1
|
Abdulla A, Yan H, Chen S, Wu L, Chen XS, Zhang Y, Zhang M, Zhuang TY, Ahmad KZ, Lin J, Ding X, Jiang L. A multichannel microfluidic device for revealing the neurotoxic effects of Bisphenol S on cerebral organoids under low-dose constant exposure. Biosens Bioelectron 2025; 267:116754. [PMID: 39332252 DOI: 10.1016/j.bios.2024.116754] [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: 06/11/2024] [Revised: 08/20/2024] [Accepted: 09/04/2024] [Indexed: 09/29/2024]
Abstract
Bisphenol S is a widely used plasticizer in manufacturing daily supplies, while little was known about its adverse effect on human health, especially on fetal brain development. Due to the complexity and subtlety of the brain, it remains challenging to reveal the hazardous effects of environmental pollution on human fetal brain development. Taking advantage of stem cell application, cerebral organoids generated from stem cells are becoming powerful tools for understanding brain development and drug toxicity testing models. Here, we developed a microfluidic chip for cerebral organoid culturing to reveal the neurotoxicity of low-dose constant BPS exposure on cerebral organoids. The organoids in our microfluidic system could be continuously cultured for 34 days and expressed all the essential properties of the cerebral organoids. Exposure to BPS was initiated from day 20 for concessive two weeks. The neurotoxic effects were evaluated by immunofluorescence staining and proteomics, and verified by quantitative real-time PCR. Our results indicated BPS exposure would inhibit neuron differentiation, hinder the Wnt signaling pathway, and cause alteration of signaling molecule expressions in brain regionalization. Even exposure to a low dose of BPS constantly might cause neurotoxicity during fetal brain development. Altogether, the multichannel microfluidic chip offers a general platform technique to reveal the effects of different hazardous chemicals on cerebral organoids.
Collapse
Affiliation(s)
- Aynur Abdulla
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200092, China; State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Haoni Yan
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200092, China
| | - Shujin Chen
- Ministry of Education, Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200092, China
| | - Leqi Wu
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Xu-Sen Chen
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Yizhi Zhang
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Manlin Zhang
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Tsz Yui Zhuang
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Khan Zara Ahmad
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institute, Sweden
| | - Jinjin Lin
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China; School of Psychology, Shaanxi Normal University, Xi' an 710062, Shaanxi Province, China
| | - Xianting Ding
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200092, China; State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China.
| | - Lai Jiang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200092, China.
| |
Collapse
|
2
|
Pavon MV, Navakkode S, Sajikumar S. Nogo-A-mediated constraints on activity-dependent synaptic plasticity and associativity in rat hippocampal CA2 synapses. Hippocampus 2024; 34:491-502. [PMID: 39091158 DOI: 10.1002/hipo.23625] [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: 12/02/2023] [Revised: 06/13/2024] [Accepted: 07/15/2024] [Indexed: 08/04/2024]
Abstract
Hippocampal area CA2 has garnered attention in recent times owing to its significant involvement in social memory and distinctive plasticity characteristics. Research has revealed that the CA2 region demonstrates a remarkable resistance to plasticity, particularly in the Schaffer Collateral (SC)-CA2 pathway. In this study we investigated the role of Nogo-A, a well-known axon growth inhibitor and more recently discovered plasticity regulator, in modulating plasticity within the CA2 region. The findings demonstrate that blocking Nogo-A in male rat hippocampal slices facilitates the establishment of both short-term and long-term plasticity in the SC-CA2 pathway, while having no impact on the Entorhinal Cortical (EC)-CA2 pathway. Additionally, the study reveals that inhibiting Nogo-A enables association between the SC and EC pathways. Mechanistically, we confirm that Nogo-A operates through its well-known co-receptor, p75 neurotrophin receptor (p75NTR), and its downstream signaling factor such as Rho-associated protein kinase (ROCK), as their inhibition also allows plasticity induction in the SC-CA2 pathway. Additionally, the induction of long-term depression (LTD) in both the EC and SC-CA2 pathways led to persistent LTD, which was not affected by Nogo-A inhibition. Our study demonstrates the involvement of Nogo-A mediated signaling mechanisms in limiting synaptic plasticity within the CA2 region.
Collapse
Affiliation(s)
- Maria Vazquez Pavon
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Neurobiology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Sheeja Navakkode
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Sreedharan Sajikumar
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Neurobiology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
3
|
Rust R, Holm MM, Egger M, Weinmann O, van Rossum D, Walter FR, Santa-Maria AR, Grönnert L, Maurer MA, Kraler S, Akhmedov A, Cideciyan R, Lüscher TF, Deli MA, Herrmann IK, Schwab ME. Nogo-A is secreted in extracellular vesicles, occurs in blood and can influence vascular permeability. J Cereb Blood Flow Metab 2024; 44:938-954. [PMID: 38000040 PMCID: PMC11318402 DOI: 10.1177/0271678x231216270] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 10/10/2023] [Accepted: 10/20/2023] [Indexed: 11/26/2023]
Abstract
Nogo-A is a transmembrane protein with multiple functions in the central nervous system (CNS), including restriction of neurite growth and synaptic plasticity. Thus far, Nogo-A has been predominantly considered a cell contact-dependent ligand signaling via cell surface receptors. Here, we show that Nogo-A can be secreted by cultured cells of neuronal and glial origin in association with extracellular vesicles (EVs). Neuron- and oligodendrocyte-derived Nogo-A containing EVs inhibited fibroblast spreading, and this effect was partially reversed by Nogo-A receptor S1PR2 blockage. EVs purified from HEK cells only inhibited fibroblast spreading upon Nogo-A over-expression. Nogo-A-containing EVs were found in vivo in the blood of healthy mice and rats, as well as in human plasma. Blood Nogo-A concentrations were elevated after acute stroke lesions in mice and rats. Nogo-A active peptides decreased barrier integrity in an in vitro blood-brain barrier model. Stroked mice showed increased dye permeability in peripheral organs when tested 2 weeks after injury. In the Miles assay, an in vivo test to assess leakage of the skin vasculature, a Nogo-A active peptide increased dye permeability. These findings suggest that blood borne, possibly EV-associated Nogo-A could exert long-range regulatory actions on vascular permeability.
Collapse
Affiliation(s)
- Ruslan Rust
- Brain Research Institute, University of Zürich, Switzerland
- Department of Health Sciences and Technology, ETH Zürich, Switzerland
- Institute for Regenerative Medicine (IREM), University of Zurich, Switzerland
| | - Mea M Holm
- Brain Research Institute, University of Zürich, Switzerland
- Department of Health Sciences and Technology, ETH Zürich, Switzerland
| | - Matteo Egger
- Department of Health Sciences and Technology, ETH Zürich, Switzerland
| | | | | | - Fruzsina R Walter
- Biological Barriers Research Group, ELKH Biological Research Centre, Szeged, Hungary
| | | | - Lisa Grönnert
- Brain Research Institute, University of Zürich, Switzerland
| | | | - Simon Kraler
- Center for Molecular Cardiology, University of Zurich, Switzerland
| | | | - Rose Cideciyan
- Center for Molecular Cardiology, University of Zurich, Switzerland
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zurich, Switzerland
- Royal Brompton and Harefield Hospitals and Imperial College, London, United Kingdom
| | - Maria A Deli
- Biological Barriers Research Group, ELKH Biological Research Centre, Szeged, Hungary
| | - Inge K Herrmann
- Particles Biology Interactions Laboratory, Swiss Federal Laboratories for Materials Science and Technology (Empa), St. Gallen, Switzerland
- Nanoparticle Systems Engineering Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, Zurich, Switzerland
| | - Martin E Schwab
- Brain Research Institute, University of Zürich, Switzerland
- Department of Health Sciences and Technology, ETH Zürich, Switzerland
- Institute for Regenerative Medicine (IREM), University of Zurich, Switzerland
| |
Collapse
|
4
|
Joly S, Augusto G, Mdzomba B, Meli I, Vogel M, Chan A, Pernet V. Nogo-A neutralization in the central nervous system with a blood-brain barrier-penetrating antibody. J Control Release 2024; 366:52-64. [PMID: 38154541 DOI: 10.1016/j.jconrel.2023.12.041] [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: 08/11/2023] [Revised: 11/26/2023] [Accepted: 12/24/2023] [Indexed: 12/30/2023]
Abstract
The poor penetration of monoclonal antibodies (mAb) across the blood-brain barrier (BBB) impedes the development of regenerative therapies for neurological diseases. For example, Nogo-A is a myelin-associated protein highly expressed in the central nervous system (CNS) whose inhibitory effects on neuronal plasticity can be neutralized with direct administration of 11C7 mAb in CNS tissues/fluids, but not with peripheral administrations such as intravenous injections. Therefore, in the present study, we engineered a CNS-penetrating antibody against Nogo-A by combining 11C7 mAb and the single-chain variable fragment (scFv) of 8D3, a rat antibody binding transferrin receptor 1 (TfR) and mediating BBB transcytosis (11C7-scFv8D3). The binding of 11C7-scFv8D3 to Nogo-A and to TfR/CD71 was validated by capture ELISA and Biolayer Interferometry. After intravenous injection in mice, capture ELISA measurements revealed fast plasma clearance of 11C7-scFv8D3 concomitantly with brain and spinal cord accumulation at levels up to 19 fold as high as those of original 11C7 mAb. 11C7-scFv8D3 detection in the parenchyma indicated effective blood-to-CNS transfer. A single dose of 11C7-scFv8D3 induced stronger activation of the growth-promoting AkT/mTOR/S6 signaling pathway than 11C7 mAb or control antibody. Taken together, our results show that BBB-crossing 11C7-scFv8D3 engages Nogo-A in the mouse CNS and stimulates neuronal growth mechanisms.
Collapse
Affiliation(s)
- Sandrine Joly
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Switzerland; Center for experimental neurology (ZEN), Bern University Hospital, University of Bern, Switzerland; Centre de recherche du CHU de Québec-Université Laval and Department of Molecular Medicine, Faculté de médecine, Université Laval, Québec, Québec, Canada; Department of Ophthalmology, Bern University Hospital, University of Bern, Switzerland; Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Gilles Augusto
- Department of Biomedical Research, University of Bern, Bern, Switzerland; Department of Immunology, Inselspital, Bern University Hospital, University of Bern, Switzerland; Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Baya Mdzomba
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Switzerland; Center for experimental neurology (ZEN), Bern University Hospital, University of Bern, Switzerland; Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Ivo Meli
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Switzerland; Center for experimental neurology (ZEN), Bern University Hospital, University of Bern, Switzerland; Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Monique Vogel
- Department of Biomedical Research, University of Bern, Bern, Switzerland; Department of Immunology, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Andrew Chan
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Switzerland; Center for experimental neurology (ZEN), Bern University Hospital, University of Bern, Switzerland; Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Vincent Pernet
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Switzerland; Center for experimental neurology (ZEN), Bern University Hospital, University of Bern, Switzerland; Centre de recherche du CHU de Québec-Université Laval and Department of Molecular Medicine, Faculté de médecine, Université Laval, Québec, Québec, Canada; Department of Biomedical Research, University of Bern, Bern, Switzerland.
| |
Collapse
|
5
|
Addison RN, Van Gemmert AWA. Bilateral Transfer of a Visuomotor Task in Different Workspace Configurations. J Mot Behav 2023; 56:290-304. [PMID: 38108224 DOI: 10.1080/00222895.2023.2293002] [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: 05/25/2022] [Accepted: 11/19/2023] [Indexed: 12/19/2023]
Abstract
Bilateral transfer occurs when a learned behavior transfers from one (group of) effectors(s) to another. Researchers investigating bilateral transfer of a visuomotor adaptation task between limbs used across workspaces have observed divergent results. This study assessed whether bilateral transfer of a visuomotor adaptation task changes with workspace configuration manipulation. Ninety-six right-handed young adults were assigned to one of three workspace locations, i.e., ipsilateral, contralateral, and central. Within each workspace were two retention groups (RRR/LLL) and two bilateral transfer groups (RLR/LRL). Performance before and after training was collected to determine direct and after-effects. We observed an asymmetric transfer of pathlength (left to right) but no ensuing after-effect. However, the transfer of movement time and normalized jerk was symmetric in the contralateral workspace. These findings showed differences in the pattern of bilateral transfer asymmetry in the different workspace configurations, which was parameter specific.
Collapse
Affiliation(s)
- Reuben N Addison
- School of Kinesiology, Louisiana State University, Baton Rouge, LA, USA
- MGH Institute of Health Professions, Charlestown Navy Yard, Boston, MA, USA
| | | |
Collapse
|
6
|
Kogan E, Lu J, Zuo Y. Cortical circuit dynamics underlying motor skill learning: from rodents to humans. Front Mol Neurosci 2023; 16:1292685. [PMID: 37965043 PMCID: PMC10641381 DOI: 10.3389/fnmol.2023.1292685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 10/11/2023] [Indexed: 11/16/2023] Open
Abstract
Motor learning is crucial for the survival of many animals. Acquiring a new motor skill involves complex alterations in both local neural circuits in many brain regions and long-range connections between them. Such changes can be observed anatomically and functionally. The primary motor cortex (M1) integrates information from diverse brain regions and plays a pivotal role in the acquisition and refinement of new motor skills. In this review, we discuss how motor learning affects the M1 at synaptic, cellular, and circuit levels. Wherever applicable, we attempt to relate and compare findings in humans, non-human primates, and rodents. Understanding the underlying principles shared by different species will deepen our understanding of the neurobiological and computational basis of motor learning.
