [Accepted: 06/01/2014] [Indexed: 01/21/2023]
Affiliation(s)
| | - Ajinkya Sase
- Department of Pediatrics; Medical University of Vienna; Vienna Austria
| | - Susanne G. Kircher
- Department of Medical Chemistry; Medical University of Vienna; Vienna Austria
| | - Jia Wan
- Department of Pediatrics; Medical University of Vienna; Vienna Austria
| | - Johannes Berger
- Department of Pathobiology of the Nervous System; Center for Brain Research; Medical University of Vienna; Vienna Austria
| | - Harald Höger
- Core Unit of Biomedical Research; Division of Laboratory Animal Science and Genetics; Medical University of Vienna; Himberg Austria
| | - Arnold Pollak
- Department of Pediatrics; Medical University of Vienna; Vienna Austria
| | - Gert Lubec
- Department of Pediatrics; Medical University of Vienna; Vienna Austria
| |
Collapse
|
42
|
Manrique C, Migliorati M, Gilbert V, Brezun JM, Chaillan FA, Truchet B, Khrestchatisky M, Guiraudie-Capraz G, Roman FS. Dynamic expression of the polysialyltransferase in adult rat hippocampus performing an olfactory associative task. Hippocampus 2014; 24:979-89. [DOI: 10.1002/hipo.22284] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Revised: 04/07/2014] [Accepted: 04/09/2014] [Indexed: 01/16/2023]
Affiliation(s)
| | | | - Valérie Gilbert
- Aix Marseille Université, CNRS; FR 3512 13331 Marseille France
| | | | | | - Bruno Truchet
- Aix Marseille Université, CNRS; UMR 7291 13331 Marseille France
| | | | | | - François S. Roman
- Aix Marseille Université, CNRS, NICN; UMR 7259 13344 Marseille France
| |
Collapse
|
43
|
Pinner AL, Haroutunian V, Meador-Woodruff JH. Alterations of the myristoylated, alanine-rich C kinase substrate (MARCKS) in prefrontal cortex in schizophrenia. Schizophr Res 2014; 154:36-41. [PMID: 24568864 PMCID: PMC3999918 DOI: 10.1016/j.schres.2014.02.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 01/27/2014] [Accepted: 02/01/2014] [Indexed: 11/29/2022]
Abstract
Abnormal synaptic plasticity has been implicated in the cognitive deficits seen in schizophrenia, where alterations have been found in neurotransmission, signaling and dendritic dynamics. Rapid rearrangement of the actin cytoskeleton is critical for plasticity and abnormalities of molecular regulators of this process are candidates for understanding mechanisms underlying these changes in schizophrenia. The myristoylated, alanine-rich C-kinase substrate (MARCKS) is crucial for many roles associated with synaptic plasticity, including facilitation of neurotransmission, dendritic branching and in turn cognitive function. Accordingly, we hypothesized that this protein is abnormally expressed or regulated in schizophrenia. We measured protein expression of MARCKS by Western blot analysis in postmortem samples of dorsolateral prefrontal cortex (DLPFC) from elderly schizophrenia patients (N=16) and a comparison group (N=20). We also assayed phosphorylated-MARCKS (pMARCKS), given the role of phosphorylation in reversing membrane association by MARCKS. We found decreased expression of both MARCKS and pMARCKS in schizophrenia. Altered myristoylation may be a mechanism that explains this down-regulation of MARCKS, so we also assayed expression of the two isoforms of the key myristoylation enzyme, NMT, and an enzymatic inhibitor of this enzyme, NMT-inhibitor protein (NIP71) by Western blotting in these same subjects. Expression did not change between groups for these proteins, suggesting a mechanism other than myristoylation is responsible for decreased MARCKS expression in schizophrenia. These data suggest a potential mechanism underlying aspects of altered synaptic plasticity observed in schizophrenia.