Collapse
Affiliation(s)
| | | | - Yi Zuo
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA, United States
| |
Collapse
|
7
|
Liu HL, Lu XM, Wang HY, Hu KB, Wu QY, Liao P, Li S, Long ZY, Wang YT. The role of RNA splicing factor PTBP1 in neuronal development. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119506. [PMID: 37263298 DOI: 10.1016/j.bbamcr.2023.119506] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 05/23/2023] [Accepted: 05/26/2023] [Indexed: 06/03/2023]
Abstract
Alternative pre-mRNA splicing, which produces various mRNA isoforms with distinct structures and functions from a single gene, is regulated by specific RNA-binding proteins and is an essential method for regulating gene expression in mammals. Recent studies have shown that abnormal change during neuronal development triggered by splicing mis-regulation is an important feature of various neurological diseases. Polypyrimidine tract binding protein 1 (PTBP1) is a kind of RNA-binding proteins with extensive biological functions. As a well-known splicing regulator, it affects the neuronal development process through its involvement in axon formation, synaptogenesis, and neuronal apoptosis, according to the most recent studies. Here, we summarized the mechanism of alternative splicing, structure and function of PTBP1, and the latest research progress on the role of alternative splicing events regulated by PTBP1 in axon formation, synaptogenesis and neuronal apoptosis, to reveal the mechanism of PTBP1-regulated changes in neuronal development process.
Collapse
Affiliation(s)
- Hui-Lin Liu
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, PR China; State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, PR China
| | - Xiu-Min Lu
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, PR China
| | - Hai-Yan Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, PR China
| | - Kai-Bin Hu
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, PR China
| | - Qing-Yun Wu
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, PR China
| | - Ping Liao
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, PR China
| | - Sen Li
- State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, PR China
| | - Zai-Yun Long
- State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, PR China
| | - Yong-Tang Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, PR China.
| |
Collapse
|
8
|
Glotfelty EJ, Hsueh SC, Claybourne Q, Bedolla A, Kopp KO, Wallace T, Zheng B, Luo Y, Karlsson TE, McDevitt RA, Olson L, Greig NH. Microglial Nogo delays recovery following traumatic brain injury in mice. Glia 2023; 71:2473-2494. [PMID: 37401784 PMCID: PMC10528455 DOI: 10.1002/glia.24436] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/16/2023] [Accepted: 06/20/2023] [Indexed: 07/05/2023]
Abstract
Nogo-A, B, and C are well described members of the reticulon family of proteins, most well known for their negative regulatory effects on central nervous system (CNS) neurite outgrowth and repair following injury. Recent research indicates a relationship between Nogo-proteins and inflammation. Microglia, the brain's immune cells and inflammation-competent compartment, express Nogo protein, although specific roles of the Nogo in these cells is understudied. To examine inflammation-related effects of Nogo, we generated a microglial-specific inducible Nogo KO (MinoKO) mouse and challenged the mouse with a controlled cortical impact (CCI) traumatic brain injury (TBI). Histological analysis shows no difference in brain lesion sizes between MinoKO-CCI and Control-CCI mice, although MinoKO-CCI mice do not exhibit the levels of ipsilateral lateral ventricle enlargement as injury matched controls. Microglial Nogo-KO results in decreased lateral ventricle enlargement, microglial and astrocyte immunoreactivity, and increased microglial morphological complexity compared to injury matched controls, suggesting decreased tissue inflammation. Behaviorally, healthy MinoKO mice do not differ from control mice, but automated tracking of movement around the home cage and stereotypic behavior, such as grooming and eating (termed cage "activation"), following CCI is significantly elevated. Asymmetrical motor function, a deficit typical of unilaterally brain lesioned rodents, was not detected in CCI injured MinoKO mice, while the phenomenon was present in CCI injured controls 1-week post-injury. Overall, our studies show microglial Nogo as a negative regulator of recovery following brain injury. To date, this is the first evaluation of the roles microglial specific Nogo in a rodent injury model.
Collapse
Affiliation(s)
- Elliot J. Glotfelty
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, NIH, Baltimore, MD 21224, USA
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Shih-Chang Hsueh
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Quia Claybourne
- Comparative Medicine Section, National Institute on Aging, NIH, Baltimore, Maryland 21224, USA
| | - Alicia Bedolla
- Department of Molecular Genetics and Biochemistry, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Katherine O. Kopp
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Tonya Wallace
- Flow Cytometry Unit, National Institute on Aging, Baltimore, MD, USA
| | - Binhai Zheng
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Yu Luo
- Department of Molecular Genetics and Biochemistry, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | | | - Ross A. McDevitt
- Comparative Medicine Section, National Institute on Aging, NIH, Baltimore, Maryland 21224, USA
| | - Lars Olson
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Nigel H. Greig
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, NIH, Baltimore, MD 21224, USA
| |
Collapse
|
9
|
Inoue R, Nishimune H. Neuronal Plasticity and Age-Related Functional Decline in the Motor Cortex. Cells 2023; 12:2142. [PMID: 37681874 PMCID: PMC10487126 DOI: 10.3390/cells12172142] [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: 07/16/2023] [Revised: 08/16/2023] [Accepted: 08/23/2023] [Indexed: 09/09/2023] Open
Abstract
Physiological aging causes a decline of motor function due to impairment of motor cortex function, losses of motor neurons and neuromuscular junctions, sarcopenia, and frailty. There is increasing evidence suggesting that the changes in motor function start earlier in the middle-aged stage. The mechanism underlining the middle-aged decline in motor function seems to relate to the central nervous system rather than the peripheral neuromuscular system. The motor cortex is one of the responsible central nervous systems for coordinating and learning motor functions. The neuronal circuits in the motor cortex show plasticity in response to motor learning, including LTP. This motor cortex plasticity seems important for the intervention method mechanisms that revert the age-related decline of motor function. This review will focus on recent findings on the role of plasticity in the motor cortex for motor function and age-related changes. The review will also introduce our recent identification of an age-related decline of neuronal activity in the primary motor cortex of middle-aged mice using electrophysiological recordings of brain slices.
Collapse
Affiliation(s)
- Ritsuko Inoue
- Laboratory of Neurobiology of Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakaecho, Itabashi-ku, Tokyo 173-0015, Japan;
| | - Hiroshi Nishimune
- Laboratory of Neurobiology of Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakaecho, Itabashi-ku, Tokyo 173-0015, Japan;
- Department of Applied Biological Science, Tokyo University of Agriculture and Technology, 3-8-1 Harumicho, Fuchu-shi, Tokyo 183-8538, Japan
| |
Collapse
|
10
|
Pavon MV, Navakkode S, Wong LW, Sajikumar S. Inhibition of Nogo-A rescues synaptic plasticity and associativity in APP/PS1 animal model of Alzheimer's disease. Semin Cell Dev Biol 2023; 139:111-120. [PMID: 35431138 DOI: 10.1016/j.semcdb.2022.04.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 03/30/2022] [Accepted: 04/06/2022] [Indexed: 12/31/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by memory loss and cognitive decline. Synaptic impairment is one of the first events to occur in the progression of this disease. Synaptic plasticity and cellular association of various plastic events have been shown to be affected in AD models. Nogo-A, a well-known axonal growth inhibitor with a recently discovered role as a plasticity suppressor, and its main receptor Nogo-66 receptor 1 (NGR1) have been found to be overexpressed in the hippocampus of Alzheimer's patients. However, the role of Nogo-A and its receptor in the pathology of AD is still widely unknown. In this work we set out to investigate whether Nogo-A is working as a plasticity suppressor in AD. Our results show that inhibition of the Nogo-A pathway via the Nogo-R antibody in an Alzheimer's mouse model, APP/PS1, leads to the restoration of both synaptic plasticity and associativity in a protein synthesis and NMDR-dependent manner. We also show that inhibition of the p75NTR pathway, which is strongly associated with NGR1, restores synaptic plasticity as well. Mechanistically, we propose that the restoration of synaptic plasticity in APP/PS1 via inhibition of the Nogo-A pathway is due to the modulation of the RhoA-ROCK2 pathway and increase in plasticity related proteins. Our study identifies Nogo-A as a plasticity suppressor in AD models hence targeting Nogo-A could be a promising strategy to understanding AD pathology.
Collapse
Affiliation(s)
- Maria Vazquez Pavon
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; Life Sciences Institute, Neurobiology Programme, National University of Singapore, Singapore 117456, Singapore
| | - Sheeja Navakkode
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Lik-Wei Wong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; Life Sciences Institute, Neurobiology Programme, National University of Singapore, Singapore 117456, Singapore
| | - Sreedharan Sajikumar
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; Life Sciences Institute, Neurobiology Programme, National University of Singapore, Singapore 117456, Singapore; Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore.
| |
Collapse
|
11
|
Dave BP, Shah KC, Shah MB, Chorawala MR, Patel VN, Shah PA, Shah GB, Dhameliya TM. Unveiling the modulation of Nogo receptor in neuroregeneration and plasticity: Novel aspects and future horizon in a new frontier. Biochem Pharmacol 2023; 210:115461. [PMID: 36828272 DOI: 10.1016/j.bcp.2023.115461] [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: 12/21/2022] [Revised: 02/16/2023] [Accepted: 02/17/2023] [Indexed: 02/25/2023]
Abstract
Neurodegenerative diseases (NDs) such as Alzheimer's, Parkinson's, Multiple Sclerosis, Hereditary Spastic Paraplegia, and Amyotrophic Lateral Sclerosis have emerged as the most dreaded diseases due to a lack of precise diagnostic tools and efficient therapies. Despite the fact that the contributing factors of NDs are still unidentified, mounting evidence indicates the possibility that genetic and cellular changes may lead to the significant production of abnormally misfolded proteins. These misfolded proteins lead to damaging effects thereby causing neurodegeneration. The association between Neurite outgrowth factor (Nogo) with neurological diseases and other peripheral diseases is coming into play. Three isoforms of Nogo have been identified Nogo-A, Nogo-B and Nogo-C. Among these, Nogo-A is mainly responsible for neurological diseases as it is localized in the CNS (Central Nervous System), whereas Nogo-B and Nogo-C are responsible for other diseases such as colitis, lung, intestinal injury, etc. Nogo-A, a membrane protein, had first been described as a CNS-specific inhibitor of axonal regeneration. Several recent studies have revealed the role of Nogo-A proteins and their receptors in modulating neurite outgrowth, branching, and precursor migration during nervous system development. It may also modulate or affect the inhibition of growth during the developmental processes of the CNS. Information about the effects of other ligands of Nogo protein on the CNS are yet to be discovered however several pieces of evidence have suggested that it may also influence the neuronal maturation of CNS and targeting Nogo-A could prove to be beneficial in several neurodegenerative diseases.
Collapse
Affiliation(s)
- Bhavarth P Dave
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Ahmedabad 380009, Gujarat, India
| | - Kashvi C Shah
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Ahmedabad 380009, Gujarat, India
| | - Maitri B Shah
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Ahmedabad 380009, Gujarat, India
| | - Mehul R Chorawala
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Ahmedabad 380009, Gujarat, India.
| | - Vishvas N Patel
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Ahmedabad 380009, Gujarat, India
| | - Palak A Shah
- Department of Pharmacology, K. B. Institute of Pharmaceutical Education and Research, Gandhinagar 380023, Gujarat, India
| | - Gaurang B Shah
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Ahmedabad 380009, Gujarat, India
| | - Tejas M Dhameliya
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad-382481, Gujarat, India
| |
Collapse
|
12
|
Inoue R, Miura M, Yanai S, Nishimune H. Coenzyme Q 10 supplementation improves the motor function of middle-aged mice by restoring the neuronal activity of the motor cortex. Sci Rep 2023; 13:4323. [PMID: 36922562 PMCID: PMC10017826 DOI: 10.1038/s41598-023-31510-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 03/13/2023] [Indexed: 03/18/2023] Open
Abstract
Physiological aging causes motor function decline and anatomical and biochemical changes in the motor cortex. We confirmed that middle-aged mice at 15-18 months old show motor function decline, which can be restored to the young adult level by supplementing with mitochondrial electron transporter coenzyme Q10 (CoQ10) as a water-soluble nanoformula by drinking water for 1 week. CoQ10 supplementation concurrently improved brain mitochondrial respiration but not muscle strength. Notably, we identified an age-related decline in field excitatory postsynaptic potential (fEPSP) amplitude in the pathway from layers II/III to V of the primary motor area of middle-aged mice, which was restored to the young adult level by supplementing with CoQ10 for 1 week but not by administering CoQ10 acutely to brain slices. Interestingly, CoQ10 with high-frequency stimulation induced NMDA receptor-dependent long-term potentiation (LTP) in layer V of the primary motor cortex of middle-aged mice. Importantly, the fEPSP amplitude showed a larger input‒output relationship after CoQ10-dependent LTP expression. These data suggest that CoQ10 restores the motor function of middle-aged mice by improving brain mitochondrial function and the basal fEPSP level of the motor cortex, potentially by enhancing synaptic plasticity efficacy. Thus, CoQ10 supplementation may ameliorate the age-related decline in motor function in humans.
Collapse
Affiliation(s)
- Ritsuko Inoue
- Laboratory of Neurobiology of Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakaecho, Itabashi-Ku, Tokyo, 173-0015, Japan.
| | - Masami Miura
- Laboratory of Neurobiology of Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakaecho, Itabashi-Ku, Tokyo, 173-0015, Japan.,Saitama Central Hospital, 2177 Kamitome, Miyoshicho, Iruma-Gun, Saitama, 354-0045, Japan
| | - Shuichi Yanai
- Laboratory of Memory Neuroscience, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakaecho, Itabashi-Ku, Tokyo, 173-0015, Japan
| | - Hiroshi Nishimune
- Laboratory of Neurobiology of Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakaecho, Itabashi-Ku, Tokyo, 173-0015, Japan. .,Department of Applied Biological Science, Tokyo University of Agriculture and Technology, 3-8-1 Harumicho, Fuchu-Shi, Tokyo, 183-8538, Japan.
| |
Collapse
|
13
|
Intranasal delivery of full-length anti-Nogo-A antibody: A potential alternative route for therapeutic antibodies to central nervous system targets. Proc Natl Acad Sci U S A 2023; 120:e2200057120. [PMID: 36649432 PMCID: PMC9942809 DOI: 10.1073/pnas.2200057120] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Antibody delivery to the CNS remains a huge hurdle for the clinical application of antibodies targeting a CNS antigen. The blood-brain barrier and blood-CSF barrier restrict access of therapeutic antibodies to their CNS targets in a major way. The very high amounts of therapeutic antibodies that are administered systemically in recent clinical trials to reach CNS targets are barely viable cost-wise for broad, routine applications. Though global CNS delivery of antibodies can be achieved by intrathecal application, these procedures are invasive. A non-invasive method to bring antibodies into the CNS reliably and reproducibly remains an important unmet need in neurology. In the present study, we show that intranasal application of a mouse monoclonal antibody against the neurite growth-inhibiting and plasticity-restricting membrane protein Nogo-A leads to a rapid transfer of significant amounts of antibody to the brain and spinal cord in intact adult rats. Daily intranasal application for 2 wk of anti-Nogo-A antibody enhanced growth and compensatory sprouting of corticofugal projections and functional recovery in rats after large unilateral cortical strokes. These findings are a starting point for clinical translation for a less invasive route of application of therapeutic antibodies to CNS targets for many neurological indications.
Collapse
|
14
|
Del Rey NLG, García-Cabezas MÁ. Cytology, architecture, development, and connections of the primate striatum: Hints for human pathology. Neurobiol Dis 2023; 176:105945. [PMID: 36481436 DOI: 10.1016/j.nbd.2022.105945] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 11/19/2022] [Accepted: 12/03/2022] [Indexed: 12/10/2022] Open
Abstract
Degeneration of neurons and circuits across the striatum shows stereotyped time-course and spatial topography patterns that are distinct for Huntington's disease, Parkinson's disease, or the Tauopathies. These patterns of neurodegeneration in humans have not yet been systematically related to developmental, connectional, cellular, and chemical factors studied in human and non-human primates, that may underlie potential differences in selective vulnerability across striatal sectors. Relating primate anatomy to human pathology could provide new venues for identifying molecular, cellular, and connectional factors linked to the degeneration of striatal neurons and circuits. This review describes and summarizes several developmental, cellular, structural, and connectional features of the primate striatum in relation to patterns of neurodegeneration in the striatum of humans and of non-human primate models. We review (1) the types of neurons in the primate striatum, (2) the cyto-, myelo-, and chemoarchitecture of the primate striatum, (3) the developmental origin of the striatum in light of modern patterning studies, (4) the organization of corticostriatal projections in relation to cortical types, and (5) the topography and time-course of neuron loss, glial reaction, and protein aggregation induced by neurodegenerative diseases in humans and in non-human primate models across striatal sectors and their corresponding cortical areas. We summarize current knowledge about key aspects of primate striatal anatomy and human pathology and indicate knowledge gaps that should be addressed in future studies. We aim to identify factors for selective vulnerability to neurodegeneration of striatal neurons and circuits and obtain hints that could help elucidate striatal pathology in humans.
Collapse
Affiliation(s)
- Natalia López-González Del Rey
- PhD Program in Neuroscience UAM-Cajal; Madrid, Spain; HM CINAC (Centro Integral de Neurociencias Abarca Campal). Hospital Universitario HM Puerta del Sur. HM Hospitales. Madrid, Spain
| | - Miguel Ángel García-Cabezas
- PhD Program in Neuroscience UAM-Cajal; Madrid, Spain; Departamento de Anatomía, Histología y Neurociencia, Facultad de Medicina, Universidad Autónoma de Madrid; Madrid, Spain.
| |
Collapse
|
15
|
Kulczyńska-Przybik A, Dulewicz M, Słowik A, Borawska R, Kułakowska A, Kochanowicz J, Mroczko B. The Clinical Significance of Cerebrospinal Fluid Reticulon 4 (RTN4) Levels in the Differential Diagnosis of Neurodegenerative Diseases. J Clin Med 2021; 10:jcm10225281. [PMID: 34830564 PMCID: PMC8622503 DOI: 10.3390/jcm10225281] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/09/2021] [Accepted: 11/11/2021] [Indexed: 11/16/2022] Open
Abstract
Neurodegenerative diseases (NDs) belong to the top global causes of mortality. Diagnostic approaches to improve early diagnosis and differentiation of these diseases are constantly being sought. Therefore, we aimed to assess the cerebrospinal fluid (CSF) concentrations of Reticulon 4 (RTN4) in patients with neurodegenerative diseases and evaluate the potential clinical usefulness of this protein. RTNs are transmembrane proteins mediating neuroanatomical plasticity and functional recovery after central nervous system injury or diseases. According to our best knowledge, this is the first investigation providing the data concerning the dynamic of CSF RTN4 protein levels in patients with different NDs. Methods: Overall, 77 newly diagnosed patients with neurodegenerative diseases, including Alzheimer’s disease (AD), Parkinson’s disease (PD), and multiple sclerosis (MS), as well as 21 controls, were enrolled in the study. The CSF concentrations of tested proteins were assessed using immunological assays. Results: We revealed significantly higher CSF RTN4A levels in patients with AD, PD, and MS in comparison to the controls. Moreover, the comparative analysis of RTN4 concentration between different neurodegenerative diseases revealed the highest concentration of RTN4A in AD patients and a statistically significant difference between AD vs. PD, and AD vs. MS groups. The increased CSF level of the protein correlated with Tau, and pTau181 proteins in AD as well as in PD patients. Conclusions: Our study presents a previously not identified clinical utility of RTN4 in the differential diagnosis of neurodegenerative diseases.
Collapse
Affiliation(s)
- Agnieszka Kulczyńska-Przybik
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland; (M.D.); (R.B.); (B.M.)
- Correspondence:
| | - Maciej Dulewicz
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland; (M.D.); (R.B.); (B.M.)
| | - Agnieszka Słowik
- Department of Neurology, Jagiellonian University, 30-688 Kraków, Poland;
| | - Renata Borawska
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland; (M.D.); (R.B.); (B.M.)
| | - Alina Kułakowska
- Department of Neurology, Medical University of Bialystok, 15-269 Bialystok, Poland; (A.K.); (J.K.)
| | - Jan Kochanowicz
- Department of Neurology, Medical University of Bialystok, 15-269 Bialystok, Poland; (A.K.); (J.K.)
| | - Barbara Mroczko
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland; (M.D.); (R.B.); (B.M.)
- Department of Biochemical Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland
| |
Collapse
|
16
|
Metzdorf K, Fricke S, Balia MT, Korte M, Zagrebelsky M. Nogo-A Modulates the Synaptic Excitation of Hippocampal Neurons in a Ca 2+-Dependent Manner. Cells 2021; 10:cells10092299. [PMID: 34571950 PMCID: PMC8467072 DOI: 10.3390/cells10092299] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 08/27/2021] [Accepted: 08/30/2021] [Indexed: 11/16/2022] Open
Abstract
A tight regulation of the balance between inhibitory and excitatory synaptic transmission is a prerequisite for synaptic plasticity in neuronal networks. In this context, the neurite growth inhibitor membrane protein Nogo-A modulates synaptic plasticity, strength, and neurotransmitter receptor dynamics. However, the molecular mechanisms underlying these actions are unknown. We show that Nogo-A loss-of-function in primary mouse hippocampal cultures by application of a function-blocking antibody leads to higher excitation following a decrease in GABAARs at inhibitory and an increase in the GluA1, but not GluA2 AMPAR subunit at excitatory synapses. This unbalanced regulation of AMPAR subunits results in the incorporation of Ca2+-permeable GluA2-lacking AMPARs and increased intracellular Ca2+ levels due to a higher Ca2+ influx without affecting its release from the internal stores. Increased neuronal activation upon Nogo-A loss-of-function prompts the phosphorylation of the transcription factor CREB and the expression of c-Fos. These results contribute to the understanding of the molecular mechanisms underlying the regulation of the excitation/inhibition balance and thereby of plasticity in the brain.
Collapse
Affiliation(s)
- Kristin Metzdorf
- Division of Cellular Neurobiology, Zoological Institute, TU Braunschweig, D-38106 Braunschweig, Germany; (K.M.); (M.T.B.); (M.K.)
- Helmholtz Centre for Infection Research, AG NIND, Inhoffenstr. 7, D-38124 Braunschweig, Germany
| | - Steffen Fricke
- Division of Cell Physiology, Zoological Institute, TU Braunschweig, D-38106 Braunschweig, Germany;
| | - Maria Teresa Balia
- Division of Cellular Neurobiology, Zoological Institute, TU Braunschweig, D-38106 Braunschweig, Germany; (K.M.); (M.T.B.); (M.K.)
| | - Martin Korte
- Division of Cellular Neurobiology, Zoological Institute, TU Braunschweig, D-38106 Braunschweig, Germany; (K.M.); (M.T.B.); (M.K.)
- Helmholtz Centre for Infection Research, AG NIND, Inhoffenstr. 7, D-38124 Braunschweig, Germany
| | - Marta Zagrebelsky
- Division of Cellular Neurobiology, Zoological Institute, TU Braunschweig, D-38106 Braunschweig, Germany; (K.M.); (M.T.B.); (M.K.)
- Correspondence: ; Tel.: +49-(0)-531-3913225
| |
Collapse
|
17
|
Stehle JH, Sheng Z, Hausmann L, Bechstein P, Weinmann O, Hernesniemi J, Neimat JS, Schwab ME, Zemmar A. Exercise-induced Nogo-A influences rodent motor learning in a time-dependent manner. PLoS One 2021; 16:e0250743. [PMID: 33951058 PMCID: PMC8099082 DOI: 10.1371/journal.pone.0250743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 04/13/2021] [Indexed: 11/22/2022] Open
Abstract
The adult, mature central nervous system (CNS) has limited plasticity. Physical exercising can counteract this limitation by inducing plasticity and fostering processes such as learning, memory consolidation and formation. Little is known about the molecular factors that govern these mechanisms, and how they are connected with exercise. In this study, we used immunohistochemical and behavioral analyses to investigate how running wheel exercise affects expression of the neuronal plasticity-inhibiting protein Nogo-A in the rat cortex, and how it influences motor learning in vivo. Following one week of exercise, rats exhibited a decrease in Nogo-A levels, selectively in motor cortex layer 2/3, but not in layer 5. Nogo-A protein levels returned to baseline after two weeks of running wheel exercise. In a skilled motor task (forelimb-reaching), administration of Nogo-A function-blocking antibodies over the course of the first training week led to improved motor learning. By contrast, Nogo-A antibody application over two weeks of training resulted in impaired learning. Our findings imply a bimodal, time-dependent function of Nogo-A in exercise-induced neuronal plasticity: While an activity-induced suppression of the plasticity-inhibiting protein Nogo-A appears initially beneficial for enhanced motor learning, presumably by allowing greater plasticity in establishing novel synaptic connections, this process is not sustained throughout continued exercise. Instead, upregulation of Nogo-A over the course of the second week of running wheel exercise in rats implies that Nogo-A is required for consolidation of acquired motor skills during the delayed memory consolidation process, possibly by inhibiting ongoing neuronal morphological reorganization to stabilize established synaptic pathways. Our findings suggest that Nogo-A downregulation allows leaning to occur, i.e. opens a 'learning window', while its later upregulation stabilizes the learnt engrams. These findings underline the importance of appropriately timing of application of Nogo-A antibodies in future clinical trials that aim to foster memory performance while avoiding adverse effects.
Collapse
Affiliation(s)
- Jörg H. Stehle
- Department of Neurosurgery, Henan Provincial People´s Hospital, Henan University People’s Hospital, Henan University School of Medicine, People’s Hospital of Zhengzhou University, Zhengzhou, China
- Dr. Senckenbergische Anatomie, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Zhiyuan Sheng
- Department of Neurosurgery, Henan Provincial People´s Hospital, Henan University People’s Hospital, Henan University School of Medicine, People’s Hospital of Zhengzhou University, Zhengzhou, China
| | - Laura Hausmann
- Department of Neurology, University Hospital RWTH Aachen, Aachen, Germany
| | - Philipp Bechstein
- Dr. Senckenbergische Anatomie, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Oliver Weinmann
- Brain Research Institute, University of Zurich, Zurich, Switzerland
- Department of Biology and Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Juha Hernesniemi
- Department of Neurosurgery, Henan Provincial People´s Hospital, Henan University People’s Hospital, Henan University School of Medicine, People’s Hospital of Zhengzhou University, Zhengzhou, China
| | - Joseph S. Neimat
- Department of Neurosurgery, University of Louisville, School of Medicine, Louisville, Kentucky, United States of America
| | - Martin E. Schwab
- Brain Research Institute, University of Zurich, Zurich, Switzerland
- Department of Biology and Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Ajmal Zemmar
- Department of Neurosurgery, Henan Provincial People´s Hospital, Henan University People’s Hospital, Henan University School of Medicine, People’s Hospital of Zhengzhou University, Zhengzhou, China
- Brain Research Institute, University of Zurich, Zurich, Switzerland
- Department of Biology and Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
18
|
The Implication of Reticulons (RTNs) in Neurodegenerative Diseases: From Molecular Mechanisms to Potential Diagnostic and Therapeutic Approaches. Int J Mol Sci 2021; 22:ijms22094630. [PMID: 33924890 PMCID: PMC8125174 DOI: 10.3390/ijms22094630] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/21/2021] [Accepted: 04/23/2021] [Indexed: 02/07/2023] Open
Abstract
Reticulons (RTNs) are crucial regulatory factors in the central nervous system (CNS) as well as immune system and play pleiotropic functions. In CNS, RTNs are transmembrane proteins mediating neuroanatomical plasticity and functional recovery after central nervous system injury or diseases. Moreover, RTNs, particularly RTN4 and RTN3, are involved in neurodegeneration and neuroinflammation processes. The crucial role of RTNs in the development of several neurodegenerative diseases, including Alzheimer's disease (AD), multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), or other neurological conditions such as brain injury or spinal cord injury, has attracted scientific interest. Reticulons, particularly RTN-4A (Nogo-A), could provide both an understanding of early pathogenesis of neurodegenerative disorders and be potential therapeutic targets which may offer effective treatment or inhibit disease progression. This review focuses on the molecular mechanisms and functions of RTNs and their potential usefulness in clinical practice as a diagnostic tool or therapeutic strategy.