Collapse
Affiliation(s)
- Anita L. Pinner
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
,Corresponding author: CIRC 593, 1719 6 Ave South, Birmingham, AL 35294-0021, USA, Tel: +1 205 996 6212, Fax: + 1 205 975 4879,
| | - Vahram Haroutunian
- Department of Psychiatry, Mt. Sinai School of Medicine, New York, NY, USA
| | - James H. Meador-Woodruff
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
44
|
Neural ECM molecules in synaptic plasticity, learning, and memory. PROGRESS IN BRAIN RESEARCH 2014; 214:53-80. [DOI: 10.1016/b978-0-444-63486-3.00003-7] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
45
|
Loers G, Saini V, Mishra B, Papastefanaki F, Lutz D, Chaudhury S, Ripoll DR, Wallqvist A, Gul S, Schachner M, Kaur G. Nonyloxytryptamine mimics polysialic acid and modulates neuronal and glial functions in cell culture. J Neurochem 2013; 128:88-100. [DOI: 10.1111/jnc.12408] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Revised: 07/19/2013] [Accepted: 08/12/2013] [Indexed: 01/09/2023]
Affiliation(s)
- Gabriele Loers
- Zentrum für Molekulare Neurobiologie Hamburg; Universitätsklinikum Hamburg-Eppendorf; Hamburg Germany
| | - Vedangana Saini
- Department of Biotechnology; Guru Nanak Dev University; Amritsar Punjab India
| | - Bibhudatta Mishra
- Zentrum für Molekulare Neurobiologie Hamburg; Universitätsklinikum Hamburg-Eppendorf; Hamburg Germany
| | - Florentia Papastefanaki
- Laboratory of Cellular and Molecular Neurobiology; Hellenic Pasteur Institute; Athens Greece
| | - David Lutz
- Zentrum für Molekulare Neurobiologie Hamburg; Universitätsklinikum Hamburg-Eppendorf; Hamburg Germany
| | - Sidhartha Chaudhury
- DoD Biotechnology High Performance Computing Software Applications Institute; Telemedicine and Advanced Technology Research Center; US Army Medical Research and Materiel Command; Fort Detrick Maryland USA
| | - Daniel R. Ripoll
- DoD Biotechnology High Performance Computing Software Applications Institute; Telemedicine and Advanced Technology Research Center; US Army Medical Research and Materiel Command; Fort Detrick Maryland USA
| | - Anders Wallqvist
- DoD Biotechnology High Performance Computing Software Applications Institute; Telemedicine and Advanced Technology Research Center; US Army Medical Research and Materiel Command; Fort Detrick Maryland USA
| | - Sheraz Gul
- European ScreeningPort GmbH; Schnackenburgalle114; Hamburg Germany
| | - Melitta Schachner
- Zentrum für Molekulare Neurobiologie Hamburg; Universitätsklinikum Hamburg-Eppendorf; Hamburg Germany
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience; Rutgers University; Piscataway New Jersey USA
| | - Gurcharan Kaur
- Department of Biotechnology; Guru Nanak Dev University; Amritsar Punjab India
| |
Collapse
|
46
|
Monzo HJ, Park TIH, Dieriks BV, Jansson D, Faull RLM, Dragunow M, Curtis MA. Insulin and IGF1 modulate turnover of polysialylated neural cell adhesion molecule (PSA-NCAM) in a process involving specific extracellular matrix components. J Neurochem 2013; 126:758-70. [PMID: 23844825 DOI: 10.1111/jnc.12363] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Revised: 06/20/2013] [Accepted: 07/09/2013] [Indexed: 12/19/2022]
Abstract
Cellular interactions mediated by the neural cell adhesion molecule (NCAM) are critical in cell migration, differentiation and plasticity. Switching of the NCAM-interaction mode, from adhesion to signalling, is determined by NCAM carrying a particular post-translational modification, polysialic acid (PSA). Regulation of cell-surface PSA-NCAM is traditionally viewed as a direct consequence of polysialyltransferase activity. Taking advantage of the polysialyltransferase Ca²⁺-dependent activity, we demonstrate in TE671 cells that downregulation of PSA-NCAM synthesis constitutes a necessary but not sufficient condition to reduce cell-surface PSA-NCAM; instead, PSA-NCAM turnover required internalization of the molecule into the cytosol. PSA-NCAM internalization was specifically triggered by collagen in the extracellular matrix (ECM) and prevented by insulin-like growth factor (IGF1) and insulin. Our results pose a novel role for IGF1 and insulin in controlling cell migration through modulation of PSA-NCAM turnover at the cell surface. Neural cell adhesion molecules (NCAMs) are critically involved in cell differentiation and migration. Polysialylation (PSA)/desialylation of NCAMs switches their functional interaction mode and, in turn, migration and differentiation. We have found that the desialylation process of PSA-NCAM occurs via endocytosis, induced by collagen-IV and blocked by insulin-like growth factor (IGF1) and insulin, suggesting a novel association between PSA-NCAM, IGF1/insulin and brain/tumour plasticity.