Collapse
|
19
|
Fletcher JL, Makowiecki K, Cullen CL, Young KM. Oligodendrogenesis and myelination regulate cortical development, plasticity and circuit function. Semin Cell Dev Biol 2021; 118:14-23. [PMID: 33863642 DOI: 10.1016/j.semcdb.2021.03.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 03/23/2021] [Accepted: 03/25/2021] [Indexed: 12/17/2022]
Abstract
During cortical development and throughout adulthood, oligodendrocytes add myelin internodes to glutamatergic projection neurons and GABAergic inhibitory neurons. In addition to directing node of Ranvier formation, to enable saltatory conduction and influence action potential transit time, oligodendrocytes support axon health by communicating with axons via the periaxonal space and providing metabolic support that is particularly critical for healthy ageing. In this review we outline the timing of oligodendrogenesis in the developing mouse and human cortex and describe the important role that oligodendrocytes play in sustaining and modulating neuronal function. We also provide insight into the known and speculative impact that myelination has on cortical axons and their associated circuits during the developmental critical periods and throughout life, particularly highlighting their life-long role in learning and remembering.
Collapse
Affiliation(s)
- Jessica L Fletcher
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Kalina Makowiecki
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Carlie L Cullen
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Kaylene M Young
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia.
| |
Collapse
|
20
|
Liu H, He J, Wu Y, Du Y, Jiang Y, Chen C, Yu Z, Zhong J, Wang Z, Cheng C, Sun X, Huang Z. Endothelial Regulation by Exogenous Annexin A1 in Inflammatory Response and BBB Integrity Following Traumatic Brain Injury. Front Neurosci 2021; 15:627110. [PMID: 33679307 PMCID: PMC7930239 DOI: 10.3389/fnins.2021.627110] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 01/15/2021] [Indexed: 12/19/2022] Open
Abstract
Background and Target Following brain trauma, blood–brain barrier (BBB) disruption and inflammatory response are critical pathological steps contributing to secondary injury, leading to high mortality and morbidity. Both pathologies are closely associated with endothelial remodeling. In the present study, we concentrated on annexin A1 (ANXA1) as a novel regulator of endothelial function after traumatic brain injury. Methods After establishing controlled cortical impact (CCI) model in male mice, human recombinant ANXA1 (rANXA1) was administered intravenously, followed by assessments of BBB integrity, brain edema, inflammatory response, and neurological deficits. Result Animals treated with rANXA1 (1 μg/kg) at 1 h after CCI exhibited optimal BBB protection including alleviated BBB disruption and brain edema, as well as endothelial junction proteins loss. The infiltrated neutrophils and inflammatory cytokines were suppressed by rANXA1, consistent with decreased adhesive and transmigrating molecules from isolated microvessels. Moreover, rANXA1 attenuated the neurological deficits induced by CCI. We further found that the Ras homolog gene family member A (RhoA) inhibition has similar effect as rANXA1 in ameliorating brain injuries after CCI, whereas rANXA1 suppressed CCI-induced RhoA activation. Conclusion Our findings suggest that the endothelial remodeling by exogenous rANXA1 corrects BBB disruption and inflammatory response through RhoA inhibition, hence improving functional outcomes in CCI mice.
Collapse
Affiliation(s)
- Han Liu
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Department of Neurosurgery, Qilu Hospital of Shandong University (Qingdao Campus), Qingdao, China
| | - Junchi He
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yue Wu
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yang Du
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Yinghua Jiang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Chengzhi Chen
- Department of Occupational and Environmental Health, School of Public Health and Management, Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing Medical University, Chongqing, China
| | - Zhanyang Yu
- Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Jianjun Zhong
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhigang Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University (Qingdao Campus), Qingdao, China
| | - Chongjie Cheng
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaochuan Sun
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhijian Huang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
21
|
Espírito-Santo S, Coutinho VG, Dezonne RS, Stipursky J, Dos Santos-Rodrigues A, Batista C, Paes-de-Carvalho R, Fuss B, Gomes FCA. Astrocytes as a target for Nogo-A and implications for synapse formation in vitro and in a model of acute demyelination. Glia 2021; 69:1429-1443. [PMID: 33497496 DOI: 10.1002/glia.23971] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 12/21/2020] [Accepted: 01/14/2021] [Indexed: 12/20/2022]
Abstract
Central nervous system (CNS) function depends on precise synaptogenesis, which is shaped by environmental cues and cellular interactions. Astrocytes are outstanding regulators of synapse development and plasticity through contact-dependent signals and through the release of pro- and antisynaptogenic factors. Conversely, myelin and its associated proteins, including Nogo-A, affect synapses in a inhibitory fashion and contribute to neural circuitry stabilization. However, the roles of Nogo-A-astrocyte interactions and their implications in synapse development and plasticity have not been characterized. Therefore, we aimed to investigate whether Nogo-A affects the capacity of astrocytes to induce synaptogenesis. Additionally, we assessed whether downregulation of Nogo-A signaling in an in vivo demyelination model impacts the synaptogenic potential of astrocytes. Our in vitro data show that cortical astrocytes respond to Nogo-A through RhoA pathway activation, exhibiting stress fiber formation and decreased ramified morphology. This phenotype was associated with reduced levels of GLAST protein and aspartate uptake, decreased mRNA levels of the synaptogenesis-associated genes Hevin, glypican-4, TGF-β1 and BDNF, and decreased and increased protein levels of Hevin and SPARC, respectively. Corroborating these findings, conditioned medium from Nogo-A-treated astrocytes suppressed the formation of structurally and functionally mature synapses in cortical neuronal cultures. After cuprizone-induced acute demyelination, we observed reduced immunostaining for Nogo-A in the visual cortex accompanied by higher levels of Hevin expression in astrocytes and an increase in excitatory synapse density. Hence, we suggest that interactions between Nogo-A and astrocytes might represent an important pathway of plasticity regulation and could be a target for therapeutic intervention in demyelinating diseases in the future.
Collapse
Affiliation(s)
- Sheila Espírito-Santo
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Departamento de Ciências Biológicas, Universidade do Estado de Minas Gerais, Minas Gerais, Brazil
| | - Vinícius G Coutinho
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rômulo S Dezonne
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Joice Stipursky
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Carolina Batista
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Roberto Paes-de-Carvalho
- Instituto de Biologia, Programa de Neurociências, Universidade Federal Fluminense, Niterói, Brazil
| | - Babette Fuss
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | | |
Collapse
|
22
|
Abstract
Nogo-A is considered one of the most important inhibitors of myelin-associated axonal regeneration in the central nervous system. It is mainly expressed by oligodendrocytes. Although previous studies have found regulatory roles for Nogo-A in neurite outgrowth inhibition, neuronal homeostasis, precursor migration, plasticity, and neurodegeneration, its functions in the process of oxidative injury are largely uncharacterized. In this study, oligodendrocytes were extracted from the cerebral cortex of newborn Sprague-Dawley rats. We used hydrogen peroxide (H2O2) to induce an in vitro oligodendrocyte oxidative damage model and found that endogenously expressed Nogo-A is significantly upregulated in oligodendrocytes. After recombinant virus Ad-ZsGreen-rat Nogo-A infection of oligodendrocytes, Nogo-A expression was increased, and the infected oligodendrocytes were more susceptible to acute oxidative insults and exhibited a markedly elevated rate of cell death. Furthermore, knockdown of Nogo-A expression in oligodendrocytes by Ad-ZsGreen-shRNA-Nogo-A almost completely protected against oxidative stress induced by exogenous H2O2. Intervention with a Nogo-66 antibody, a LINGO1 blocker, or Y27632, an inhibitor in the Nogo-66-NgR/p75/LINGO-1-RhoA-ROCK pathway, did not affect the death of oligodendrocytes. Ad-ZsGreen-shRNA-Nogo-A also increased the levels of phosphorylated extracellular signal-regulated kinase 1/2 and inhibited BCL2 expression in oligodendrocytes. In conclusion, Nogo-A aggravated reactive oxygen species damage in oligodendrocytes, and phosphorylated extracellular signal-regulated kinase 1/2 and BCL2 might be involved in this process. This study was approved by the Ethics Committee of Peking University People’s Hospital, China (approval No. 2018PHC081) on December 18, 2018.
Collapse
Affiliation(s)
- Yang-Yang Wang
- Department of Neurology, Peking University People's Hospital, Beijing, China
| | - Na Han
- Department of Neurology, Peking University People's Hospital, Beijing, China
| | - Dao-Jun Hong
- Department of Neurology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Jun Zhang
- Department of Neurology, Peking University People's Hospital, Beijing, China
| |
Collapse
|
23
|
Christiansen L, Larsen MN, Madsen MJ, Grey MJ, Nielsen JB, Lundbye-Jensen J. Long-term motor skill training with individually adjusted progressive difficulty enhances learning and promotes corticospinal plasticity. Sci Rep 2020; 10:15588. [PMID: 32973251 PMCID: PMC7518278 DOI: 10.1038/s41598-020-72139-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 08/21/2020] [Indexed: 12/16/2022] Open
Abstract
Motor skill acquisition depends on central nervous plasticity. However, behavioural determinants leading to long lasting corticospinal plasticity and motor expertise remain unexplored. Here we investigate behavioural and electrophysiological effects of individually tailored progressive practice during long-term motor skill training. Two groups of participants practiced a visuomotor task requiring precise control of the right digiti minimi for 6 weeks. One group trained with constant task difficulty, while the other group trained with progressively increasing task difficulty, i.e. continuously adjusted to their individual skill level. Compared to constant practice, progressive practice resulted in a two-fold greater performance at an advanced task level and associated increases in corticospinal excitability. Differences were maintained 8 days later, whereas both groups demonstrated equal retention 14 months later. We demonstrate that progressive practice enhances motor skill learning and promotes corticospinal plasticity. These findings underline the importance of continuously challenging patients and athletes to promote neural plasticity, skilled performance, and recovery.
Collapse
Affiliation(s)
- Lasse Christiansen
- Department of Nutrition Exercise and Sports, University of Copenhagen, Copenhagen, Denmark. .,Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital, Amager and Hvidovre, Hvidovre, Denmark.
| | - Malte Nejst Larsen
- Department of Nutrition Exercise and Sports, University of Copenhagen, Copenhagen, Denmark.,Department of Sports Science and Clinical Biomechanics, University of Southern Denmark, Odense, Denmark
| | - Mads Just Madsen
- Department of Nutrition Exercise and Sports, University of Copenhagen, Copenhagen, Denmark.,Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital, Amager and Hvidovre, Hvidovre, Denmark
| | - Michael James Grey
- School of Health Sciences, Faculty of Medicine and Health Sciences, University of East Anglia, Norwich Research Park, Norwich, United Kingdom
| | - Jens Bo Nielsen
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
| | - Jesper Lundbye-Jensen
- Department of Nutrition Exercise and Sports, University of Copenhagen, Copenhagen, Denmark.,Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
24
|
Fricke S, Metzdorf K, Ohm M, Haak S, Heine M, Korte M, Zagrebelsky M. Fast Regulation of GABA AR Diffusion Dynamics by Nogo-A Signaling. Cell Rep 2020; 29:671-684.e6. [PMID: 31618635 DOI: 10.1016/j.celrep.2019.09.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 07/02/2019] [Accepted: 09/06/2019] [Indexed: 12/29/2022] Open
Abstract
Precisely controlling the excitatory and inhibitory balance is crucial for the stability and information-processing ability of neuronal networks. However, the molecular mechanisms maintaining this balance during ongoing sensory experiences are largely unclear. We show that Nogo-A signaling reciprocally regulates excitatory and inhibitory transmission. Loss of function for Nogo-A signaling through S1PR2 rapidly increases GABAAR diffusion, thereby decreasing their number at synaptic sites and the amplitude of GABAergic mIPSCs at CA3 hippocampal neurons. This increase in GABAAR diffusion rate is correlated with an increase in Ca2+ influx and requires the calcineurin-mediated dephosphorylation of the γ2 subunit at serine 327. These results suggest that Nogo-A signaling rapidly strengthens inhibitory GABAergic transmission by restricting the diffusion dynamics of GABAARs. Together with the observation that Nogo-A signaling regulates excitatory transmission in an opposite manner, these results suggest a crucial role for Nogo-A signaling in modulating the excitation and inhibition balance to restrict synaptic plasticity.
Collapse
Affiliation(s)
- Steffen Fricke
- Zoological Institute, Division of Cellular Neurobiology, TU Braunschweig, Braunschweig 38108, Germany
| | - Kristin Metzdorf
- Zoological Institute, Division of Cellular Neurobiology, TU Braunschweig, Braunschweig 38108, Germany
| | - Melanie Ohm
- Zoological Institute, Division of Cellular Neurobiology, TU Braunschweig, Braunschweig 38108, Germany
| | - Stefan Haak
- Zoological Institute, Division of Cellular Neurobiology, TU Braunschweig, Braunschweig 38108, Germany
| | - Martin Heine
- Molecular Physiology Group, Leibniz Institute of Neurobiology, Magdeburg 39118, Germany; Functional Neurobiology, Institute for Developmental Biology and Neurobiology, Johannes Gutenberg University, Mainz 55128, Germany
| | - Martin Korte
- Zoological Institute, Division of Cellular Neurobiology, TU Braunschweig, Braunschweig 38108, Germany; Helmholtz Centre for Infection Research, AG NIND, Inhoffenstr. 7, Braunschweig 38124, Germany
| | - Marta Zagrebelsky
- Zoological Institute, Division of Cellular Neurobiology, TU Braunschweig, Braunschweig 38108, Germany.
| |
Collapse
|
25
|
Yu C, Sun X, Li J, Chan SO, Wang L. Analysis of axon divergence at the optic chiasm in nogo-a knockout mice. Neurosci Lett 2020; 731:135109. [PMID: 32492476 DOI: 10.1016/j.neulet.2020.135109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 05/26/2020] [Accepted: 05/29/2020] [Indexed: 10/24/2022]
Abstract
Our earlier studies have shown that the axon growth inhibitory molecule Nogo affects axon routing at the optic chiasm likely through a differential regulation of Nogo receptor on the optic axons. Using isoform specific antibodies, we further showed that Nogo-A was predominantly expressed by retinal ganglion cells and their axons, while Nogo-B was highly localized on the radial glia at the midline of the chiasm, suggesting a role of Nogo-B in regulating turning of uncrossed axons. To further investigate the roles of Nogo-A in axon divergence, we analyzed the routing of axons in the chiasm of Nogo-A knockout mice during the growth of axons across the midline. At E13 to E16, there was no significant difference in the contralateral projection (P = 0.6943 for E13; P = 0.9867 for E14; P = 0.4121 for E15 and P = 0.3402 for E16). The results also showed the absence of Nogo-A did not cause any obvious change to the ipsilateral projection at the optic chiasm, both for the early generated uncrossed axons at E13 and E14 and the late cohorts at E15-E16, when compared with the wild-type mice (P = 0.4788 for E13; P = 0.188 for E14; P = 0.3152 for E15 and P = 0.432 for E16). These findings support that Nogo-A is not the major isoform to guide the axon divergence in the mouse optic chiasm.
Collapse
Affiliation(s)
- Chao Yu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China; Medical Examination Center, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China.
| | - Xiaobo Sun
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China.
| | - Jing Li
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China.
| | - Sun-On Chan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China.
| | - Liqing Wang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China; School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China.
| |
Collapse
|
26
|
Affiliation(s)
- Bor Luen Tang
- Department of Biochemistry, Yong Loo Lin School of Medicine; National University of Singapore Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore
| |
Collapse
|
27
|
Zhang J, Li Z, Liu W, Zeng W, Duan C, He X. Effects of bone marrow mesenchymal stem cells transplantation on the recovery of neurological functions and the expression of Nogo-A, NgR, Rhoa, and ROCK in rats with experimentally-induced convalescent cerebral ischemia. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:390. [PMID: 32355834 PMCID: PMC7186734 DOI: 10.21037/atm.2020.03.144] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Background To investigate the effects of intravenous transplantation of bone marrow mesenchymal stem cells (BMSCs) on neurological function in rats with experimentally-induced convalescent cerebral ischemia and the expression of Nogo-A, NgR, Rhoa, and ROCK expression. Methods BMSCs were isolated and cultured in vitro using the whole bone marrow adherent method. Eighty-one adult male Sprague-Dawley rats were divided at random into three groups: the sham-operated group, the cerebral ischemia group, and the BMSC treatment group (n=27 rats per group). In the latter two groups, the middle cerebral artery occlusion (MCAO) model was performed by the modified Zea Longa method. After MCAO, rats in the sham-operated and cerebral ischemic groups were injected with 1 mL of phosphate buffered saline (PBS) via the tail vein. In the BMSC-treatment group, 1 mL of the BMSC suspension (containing 3×106 BMSCs) was injected through the rats’ femoral vein. At 12, 24, and 72 h after BMSC transplantation, modified neurological deficit scores (mNSS) were used to assess neurological function. TTC (2,3,5-triphenyl tetrazolium chloride) staining was used to measure the ischemic lesion volume, and the distribution of Nogo-A protein was observed by immunohistochemistry. The expressions of Nogo-A, NgR, Rhoa, and ROCK were detected by Western blot. Results At 72 h after BMSC transplantation, the mNSS scores were significantly lower in the BMSC treatment group than those in the cerebral ischemia group (7.50±0.55 vs. 8.67±0.52, P<0.01), and the ischemic lesions volume was significantly reduced. The expressions of Nogo-A, NgR, RhoA, and ROCK were significantly decreased compared with the controls (P<0.05). Conclusions The transplantation of BMSCs can improve neurological function in rats after convalescent cerebral ischemia, and their therapeutic effect may be related to the downregulation of Nogo-A, NgR, RhoA, and ROCK expression.
Collapse
Affiliation(s)
- Jianbo Zhang
- Department of Neurosurgery, The National Key Clinic Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Province Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Zhenjun Li
- Department of Neurosurgery, The National Key Clinic Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Province Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Wenchao Liu
- Department of Neurosurgery, The National Key Clinic Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Province Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Wenxian Zeng
- Department of Neurosurgery, The National Key Clinic Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Province Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Chuanzhi Duan
- Department of Neurosurgery, The National Key Clinic Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Province Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Xuying He
- Department of Neurosurgery, The National Key Clinic Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Province Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China.,Department of Neurosurgery, Southern Medical University, Zhujiang Hospital, Guangzhou 510282, China
| |
Collapse
|
28
|
Ueno R, Takase H, Suenaga J, Kishimoto M, Kurihara Y, Takei K, Kawahara N, Yamamoto T. Axonal regeneration and functional recovery driven by endogenous Nogo receptor antagonist LOTUS in a rat model of unilateral pyramidotomy. Exp Neurol 2019; 323:113068. [PMID: 31629859 DOI: 10.1016/j.expneurol.2019.113068] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 09/16/2019] [Accepted: 09/18/2019] [Indexed: 12/26/2022]
Abstract
The adult mammalian central nervous system (CNS) rarely recovers from injury. Myelin fragments contain axonal growth inhibitors that limit axonal regeneration, thus playing a major role in determining neural recovery. Nogo receptor-1 (NgR1) and its ligands are among the inhibitors that limit axonal regeneration. It has been previously shown that the endogenous protein, lateral olfactory tract usher substance (LOTUS), antagonizes NgR1-mediated signaling and accelerates neuronal plasticity after spinal cord injury and cerebral ischemia in mice. However, it remained unclear whether LOTUS-mediated reorganization of descending motor pathways in the adult brain is physiologically functional and contributes to functional recovery. Here, we generated LOTUS-overexpressing transgenic (LOTUS-Tg) rats to investigate the role of LOTUS in neuronal function after damage. After unilateral pyramidotomy, motor function in LOTUS-Tg rats recovered significantly compared to that in wild-type animals. In a retrograde tracing study, labeled axons spanning from the impaired side of the cervical spinal cord to the unlesioned hemisphere of the red nucleus and sensorimotor cortex were increased in LOTUS-Tg rats. Anterograde tracing from the unlesioned cortex also revealed enhanced ipsilateral connectivity to the impaired side of the cervical spinal cord in LOTUS-Tg rats. Moreover, electrophysiological analysis showed that contralesional cortex stimulation significantly increased ipsilateral forelimb movement in LOTUS-Tg rats, which was consistent with the histological findings. According to these data, LOTUS overexpression accelerates ipsilateral projection from the unlesioned cortex and promotes functional recovery after unilateral pyramidotomy. LOTUS could be a future therapeutic option for CNS injury.
Collapse
Affiliation(s)
- Ryu Ueno
- Department of Neurosurgery, Yokohama City University, Yokohama, Japan.
| | - Hajime Takase
- Department of Neurosurgery, Yokohama City University, Yokohama, Japan.
| | - Jun Suenaga
- Department of Neurosurgery, Yokohama City University, Yokohama, Japan.
| | - Masao Kishimoto
- Department of Neurosurgery, Yokohama City University, Yokohama, Japan.
| | - Yuji Kurihara
- Molecular Medical Bioscience Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, Yokohama, Japan.
| | - Kohtaro Takei
- Molecular Medical Bioscience Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, Yokohama, Japan.
| | - Nobutaka Kawahara
- Department of Neurosurgery, Yokohama City University, Yokohama, Japan
| | - Tetsuya Yamamoto
- Department of Neurosurgery, Yokohama City University, Yokohama, Japan.
| |
Collapse
|
29
|
Zemmar A, Chen CC, Weinmann O, Kast B, Vajda F, Bozeman J, Isaad N, Zuo Y, Schwab ME. Oligodendrocyte- and Neuron-Specific Nogo-A Restrict Dendritic Branching and Spine Density in the Adult Mouse Motor Cortex. Cereb Cortex 2019; 28:2109-2117. [PMID: 28505229 PMCID: PMC6018724 DOI: 10.1093/cercor/bhx116] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Indexed: 01/27/2023] Open
Abstract
Nogo-A has been well described as a myelin-associated inhibitor of neurite outgrowth and functional neuroregeneration after central nervous system (CNS) injury. Recently, a new role of Nogo-A has been identified as a negative regulator of synaptic plasticity in the uninjured adult CNS. Nogo-A is present in neurons and oligodendrocytes. However, it is yet unclear which of these two pools regulate synaptic plasticity. To address this question we used newly generated mouse lines in which Nogo-A is specifically knocked out in (1) oligodendrocytes (oligoNogo-A KO) or (2) neurons (neuroNogo-A KO). We show that both oligodendrocyte- and neuron-specific Nogo-A KO mice have enhanced dendritic branching and spine densities in layer 2/3 cortical pyramidal neurons. These effects are compartmentalized: neuronal Nogo-A affects proximal dendrites whereas oligodendrocytic Nogo-A affects distal regions. Finally, we used two-photon laser scanning microscopy to measure the spine turnover rate of adult mouse motor cortex layer 5 cells and find that both Nogo-A KO mouse lines show enhanced spine remodeling after 4 days. Our results suggest relevant control functions of glial as well as neuronal Nogo-A for synaptic plasticity and open new possibilities for more selective and targeted plasticity enhancing strategies.
Collapse
Affiliation(s)
- Ajmal Zemmar
- Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland.,Department of Biology and Department of Health Sciences and Technology, ETH Zurich, 8057 Zurich, Switzerland.,Department of Neurosurgery, University Hospital Zurich, University of Zurich, CH-8091, Zurich, Switzerland
| | - Chia-Chien Chen
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA 95064, USA
| | - Oliver Weinmann
- Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland.,Department of Biology and Department of Health Sciences and Technology, ETH Zurich, 8057 Zurich, Switzerland
| | - Brigitt Kast
- Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland.,Department of Biology and Department of Health Sciences and Technology, ETH Zurich, 8057 Zurich, Switzerland
| | - Flora Vajda
- Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland.,Department of Biology and Department of Health Sciences and Technology, ETH Zurich, 8057 Zurich, Switzerland
| | - James Bozeman
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA 95064, USA
| | - Noel Isaad
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA 95064, USA
| | - Yi Zuo
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA 95064, USA
| | - Martin E Schwab
- Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland.,Department of Biology and Department of Health Sciences and Technology, ETH Zurich, 8057 Zurich, Switzerland
| |
Collapse
|
30
|
Germany CE, Reker AN, Hinton DJ, Oliveros A, Shen X, Andres-Beck LG, Wininger KM, Trutschl M, Cvek U, Choi DS, Nam HW. Pharmacoproteomics Profile in Response to Acamprosate Treatment of an Alcoholism Animal Model. Proteomics 2019; 18:e1700417. [PMID: 29437267 DOI: 10.1002/pmic.201700417] [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: 11/10/2017] [Revised: 01/19/2018] [Indexed: 12/20/2022]
Abstract
Acamprosate is an FDA-approved medication for the treatment of alcoholism that is unfortunately only effective in certain patients. Although acamprosate is known to stabilize the hyper-glutamatergic state in alcoholism, pharmacological mechanisms of action in brain tissue remains unknown. To investigate the mechanism of acamprosate efficacy, the authors employ a pharmacoproteomics approach using an animal model of alcoholism, type 1 equilibrative nucleoside transporter (ENT1) null mice. The results demonstrate that acamprosate treatment significantly decreased both ethanol drinking and preference in ENT1 null mice compared to that of wild-type mice. Then, to elucidate acamprosate efficacy mechanism in ENT1 null mice, the authors utilize label-free quantification proteomics comparing both genotype and acamprosate treatment effects in the nucleus accumbens (NAc). A total of 1040 protein expression changes are identified in the NAc among 3634 total proteins detected. The proteomics and Western blot result demonstrate that acamprosate treatment decreased EAAT expression implicating stabilization of the hyper-glutamatergic condition in ENT1 null mice. Pathway analysis suggests that acamprosate treatment in ENT1 null mice seems to rescue glutamate toxicity through restoring of RTN4 and NF-κB medicated neuroimmune signaling compared to wild-type mice. Overall, pharmacoproteomics approaches suggest that neuroimmune restoration is a potential efficacy mechanism in the acamprosate treatment of certain sub-populations of alcohol dependent subjects.