Collapse
Affiliation(s)
- Hector J Monzo
- Faculty of Medical and Health Sciences, Centre for Brain Research, The University of Auckland, Auckland, New Zealand
| | | | | | | | | | | | | |
Collapse
|
47
|
Guirado R, Perez-Rando M, Sanchez-Matarredona D, Castillo-Gómez E, Liberia T, Rovira-Esteban L, Varea E, Crespo C, Blasco-Ibáñez JM, Nacher J. The dendritic spines of interneurons are dynamic structures influenced by PSA-NCAM expression. ACTA ACUST UNITED AC 2013; 24:3014-24. [PMID: 23780867 DOI: 10.1093/cercor/bht156] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Excitatory neurons undergo dendritic spine remodeling in response to different stimuli. However, there is scarce information about this type of plasticity in interneurons. The polysialylated form of the neural cell adhesion molecule (PSA-NCAM) is a good candidate to mediate this plasticity as it participates in neuronal remodeling and is expressed by some mature cortical interneurons, which have reduced dendritic arborization, spine density, and synaptic input. To study the connectivity of the dendritic spines of interneurons and the influence of PSA-NCAM on their dynamics, we have analyzed these structures in a subpopulation of fluorescent spiny interneurons in the hippocampus of glutamic acid decarboxylase-enhanced green fluorescent protein transgenic mice. Our results show that these spines receive excitatory synapses. The depletion of PSA in vivo using the enzyme Endo-Neuraminidase-N (Endo-N) increases spine density when analyzed 2 days after, but decreases it 7 days after. The dendritic spine turnover was also analyzed in real time using organotypic hippocampal cultures: 24 h after the addition of EndoN, we observed an increase in the apparition rate of spines. These results indicate that dendritic spines are important structures in the control of the synaptic input of hippocampal interneurons and suggest that PSA-NCAM is relevant in the regulation of their morphology and connectivity.