Collapse
Affiliation(s)
- Caroline E Germany
- Department of Pharmacology, Toxicology, and Neuroscience, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Ashlie N Reker
- Department of Pharmacology, Toxicology, and Neuroscience, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - David J Hinton
- Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Alfredo Oliveros
- Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Xinggui Shen
- Department of Pharmacology, Toxicology, and Neuroscience, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Lindsey G Andres-Beck
- Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Katheryn M Wininger
- Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Marjan Trutschl
- Department of Computer Science, Louisiana State University-Shreveport, Shreveport, LA, USA
| | - Urska Cvek
- Department of Computer Science, Louisiana State University-Shreveport, Shreveport, LA, USA
| | - Doo-Sup Choi
- Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Hyung W Nam
- Department of Pharmacology, Toxicology, and Neuroscience, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| |
Collapse
|
31
|
Berry S, Weinmann O, Fritz AK, Rust R, Wolfer D, Schwab ME, Gerber U, Ster J. Loss of Nogo-A, encoded by the schizophrenia risk gene Rtn4, reduces mGlu3 expression and causes hyperexcitability in hippocampal CA3 circuits. PLoS One 2018; 13:e0200896. [PMID: 30040841 PMCID: PMC6057643 DOI: 10.1371/journal.pone.0200896] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 07/04/2018] [Indexed: 11/19/2022] Open
Abstract
Recent investigations of Nogo-A, a well characterized protein inhibitor of neurite outgrowth in the brain, have revealed additional functions including a role in neuropsychiatric disorders such as schizophrenia. Here we examined Nogo-A functions in mouse CA3 hippocampal circuitry. Patch clamp recordings showed that the absence of Nogo-A results in a hyperactive network. In addition, mGlu3 metabotropic glutamate receptors, which exhibit mutations in certain forms of schizophrenia, were downregulated specifically in the CA3 area. Furthermore, Nogo-A-/- mice showed disordered theta oscillations with decreased incidence and frequency, similar to those observed in mGlu3-/- mice. As disruptions in theta rhythmicity are associated with impaired spatial navigation, we tested mice using modified Morris water maze tasks. Mice lacking Nogo-A exhibited altered search strategies, displaying greater dependence on global as opposed to local reference frames. This link between Nogo-A and mGlu3 receptors may provide new insights into mechanisms underlying schizophrenia.
Collapse
Affiliation(s)
- Stewart Berry
- Brain Research Institute, University of Zurich, Zurich, Switzerland
| | - Oliver Weinmann
- Brain Research Institute, University of Zurich, Zurich, Switzerland
| | | | - Ruslan Rust
- Brain Research Institute, University of Zurich, Zurich, Switzerland
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - David Wolfer
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Martin E. Schwab
- Brain Research Institute, University of Zurich, Zurich, Switzerland
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Urs Gerber
- Brain Research Institute, University of Zurich, Zurich, Switzerland
| | - Jeanne Ster
- Brain Research Institute, University of Zurich, Zurich, Switzerland
- * E-mail:
| |
Collapse
|
32
|
Nogo-A inactivation improves visual plasticity and recovery after retinal injury. Cell Death Dis 2018; 9:727. [PMID: 29950598 PMCID: PMC6021388 DOI: 10.1038/s41419-018-0780-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 06/02/2018] [Accepted: 06/06/2018] [Indexed: 12/24/2022]
Abstract
Myelin-associated proteins such as Nogo-A are major inhibitors of neuronal plasticity that contribute to permanent neurological impairments in the injured CNS. In the present study, we investigated the influence of Nogo-A on visual recovery after retinal injuries in mice. Different doses of N-methyl-d-aspartate (NMDA) were injected in the vitreous of the left eye to induce retinal neuron death. The visual function was monitored using the optokinetic response (OKR) as a behavior test, and electroretinogram (ERG) and local field potential (LFP) recordings allowed to assess changes in retinal and cortical neuron activity, respectively. Longitudinal OKR follow-ups revealed reversible visual deficits after injection of NMDA ≤ 1 nmole in the left eye and concomitant functional improvement in the contralateral visual pathway of the right eye that was let intact. Irreversible OKR loss observed with NMDA ≥ 2 nmol was correlated with massive retinal cell death and important ERG response decline. Strikingly, the OKR mediated by injured and intact eye stimulation was markedly improved in Nogo-A KO mice compared with WT animals, suggesting that the inactivation of Nogo-A promotes visual recovery and plasticity. Moreover, OKR improvement was associated with shorter latency of the N2 wave of Nogo-A KO LFPs relative to WT animals. Strikingly, intravitreal injection of anti-Nogo-A antibody (11C7) in the injured eye exerted positive effects on cortical LFPs. This study presents the intrinsic ability of the visual system to recover from NMDA-induced retinal injury and its limitations. Nogo-A neutralization may promote visual recovery in retinal diseases such as glaucoma.
Collapse
|
33
|
The Extracellular Environment of the CNS: Influence on Plasticity, Sprouting, and Axonal Regeneration after Spinal Cord Injury. Neural Plast 2018; 2018:2952386. [PMID: 29849554 PMCID: PMC5932463 DOI: 10.1155/2018/2952386] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 01/22/2018] [Accepted: 02/06/2018] [Indexed: 11/17/2022] Open
Abstract
The extracellular environment of the central nervous system (CNS) becomes highly structured and organized as the nervous system matures. The extracellular space of the CNS along with its subdomains plays a crucial role in the function and stability of the CNS. In this review, we have focused on two components of the neuronal extracellular environment, which are important in regulating CNS plasticity including the extracellular matrix (ECM) and myelin. The ECM consists of chondroitin sulfate proteoglycans (CSPGs) and tenascins, which are organized into unique structures called perineuronal nets (PNNs). PNNs associate with the neuronal cell body and proximal dendrites of predominantly parvalbumin-positive interneurons, forming a robust lattice-like structure. These developmentally regulated structures are maintained in the adult CNS and enhance synaptic stability. After injury, however, CSPGs and tenascins contribute to the structure of the inhibitory glial scar, which actively prevents axonal regeneration. Myelin sheaths and mature adult oligodendrocytes, despite their important role in signal conduction in mature CNS axons, contribute to the inhibitory environment existing after injury. As such, unlike the peripheral nervous system, the CNS is unable to revert to a “developmental state” to aid neuronal repair. Modulation of these external factors, however, has been shown to promote growth, regeneration, and functional plasticity after injury. This review will highlight some of the factors that contribute to or prevent plasticity, sprouting, and axonal regeneration after spinal cord injury.
Collapse
|
34
|
Iobbi C, Korte M, Zagrebelsky M. Nogo-66 Restricts Synaptic Strengthening via Lingo1 and the ROCK2-Cofilin Pathway to Control Actin Dynamics. Cereb Cortex 2018; 27:2779-2792. [PMID: 27166169 DOI: 10.1093/cercor/bhw122] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Nogo-A restricts long-term potentiation (LTP) at the Schaffer collateral-CA1 pathway in the adult hippocampus via 2 extracellular domains: Nogo-A-Δ20 and Nogo-66. Nogo-66 signals via Nogo Receptor 1 (NgR1) to regulate synaptic function. Whether the NgR1 coreceptors Lingo1 and p75NTR are involved in the signaling in this context is still not known. Moreover, the intracellular cascade mediating the activity of Nogo-66 in restricting LTP is unexplored. We combine electrophysiology and biochemistry in acute hippocampal slices and demonstrate that a loss of function for Lingo1 results in a significant increase in LTP levels at the Schaffer collateral-CA1 pathway, and that Lingo1 is the NgR1 coreceptor mediating the role of Nogo-66 in restricting LTP. Our data show that p75NTR is not involved in mediating the Nogo-66 effect on LTP. Moreover, loss of function for p75NTR and NgR1 equally attenuate LTD, suggesting that p75NTR might mediate the NgR1-dependent regulation of LTD, independently of Nogo-66. Finally, our results indicate that Nogo-66 signaling limits LTP via the ROCK2-Cofilin pathway to control the dynamics of the actin cytoskeleton. The present results elucidate the signaling pathway activated by Nogo-66 to control LTP and contribute to the understanding of how Nogo-A stabilizes the neural circuits to limit activity-dependent plasticity events in the mature hippocampus.
Collapse
Affiliation(s)
- Cristina Iobbi
- Zoological Institute, Division of Cellular Neurobiology, TU Braunschweig, 38106, Braunschweig, Germany
| | - Martin Korte
- Zoological Institute, Division of Cellular Neurobiology, TU Braunschweig, 38106, Braunschweig, Germany.,Helmholtz Centre for Infection Research, AG NIND, 38124, Braunschweig, Germany
| | - Marta Zagrebelsky
- Zoological Institute, Division of Cellular Neurobiology, TU Braunschweig, 38106, Braunschweig, Germany
| |
Collapse
|
35
|
Smedfors G, Olson L, Karlsson TE. A Nogo-Like Signaling Perspective from Birth to Adulthood and in Old Age: Brain Expression Patterns of Ligands, Receptors and Modulators. Front Mol Neurosci 2018. [PMID: 29520216 PMCID: PMC5827527 DOI: 10.3389/fnmol.2018.00042] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
An appropriate strength of Nogo-like signaling is important to maintain synaptic homeostasis in the CNS. Disturbances have been associated with schizophrenia, MS and other diseases. Blocking Nogo-like signaling may improve recovery after spinal cord injury, stroke and traumatic brain injury. To understand the interacting roles of an increasing number of ligands, receptors and modulators engaged in Nogo-like signaling, the transcriptional activity of these genes in the same brain areas from birth to old age in the normal brain is needed. Thus, we have quantitatively mapped the innate expression of 11 important genes engaged in Nogo-like signaling. Using in situ hybridization, we located and measured the amount of mRNA encoding Nogo-A, OMgp, NgR1, NgR2, NgR3, Lingo-1, Troy, Olfactomedin, LgI1, ADAM22, and MAG, in 18 different brain areas at six different ages (P0, 1, 2, 4, 14, and 104 weeks). We show gene- and area-specific activities and how the genes undergo dynamic regulation during postnatal development and become stable during adulthood. Hippocampal areas underwent the largest changes over time. We only found differences between individual cortical areas in Troy and MAG. Subcortical areas presented the largest inter-regional differences; lateral and basolateral amygdala had markedly higher expression than other subcortical areas. The widespread differences and unique expression patterns of the different genes involved in Nogo-like signaling suggest that the functional complexes could look vastly different in different areas.
Collapse
Affiliation(s)
| | - Lars Olson
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | | |
Collapse
|
36
|
Liu H, He J, Zhong J, Zhang H, Zhang Z, Liu L, Huang Z, Wu Y, Jiang L, Guo Z, Xu R, Chai W, Huo G, Sun X, Cheng C. Clinical and Basic Evaluation of the Prognostic Value of Uric Acid in Traumatic Brain Injury. Int J Med Sci 2018; 15:1072-1082. [PMID: 30013449 PMCID: PMC6036155 DOI: 10.7150/ijms.25799] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 06/08/2018] [Indexed: 02/07/2023] Open
Abstract
Background: As a major antioxidant in serum, uric acid (UA) was once considered only as the leading cause of gout; however, recent studies have validated its neuroprotective role in ischemic stroke. Because the potential protective effects of UA in traumatic brain injury (TBI) remain largely unknown, this study investigated the role of UA in TBI in both clinical patients and experimental animals. Methods: In TBI patients, serum UA concentrations were measured within 3 days after injury. Clinical outcomes at discharge were classified according to the Glasgow Outcome Scale: good outcome (4-5) and poor outcome (1-3). Risk factors for good outcome were identified via backward logistic regression analysis. For the animal study, a controlled cortical impact (CCI) injury model was established in mice. These mice were given UA at different doses intraperitoneally, and subsequent UA concentrations in mouse serum and brain tissue were determined. Neurological function, oxidative stress, inflammatory response, neuronal maintenance, cerebral blood flow, and lesion size were also assessed. Results: The serum UA level was significantly lower in TBI patients who had a good outcome (P<0.01), and low serum UA was an independent predictor of good outcome after TBI (P<0.01; odds ratio, 0.023; 95% confidence interval, 0.006-0.082). Consistently, decreased levels of serum UA were observed in both TBI patients and CCI animals (P<0.05), whereas the UA concentration was increased in CCI brain tissue (P<0.05). Administration of UA further increased the UA level in brain tissue as compared to that in control animals (P<0.05). Among the different doses administered, 16 mg/kg UA improved sensorimotor functional recovery, spatial learning, and memory in CCI mice (P<0.05). Moreover, oxidative stress and the inflammatory response were inhibited by UA treatment (P<0.05). UA treatment also improved neuronal maintenance and cortical blood flow (P<0.05) but not lesion size (P>0.05). Conclusions: UA acted to attenuate neuronal loss, cerebral perfusion impairment and neurological deficits in TBI mice through suppression of neuronal and vascular oxidative stress. Following TBI, active antioxidant defense in the brain may result in consumption of UA in the serum, and thus, a decreased serum UA level could be predictive of good clinical recovery.
Collapse
Affiliation(s)
- Han Liu
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Junchi He
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jianjun Zhong
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hongrong Zhang
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhaosi Zhang
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Liu Liu
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhijian Huang
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yue Wu
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Jiang
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zongduo Guo
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Rui Xu
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Weina Chai
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Gang Huo
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaochuan Sun
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chongjie Cheng
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
37
|
Xie Q, Chen X, Deng H, Liu D, Sun Y, Zhou X, Yang Y, Han H. An automated pipeline for bouton, spine, and synapse detection of in vivo two-photon images. BioData Min 2017; 10:40. [PMID: 29270230 PMCID: PMC5738741 DOI: 10.1186/s13040-017-0161-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Accepted: 12/04/2017] [Indexed: 11/15/2022] Open
Abstract
Background In the nervous system, the neurons communicate through synapses. The size, morphology, and connectivity of these synapses are significant in determining the functional properties of the neural network. Therefore, they have always been a major focus of neuroscience research. Two-photon laser scanning microscopy allows the visualization of synaptic structures in vivo, leading to many important findings. However, the identification and quantification of structural imaging data currently rely heavily on manual annotation, a method that is both time-consuming and prone to bias. Results We present an automated approach for the identification of synaptic structures in two-photon images. Axon boutons and dendritic spines are structurally distinct. They can be detected automatically using this image processing method. Then, synapses can be identified by integrating information from adjacent axon boutons and dendritic spines. In this study, we first detected the axonal boutons and dendritic spines respectively, and then identified synapses based on these results. Experimental results were validated manually, and the effectiveness of our proposed method was demonstrated. Conclusions This approach will helpful for neuroscientists to automatically analyze and quantify the formation, elimination and destabilization of the axonal boutons, dendritic spines and synapses.