Collapse
Affiliation(s)
- Ramon Guirado
- Cell Biology Department, Neurobiology Unit and Program in Basic and Applied Neurosciences, Universitat de València, Valencia, Spain Current address: Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Marta Perez-Rando
- Cell Biology Department, Neurobiology Unit and Program in Basic and Applied Neurosciences, Universitat de València, Valencia, Spain
| | - David Sanchez-Matarredona
- Cell Biology Department, Neurobiology Unit and Program in Basic and Applied Neurosciences, Universitat de València, Valencia, Spain
| | - Esther Castillo-Gómez
- Cell Biology Department, Neurobiology Unit and Program in Basic and Applied Neurosciences, Universitat de València, Valencia, Spain
| | - Teresa Liberia
- Cell Biology Department, Neurobiology Unit and Program in Basic and Applied Neurosciences, Universitat de València, Valencia, Spain
| | - Laura Rovira-Esteban
- Cell Biology Department, Neurobiology Unit and Program in Basic and Applied Neurosciences, Universitat de València, Valencia, Spain
| | - Emilio Varea
- Cell Biology Department, Neurobiology Unit and Program in Basic and Applied Neurosciences, Universitat de València, Valencia, Spain
| | - Carlos Crespo
- Cell Biology Department, Neurobiology Unit and Program in Basic and Applied Neurosciences, Universitat de València, Valencia, Spain
| | - José Miguel Blasco-Ibáñez
- Cell Biology Department, Neurobiology Unit and Program in Basic and Applied Neurosciences, Universitat de València, Valencia, Spain
| | - Juan Nacher
- Cell Biology Department, Neurobiology Unit and Program in Basic and Applied Neurosciences, Universitat de València, Valencia, Spain Fundación Hospital Clínico Universitario de Valencia, INCLIVA, Valencia, Spain CIBERSAM, Spanish National Network for Research in Mental Health, Madrid, Spain
| |
Collapse
|
48
|
The neural plasticity theory of depression: assessing the roles of adult neurogenesis and PSA-NCAM within the hippocampus. Neural Plast 2013; 2013:805497. [PMID: 23691371 PMCID: PMC3649690 DOI: 10.1155/2013/805497] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Accepted: 03/13/2013] [Indexed: 01/01/2023] Open
Abstract
Depression is a devastating and prevalent disease, with profound effects on neural structure and function; however the etiology and neuropathology of depression remain poorly understood. Though antidepressant drugs exist, they are not ideal, as only a segment of patients are effectively treated, therapeutic onset is delayed, and the exact mechanism of these drugs remains to be elucidated. Several theories of depression do exist, including modulation of monoaminergic neurotransmission, alterations in neurotrophic factors, and the upregulation of adult hippocampal neurogenesis, and are briefly mentioned in the review. However none of these theories sufficiently explains the pathology and treatment of depression unto itself. Recently, neural plasticity theories of depression have postulated that multiple aspects of brain plasticity, beyond neurogenesis, may bridge the prevailing theories. The term “neural plasticity” encompasses an array of mechanisms, from the birth, survival, migration, and integration of new neurons to neurite outgrowth, synaptogenesis, and the modulation of mature synapses. This review critically assesses the role of adult hippocampal neurogenesis and the cell adhesion molecule, PSA-NCAM (which is known to be involved in many facets of neural plasticity), in depression and antidepressant treatment.
Collapse
|
49
|
Theis T, Mishra B, von der Ohe M, Loers G, Prondzynski M, Pless O, Blackshear PJ, Schachner M, Kleene R. Functional role of the interaction between polysialic acid and myristoylated alanine-rich C kinase substrate at the plasma membrane. J Biol Chem 2013; 288:6726-42. [PMID: 23329829 PMCID: PMC3585110 DOI: 10.1074/jbc.m112.444034] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Indexed: 12/14/2022] Open
Abstract
Polysialic acid (PSA) is a homopolymeric glycan that plays crucial roles in the developing and adult nervous system. So far only a few PSA-binding proteins have been identified. Here, we identify myristoylated alanine-rich C kinase substrate (MARCKS) as novel PSA binding partner. Binding assays showed a direct interaction between PSA and a peptide comprising the effector domain of MARCKS (MARCKS-ED). Co-immunoprecipitation of PSA-carrying neural cell adhesion molecule (PSA-NCAM) with MARCKS and co-immunostaining of MARCKS and PSA at the cell membrane of hippocampal neurons confirm the interaction between PSA and MARCKS. Co-localization and an intimate interaction of PSA and MARCKS at the cell surface was seen by confocal microscopy and fluorescence resonance energy transfer (FRET) analysis after the addition of fluorescently labeled PSA or PSA-NCAM to live CHO cells or hippocampal neurons expressing MARCKS as a fusion protein with green fluorescent protein (GFP). Cross-linking experiments showed that extracellularly applied PSA or PSA-NCAM and intracellularly expressed MARCKS-GFP are in close contact, suggesting that PSA and MARCKS interact with each other at the plasma membrane from opposite sides. Insertion of PSA and MARCKS-ED peptide into lipid bilayers from opposite sides alters the electric properties of the bilayer confirming the notion that PSA and the effector domain of MARCKS interact at and/or within the plane of the membrane. The MARCKS-ED peptide abolished PSA-induced enhancement of neurite outgrowth from cultured hippocampal neurons indicating an important functional role for the interaction between MARCKS and PSA in the developing and adult nervous system.