Collapse
Affiliation(s)
- Qiwei Xie
- Research Base of Beijing Modern Manufacturing Development, No.100, Pingleyuan, Beijing, 100124 China.,Data Mining Lab, School of Management, Beijing University of Technology, No.100, Pingleyuan, Beijing, 100124 China.,Institute of Automation, Chinese Academy of Sciences, 95 Zhongguancun East Road, Beijing, 100190 China
| | - Xi Chen
- Institute of Automation, Chinese Academy of Sciences, 95 Zhongguancun East Road, Beijing, 100190 China
| | - Hao Deng
- Faculty of Information Technology, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau, China
| | - Danqian Liu
- Institute of Neuroscience, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031 China
| | - Yingyu Sun
- Beijing Normal University, No. 19, Waida Jie, Xinjie Kou, Beijing, 100875 China
| | - Xiaojuan Zhou
- Beijing Normal University, No. 19, Waida Jie, Xinjie Kou, Beijing, 100875 China
| | - Yang Yang
- Institute of Neuroscience, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031 China.,Center for Excellence in Brain Science and Intelligence Technology Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031 China
| | - Hua Han
- Institute of Automation, Chinese Academy of Sciences, 95 Zhongguancun East Road, Beijing, 100190 China.,Center for Excellence in Brain Science and Intelligence Technology Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031 China.,University of Chinese Academy of Sciences, School of future technology, No.19(A) Yuquan Road, Beijing, 100049 China
| |
Collapse
|
38
|
MT3-MMP Promotes Excitatory Synapse Formation by Promoting Nogo-66 Receptor Ectodomain Shedding. J Neurosci 2017; 38:518-529. [PMID: 29196321 DOI: 10.1523/jneurosci.0962-17.2017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 10/23/2017] [Accepted: 11/20/2017] [Indexed: 12/14/2022] Open
Abstract
Cell-surface molecules are dynamically regulated at the synapse to assemble and disassemble adhesive contacts that are important for synaptogenesis and for tuning synaptic transmission. Metalloproteinases dynamically regulate cellular behaviors through the processing of cell surface molecules. In the present study, we evaluated the role of membrane-type metalloproteinases (MT-MMPs) in excitatory synaptogenesis. We find that MT3-MMP and MT5-MMP are broadly expressed in the mouse cerebral cortex and that MT3-MMP loss-of-function interferes with excitatory synapse development in dissociated cortical neurons and in vivo We identify Nogo-66 receptor (NgR1) as an MT3-MMP substrate that is required for MT3-MMP-dependent synapse formation. Introduction of the shed ectodomain of NgR1 is sufficient to accelerate excitatory synapse formation in dissociated cortical neurons and in vivo Together, our findings support a role for MT3-MMP-dependent shedding of NgR1 in regulating excitatory synapse development.SIGNIFICANCE STATEMENT In this study, we identify MT3-MMP, a membrane-bound zinc protease, to be necessary for the development of excitatory synapses in cortical neurons. We identify Nogo-66 receptors (NgR1) as a downstream target of MT3-MMP proteolytic activity. Furthermore, processing of surface NgR1 by MT3-MMP generates a soluble ectodomain fragment that accelerates the formation of excitatory synapses. We propose that MT3-MMP activity and NgR1 shedding could stimulate circuitry remodeling in the adult brain and enhance functional connectivity after brain injury.
Collapse
|
39
|
Zhao Y, Sivaji S, Chiang MC, Ali H, Zukowski M, Ali S, Kennedy B, Sklyar A, Cheng A, Guo Z, Reed AK, Kodali R, Borowski J, Frost G, Beukema P, Wills ZP. Amyloid Beta Peptides Block New Synapse Assembly by Nogo Receptor-Mediated Inhibition of T-Type Calcium Channels. Neuron 2017; 96:355-372.e6. [PMID: 29024660 DOI: 10.1016/j.neuron.2017.09.041] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 07/27/2017] [Accepted: 09/22/2017] [Indexed: 01/07/2023]
Abstract
Compelling evidence links amyloid beta (Aβ) peptide accumulation in the brains of Alzheimer's disease (AD) patients with the emergence of learning and memory deficits, yet a clear understanding of the events that drive this synaptic pathology are lacking. We present evidence that neurons exposed to Aβ are unable to form new synapses, resulting in learning deficits in vivo. We demonstrate the Nogo receptor family (NgR1-3) acts as Aβ receptors mediating an inhibition of synapse assembly, plasticity, and learning. Live imaging studies reveal Aβ activates NgRs on the dendritic shaft of neurons, triggering an inhibition of calcium signaling. We define T-type calcium channels as a target of Aβ-NgR signaling, mediating Aβ's inhibitory effects on calcium, synapse assembly, plasticity, and learning. These studies highlight deficits in new synapse assembly as a potential initiator of cognitive pathology in AD, and pinpoint calcium dysregulation mediated by NgRs and T-type channels as key components. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Yanjun Zhao
- Department of Neurobiology, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Sivaprakash Sivaji
- Department of Neurobiology, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Michael C Chiang
- Department of Neurobiology, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Haadi Ali
- Department of Neurobiology, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Monica Zukowski
- Department of Neurobiology, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Sareen Ali
- Department of Neurobiology, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Bryan Kennedy
- Department of Neurobiology, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Alex Sklyar
- Department of Neurobiology, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Alice Cheng
- Department of Neurobiology, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Zihan Guo
- Department of Neurobiology, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Alexander K Reed
- Department of Neurobiology, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Ravindra Kodali
- Department of Structural Biology and Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Jennifer Borowski
- Department of Neurobiology, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Georgia Frost
- Department of Neurobiology, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Patrick Beukema
- Center for the Neural Basis of Cognition, Carnegie Mellon University, Pittsburgh, PA 15213, USA; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Zachary P Wills
- Department of Neurobiology, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA.
| |
Collapse
|
40
|
Otero-Ortega L, Gómez-de Frutos MC, Laso-García F, Sánchez-Gonzalo A, Martínez-Arroyo A, Díez-Tejedor E, Gutiérrez-Fernández M. NogoA Neutralization Promotes Axonal Restoration After White Matter Injury In Subcortical Stroke. Sci Rep 2017; 7:9431. [PMID: 28842591 PMCID: PMC5573364 DOI: 10.1038/s41598-017-09705-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 07/27/2017] [Indexed: 12/19/2022] Open
Abstract
Blocking axonal growth inhibitor NogoA has been of great interest for promoting axonal recovery from neurological diseases. The present study investigates the therapeutic effects of blocking NogoA, inducing functional recovery and promoting white matter repair in an experimental animal model of stroke. Adult male rats were subjected to white matter injury by subcortical ischemic stroke. Twenty-four hours after surgery, 250 ug of anti-NogoA or anti-IgG-1 were administered through the tail vein. The quantity of NogoA protein was determined by immunohistochemistry in the brain and peripheral organs. In addition, functional status, lesion size, fiber tract integrity, axonal sprouting and white matter repair markers were analyzed. Moreover, an in vitro study was performed in order to strengthen the results obtained in vivo. A lower quantity of NogoA protein was found in the brain and peripheral organs of the animals that received anti-NogoA treatment. The animals receiving anti-NogoA treatment showed significantly better results in terms of functional recovery, fiber tract integrity, axonal sprouting and white matter repair markers compared with the control group at 28 days. White matter integrity was in part restored by antibody-mediated inhibition of NogoA administration in those animals that were subjected to an axonal injury by subcortical stroke. This white matter restoration triggered functional recovery.
Collapse
Affiliation(s)
- Laura Otero-Ortega
- Neuroscience and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Center, La Paz University Hospital, Neuroscience Area of IdiPAZ Health Research Institute, Autonomous University of Madrid, Madrid, Spain
| | - Mari Carmen Gómez-de Frutos
- Neuroscience and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Center, La Paz University Hospital, Neuroscience Area of IdiPAZ Health Research Institute, Autonomous University of Madrid, Madrid, Spain
| | - Fernando Laso-García
- Neuroscience and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Center, La Paz University Hospital, Neuroscience Area of IdiPAZ Health Research Institute, Autonomous University of Madrid, Madrid, Spain
| | - Alba Sánchez-Gonzalo
- Neuroscience and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Center, La Paz University Hospital, Neuroscience Area of IdiPAZ Health Research Institute, Autonomous University of Madrid, Madrid, Spain
| | - Arturo Martínez-Arroyo
- Neuroscience and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Center, La Paz University Hospital, Neuroscience Area of IdiPAZ Health Research Institute, Autonomous University of Madrid, Madrid, Spain
| | - Exuperio Díez-Tejedor
- Neuroscience and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Center, La Paz University Hospital, Neuroscience Area of IdiPAZ Health Research Institute, Autonomous University of Madrid, Madrid, Spain.
| | - María Gutiérrez-Fernández
- Neuroscience and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Center, La Paz University Hospital, Neuroscience Area of IdiPAZ Health Research Institute, Autonomous University of Madrid, Madrid, Spain.
| |
Collapse
|
41
|
Neural Glycosylphosphatidylinositol-Anchored Proteins in Synaptic Specification. Trends Cell Biol 2017; 27:931-945. [PMID: 28743494 DOI: 10.1016/j.tcb.2017.06.007] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Revised: 06/27/2017] [Accepted: 06/29/2017] [Indexed: 12/15/2022]
Abstract
Glycosylphosphatidylinositol (GPI)-anchored proteins are a specialized class of lipid-associated neuronal membrane proteins that perform diverse functions in the dynamic control of axon guidance, synaptic adhesion, cytoskeletal remodeling, and localized signal transduction, particularly at lipid raft domains. Recent studies have demonstrated that a subset of GPI-anchored proteins act as critical regulators of synapse development by modulating specific synaptic adhesion pathways via direct interactions with key synapse-organizing proteins. Additional studies have revealed that alteration of these regulatory mechanisms may underlie various brain disorders. In this review, we highlight the emerging role of GPI-anchored proteins as key synapse organizers that aid in shaping the properties of various types of synapses and circuits in mammals.
Collapse
|
42
|
Nogo Receptor 1 Confines a Disinhibitory Microcircuit to the Critical Period in Visual Cortex. J Neurosci 2017; 36:11006-11012. [PMID: 27798181 DOI: 10.1523/jneurosci.0935-16.2016] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 09/02/2016] [Indexed: 11/21/2022] Open
Abstract
A characteristic of the developing mammalian visual system is a brief interval of plasticity, termed the "critical period," when the circuitry of primary visual cortex is most sensitive to perturbation of visual experience. Depriving one eye of vision (monocular deprivation [MD]) during the critical period alters ocular dominance (OD) by shifting the responsiveness of neurons in visual cortex to favor the nondeprived eye. A disinhibitory microcircuit involving parvalbumin-expressing (PV) interneurons initiates this OD plasticity. The gene encoding the neuronal nogo-66-receptor 1 (ngr1/rtn4r) is required to close the critical period. Here we combined mouse genetics, electrophysiology, and circuit mapping with laser-scanning photostimulation to investigate whether disinhibition is confined to the critical period by ngr1 We demonstrate that ngr1 mutant mice retain plasticity characteristic of the critical period as adults, and that ngr1 operates within PV interneurons to restrict the loss of intracortical excitatory synaptic input following MD in adult mice, and this disinhibition induces a "lower PV network configuration" in both critical-period wild-type mice and adult ngr1-/- mice. We propose that ngr1 limits disinhibition to close the critical period for OD plasticity and that a decrease in PV expression levels reports the diminished recent cumulative activity of these interneurons. SIGNIFICANCE STATEMENT Life experience refines brain circuits throughout development during specified critical periods. Abnormal experience during these critical periods can yield enduring maladaptive changes in neural circuits that impair brain function. In the developing visual system, visual deprivation early in life can result in amblyopia (lazy-eye), a prevalent childhood disorder comprising permanent deficits in spatial vision. Here we identify that the nogo-66 receptor 1 gene restricts an early and essential step in OD plasticity to the critical period. These findings link the emerging circuit-level description of OD plasticity to the genetic regulation of the critical period. Understanding how plasticity is confined to critical periods may provide clues how to better treat amblyopia.
Collapse
|
43
|
Hypertension-induced synapse loss and impairment in synaptic plasticity in the mouse hippocampus mimics the aging phenotype: implications for the pathogenesis of vascular cognitive impairment. GeroScience 2017; 39:385-406. [PMID: 28664509 DOI: 10.1007/s11357-017-9981-y] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 05/30/2017] [Indexed: 12/22/2022] Open
Abstract
Strong epidemiological and experimental evidence indicates that hypertension has detrimental effects on the cerebral microcirculation and thereby promotes accelerated brain aging. Hypertension is an independent risk factor for both vascular cognitive impairment (VCI) and Alzheimer's disease (AD). However, the pathophysiological link between hypertension-induced cerebromicrovascular injury (e.g., blood-brain barrier disruption, increased microvascular oxidative stress, and inflammation) and cognitive decline remains elusive. The present study was designed to characterize neuronal functional and morphological alterations induced by chronic hypertension and compare them to those induced by aging. To achieve that goal, we induced hypertension in young C57BL/6 mice by chronic (4 weeks) infusion of angiotensin II. We found that long-term potentiation (LTP) of performant path synapses following high-frequency stimulation of afferent fibers was decreased in hippocampal slices obtained from hypertensive mice, mimicking the aging phenotype. Hypertension and advanced age were associated with comparable decline in synaptic density in the stratum radiatum of the mouse hippocampus. Hypertension, similar to aging, was associated with changes in mRNA expression of several genes involved in regulation of neuronal function, including down-regulation of Bdnf, Homer1, and Dlg4, which may have a role in impaired synaptic plasticity. Collectively, hypertension impairs synaptic plasticity, reduces synaptic density, and promotes dysregulation of genes involved in synaptic function in the mouse hippocampus mimicking the aging phenotype. These hypertension-induced neuronal alterations may impair establishment of memories in the hippocampus and contribute to the pathogenesis and clinical manifestation of both vascular cognitive impairment (VCI) and Alzheimer's disease (AD).
Collapse
|
44
|
Bodrikov V, Welte C, Wiechers M, Weschenfelder M, Kaur G, Shypitsyna A, Pinzon-Olejua A, Bastmeyer M, Stuermer CAO. Substrate properties of zebrafish Rtn4b/Nogo and axon regeneration in the zebrafish optic nerve. J Comp Neurol 2017; 525:2991-3009. [PMID: 28560734 DOI: 10.1002/cne.24253] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 05/16/2017] [Accepted: 05/24/2017] [Indexed: 11/08/2022]
Abstract
This study explored why lesioned retinal ganglion cell (RGC) axons regenerate successfully in the zebrafish optic nerve despite the presence of Rtn4b, the homologue of the rat neurite growth inhibitor RTN4-A/Nogo-A. Rat Nogo-A and zebrafish Rtn4b possess characteristic motifs (M1-4) in the Nogo-A-specific region, which contains delta20, the most inhibitory region of rat Nogo-A. To determine whether zebrafish M1-4 is inhibitory as rat M1-4 and Nogo-A delta20, proteins were recombinantly expressed and used as substrates for zebrafish single cell RGCs, mouse hippocampal neurons and goldfish, zebrafish and chick retinal explants. When offered as homogenous substrates, neurites of hippocampal neurons and of zebrafish single cell RGCs were inhibited by zebrafish M1-4, rat M1-4, and Nogo-A delta20. Neurite length increased when zebrafish single cell RGCs were treated with receptor-type-specific antagonists and, respectively, with morpholinos (MO) against S1PR2 and S1PR5a-which represent candidate zebrafish Nogo-A receptors. In a stripe assay, however, where M1-4 lanes alternate with polylysine-(Plys)-only lanes, RGC axons from goldfish, zebrafish, and chick retinal explants avoided rat M1-4 but freely crossed zebrafish M1-4 lanes-suggesting that zebrafish M1-4 is growth permissive and less inhibitory than rat M1-4. Moreover, immunostainings and dot blots of optic nerve and myelin showed that expression of Rtn4b is very low in tissue and myelin at 3-5 days after lesion when axons regenerate. Thus, Rtn4b seems to represent no major obstacle for axon regeneration in vivo because it is less inhibitory for RGC axons from retina explants, and because of its low abundance.
Collapse
Affiliation(s)
| | - Cornelia Welte
- Department of Biology, University of Konstanz, Konstanz, Germany
| | | | - Markus Weschenfelder
- Zoological Institute, Cell and Neurobiology Biology, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Gurjot Kaur
- Department of Biology, University of Konstanz, Konstanz, Germany
| | | | | | - Martin Bastmeyer
- Zoological Institute, Cell and Neurobiology Biology, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | | |
Collapse
|
45
|
Kurihara Y, Saito Y, Takei K. Blockade of chondroitin sulfate proteoglycans-induced axonal growth inhibition by LOTUS. Neuroscience 2017; 356:265-274. [PMID: 28571719 DOI: 10.1016/j.neuroscience.2017.05.034] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 05/19/2017] [Accepted: 05/19/2017] [Indexed: 12/14/2022]
Abstract
Chondroitin sulfate proteoglycans (CSPGs) are axon growth inhibitors in the glial scar, and restrict axon regeneration following damage to the adult mammalian central nervous system. CSPGs have recently been identified as functional ligands for Nogo receptor-1 (NgR1), which is the common receptor for Nogo proteins, myelin-associated glycoprotein (MAG), oligodendrocyte myelin glycoprotein (OMgp) and B lymphocyte stimulator (BLyS). We have previously reported that through its binding to NgR1, lateral olfactory tract usher substance (LOTUS) suppresses Nogo, MAG, OMgp, and BLyS-induced axon growth inhibition. However, it remains unknown whether LOTUS also exerts this suppressive action on CSPG-induced axon growth inhibition. LOTUS overexpression rescued CSPG-induced growth cone collapse and neurite outgrowth inhibition in cultured dorsal root ganglion neurons, which only weakly express endogenous LOTUS. In cultured olfactory bulb neurons, which endogenously express LOTUS, the growth cone was insensitive to CSPG-induced collapse, but was sensitive to collapse induced by CSPGs in lotus-deficient mice. Our data demonstrate that LOTUS suppresses CSPG-induced axon growth inhibition, suggesting that LOTUS may represent a promising therapeutic agent for promoting axon regeneration.
Collapse
Affiliation(s)
- Yuji Kurihara
- Molecular Medical Bioscience Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, Suehiro-cho 1-7-29, Tsurumi-ward, Yokohama 230-0045, Japan
| | - Yu Saito
- Molecular Medical Bioscience Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, Suehiro-cho 1-7-29, Tsurumi-ward, Yokohama 230-0045, Japan
| | - Kohtaro Takei
- Molecular Medical Bioscience Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, Suehiro-cho 1-7-29, Tsurumi-ward, Yokohama 230-0045, Japan.
| |
Collapse
|
46
|
Karlsson TE, Wellfelt K, Olson L. Spatiotemporal and Long Lasting Modulation of 11 Key Nogo Signaling Genes in Response to Strong Neuroexcitation. Front Mol Neurosci 2017; 10:94. [PMID: 28442990 PMCID: PMC5386981 DOI: 10.3389/fnmol.2017.00094] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 03/20/2017] [Indexed: 12/13/2022] Open
Abstract
Inhibition of nerve growth and plasticity in the CNS is to a large part mediated by Nogo-like signaling, now encompassing a plethora of ligands, receptors, co-receptors and modulators. Here we describe the distribution and levels of mRNA encoding 11 key genes involved in Nogo-like signaling (Nogo-A, Oligodendrocyte-Myelin glycoprotein (OMgp), Nogo receptor 1 (NgR1), NgR2, NgR3, Lingo-1, TNF receptor orphan Y (Troy), Olfactomedin, Lateral olfactory tract usher substance (Lotus) and membrane-type matrix metalloproteinase-3 (MT3-MPP)), as well as BDNF and GAPDH. Expression was analyzed in nine different brain areas before, and at eight time points during the first 3 days after a strong neuroexcitatory stimulation, caused by one kainic acid injection. A temporo-spatial pattern of orderly transcriptional regulations emerges that strengthens the role of Nogo-signaling mechanisms for synaptic plasticity in synchrony with transcriptional increases of BDNF mRNA. For most Nogo-type signaling genes, the largest alterations of mRNA levels occur in the dentate gyrus, with marked alterations also in the CA1 region. Changes occurred somewhat later in several areas of the cerebral cortex. The detailed spatio-temporal pattern of mRNA presence and kainic acid-induced transcriptional response is gene-specific. We reveal that several different gene alterations combine to decrease (and later increase) Nogo-like signaling, as expected to allow structural plasticity responses. Other genes are altered in the opposite direction, suggesting that the system prepares in advance in order to rapidly restore balance. However, the fact that Lingo-1 shows a seemingly opposite, plasticity inhibiting response to kainic acid (strong increase of mRNA in the dentate gyrus), may instead suggest a plasticity-enhancing intracellular function of this presumed NgR1 co-receptor.
Collapse
Affiliation(s)
| | - Katrin Wellfelt
- Department of Neuroscience, Karolinska InstitutetStockholm, Sweden
| | - Lars Olson
- Department of Neuroscience, Karolinska InstitutetStockholm, Sweden
| |
Collapse
|
47
|
Pernet V. Nogo-A in the visual system development and in ocular diseases. Biochim Biophys Acta Mol Basis Dis 2017; 1863:1300-1311. [PMID: 28408340 DOI: 10.1016/j.bbadis.2017.04.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 04/08/2017] [Accepted: 04/09/2017] [Indexed: 01/02/2023]
Abstract
Nogo-A is a potent myelin-associated inhibitor for neuronal growth and plasticity in the central nervous system (CNS). Its effects are mediated by the activation of specific receptors that intracellularly control cytoskeleton rearrangements, protein synthesis and gene expression. Moreover, Nogo-A has been involved in the development of the visual system and in a variety of neurodegenerative diseases and injury processes that can alter its function. For example, Nogo-A was shown to influence optic nerve myelinogenesis, the formation and maturation of retinal axon projections, and retinal angiogenesis. In adult animals, the inactivation of Nogo-A exerted remarkable effects on visual plasticity. Relieving Nogo-A-induced inhibition increased axonal sprouting after optic nerve lesion and axonal rewiring in the visual cortex of intact adult mice. This review aims at presenting our current knowledge on the role of Nogo-A in the visual system and to discuss how its therapeutic targeting may promote visual improvement in ophthalmic diseases.
Collapse
Affiliation(s)
- Vincent Pernet
- CUO-Recherche, Centre de recherche du CHU de Québec and Département d'ophtalmologie, Faculté de médecine, Université Laval, Québec, Québec, Canada.
| |
Collapse
|
48
|
Abstract
Most of the current therapies, as well as many of the clinical trials, for multiple sclerosis (MS) target the inflammatory autoimmune processes, but less than 20% of all clinical trials investigate potential therapies for the chronic progressive disease stage of MS. The latter is responsible for the steadily increasing disability in many patients, and there is an urgent need for novel therapies that protect nervous system tissue and enhance axonal growth and/or remyelination. As outlined in this review, solid pre-clinical data suggest neutralization of the neurite outgrowth inhibitor Nogo-A as a potential new way to achieve both axonal and myelin repair. Several phase I clinical studies with anti-Nogo-A antibodies have been conducted in different disease paradigms including MS and spinal cord injury. Data from spinal cord injury and amyotrophic lateral sclerosis (ALS) trials accredit a good safety profile of high doses of anti-Nogo-A antibodies administered intravenously or intrathecally. An antibody against a Nogo receptor subunit, leucine rich repeat and immunoglobulin-like domain-containing protein 1 (LINGO-1), was recently shown to improve outcome in patients with acute optic neuritis in a phase II study. Nogo-A-suppressing antibodies could be novel drug candidates for the relapsing as well as the progressive MS disease stage. In this review, we summarize the available pre-clinical and clinical evidence on Nogo-A and elucidate the potential of Nogo-A-antibodies as a therapy for progressive MS.
Collapse
|
49
|
Boghdadi AG, Teo L, Bourne JA. The Involvement of the Myelin-Associated Inhibitors and Their Receptors in CNS Plasticity and Injury. Mol Neurobiol 2017; 55:1831-1846. [PMID: 28229330 DOI: 10.1007/s12035-017-0433-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 01/31/2017] [Indexed: 12/21/2022]
Abstract
The limited capacity for the central nervous system (CNS) to repair itself was first described over 100 years ago by Spanish neuroscientist Ramon Y. Cajal. However, the exact mechanisms underlying this failure in neuronal regeneration remain unclear and, as such, no effective therapeutics yet exist. Numerous studies have attempted to elucidate the biochemical and molecular mechanisms that inhibit neuronal repair with increasing evidence suggesting that several inhibitory factors and repulsive guidance cues active during development actually persist into adulthood and may be contributing to the inhibition of repair. For example, in the injured adult CNS, there are various inhibitory factors that impede the outgrowth of neurites from damaged neurons. One of the most potent of these neurite outgrowth inhibitors is the group of proteins known as the myelin-associated inhibitors (MAIs), present mainly on the membranes of oligodendroglia. Several studies have shown that interfering with these proteins can have positive outcomes in CNS injury models by promoting neurite outgrowth and improving functional recovery. As such, the MAIs, their receptors, and downstream effectors are valid drug targets for the treatment of CNS injury. This review will discuss the current literature on MAIs in the context of CNS development, plasticity, and injury. Molecules that interfere with the MAIs and their receptors as potential candidates for the treatment of CNS injury will additionally be introduced in the context of preclinical and clinical trials.
Collapse
Affiliation(s)
- Anthony G Boghdadi
- Australian Regenerative Medicine Institute, Monash University, 15 Innovation Walk (Building 75), Clayton, VIC, 3800, Australia
| | - Leon Teo
- Australian Regenerative Medicine Institute, Monash University, 15 Innovation Walk (Building 75), Clayton, VIC, 3800, Australia
| | - James A Bourne
- Australian Regenerative Medicine Institute, Monash University, 15 Innovation Walk (Building 75), Clayton, VIC, 3800, Australia.
| |
Collapse
|
50
|
Nogo-A regulates spatial learning as well as memory formation and modulates structural plasticity in the adult mouse hippocampus. Neurobiol Learn Mem 2017; 138:154-163. [DOI: 10.1016/j.nlm.2016.06.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 06/21/2016] [Accepted: 06/23/2016] [Indexed: 11/21/2022]
|