Collapse
Affiliation(s)
- Thomas Theis
- From the Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Bibhudatta Mishra
- From the Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Maren von der Ohe
- From the Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Gabriele Loers
- From the Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, 20246 Hamburg, Germany
| | | | - Ole Pless
- European Screening Port GmbH, Schnackenburgallee 114, 22525 Hamburg, Germany
| | - Perry J. Blackshear
- the Departments of Medicine and Biochemistry, Duke University, Durham, North Carolina 27709
- Laboratory of Signal Transduction, NIEHS, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, and
| | - Melitta Schachner
- From the Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, 20246 Hamburg, Germany
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey 08854
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou 515041, China
| | - Ralf Kleene
- From the Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, 20246 Hamburg, Germany
| |
Collapse
|
50
|
Maroto M, Fernández-Morales JC, Padín JF, González JC, Hernández-Guijo JM, Montell E, Vergés J, de Diego AMG, García AG. Chondroitin sulfate, a major component of the perineuronal net, elicits inward currents, cell depolarization, and calcium transients by acting on AMPA and kainate receptors of hippocampal neurons. J Neurochem 2013; 125:205-13. [PMID: 23350646 DOI: 10.1111/jnc.12159] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Revised: 10/08/2012] [Accepted: 01/15/2013] [Indexed: 12/19/2022]
Abstract
Chondroitin sulfate (CS) proteoglycans (CSPGs) are the most abundant PGs of the brain extracellular matrix (ECM). Free CS could be released during ECM degradation and exert physiological functions; thus, we aimed to investigate the effects of CS on voltage- and current-clamped rat embryo hippocampal neurons in primary cultures. We found that CS elicited a whole-cell Na(+)-dependent inward current (ICS) that produced drastic cell depolarization, and a cytosolic calcium transient ([Ca(2+)]c). Those effects were similar to those elicited by α-amino-3-hydroxy-5-methylisoxazole-4-propionate (AMPA) and kainate, were completely blocked by NBQX and CNQX, were partially blocked by GYKI, and were unaffected by MK801 and D-APV. Furthermore, ICS and AMPA currents were similarly potentiated by cyclothiazide, a positive allosteric modulator of AMPA receptors. Because CSPGs have been attributed Ca(2) (+) -dependent roles, such as neural network development, axon pathfinding, plasticity and regeneration after CNS injury, CS action after ECM degradation could be contributing to the mediation of these effects through its interaction with AMPA and kainate receptors.
Collapse
Affiliation(s)
- Marcos Maroto
- Instituto Teófilo Hernando, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain.,Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - José-Carlos Fernández-Morales
- Instituto Teófilo Hernando, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain.,Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Juan Fernando Padín
- Instituto Teófilo Hernando, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain.,Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - José C González
- Instituto Teófilo Hernando, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain.,Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Jesús M Hernández-Guijo
- Instituto Teófilo Hernando, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain.,Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Eulalia Montell
- Pre-Clinical R&D Area, Pharmascience Division, Bioibérica, Barcelona, Spain
| | - Josep Vergés
- Pre-Clinical R&D Area, Pharmascience Division, Bioibérica, Barcelona, Spain
| | - Antonio M G de Diego
- Instituto Teófilo Hernando, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain.,Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Antonio G García
- Instituto Teófilo Hernando, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain.,Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria, Servicio de Farmacología Clínica, Hospital Universitario de la Princesa. Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|