1
|
Jiang L, Huang L, Dai C, Zheng R, Miyake M, Mori Y, Nakao S, Morino K, Ymashiro K, Miao Y, Li Q, Ren W, Ye Z, Li H, Yang Z, Shi Y. Genome-Wide Association Analysis Identifies LILRB2 Gene for Pathological Myopia. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308968. [PMID: 39207058 PMCID: PMC11516067 DOI: 10.1002/advs.202308968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 07/13/2024] [Indexed: 09/04/2024]
Abstract
Pathological myopia (PM) is one of the leading causes of blindness, especially in Asia. To identify the genetic risk factors of PM, a two-stage genome-wide association study (GWAS) and replication analysis in East Asian populations is conducted. The analysis identified LILRB2 in 19q13.42 as a new candidate locus for PM. The increased protein expression of LILRB2/Pirb (mouse orthologous protein) in PM patients and myopia mouse models is validated. It is further revealed that the increase in LILRB2/Pirb promoted fatty acid synthesis and lipid accumulation, leading to the destruction of choroidal function and the development of PM. This study revealed the association between LILRB2 and PM, uncovering the molecular mechanism of lipid metabolism disorders leading to the pathogenesis of PM due to LILRB2 upregulation.
Collapse
Affiliation(s)
- Lingxi Jiang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical GeneticsDepartment of Laboratory MedicineSichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduSichuan610072China
- Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026)Sichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalChengduSichuan610072China
| | - Lulin Huang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical GeneticsDepartment of Laboratory MedicineSichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduSichuan610072China
- Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026)Sichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalChengduSichuan610072China
| | - Chao Dai
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical GeneticsDepartment of Laboratory MedicineSichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduSichuan610072China
| | - Rui Zheng
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical GeneticsDepartment of Laboratory MedicineSichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduSichuan610072China
| | - Masahiro Miyake
- Department of Ophthalmology and Visual SciencesKyoto University Graduate School of MedicineKyoto606‐8501Japan
| | - Yuki Mori
- Department of Ophthalmology and Visual SciencesKyoto University Graduate School of MedicineKyoto606‐8501Japan
| | - Shin‐ya Nakao
- Department of Ophthalmology and Visual SciencesKyoto University Graduate School of MedicineKyoto606‐8501Japan
| | - Kazuya Morino
- Department of Ophthalmology and Visual SciencesKyoto University Graduate School of MedicineKyoto606‐8501Japan
| | - Kenji Ymashiro
- Department of Ophthalmology and Visual SciencesKyoto University Graduate School of MedicineKyoto606‐8501Japan
| | - Yang‐Bao Miao
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical GeneticsDepartment of Laboratory MedicineSichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduSichuan610072China
| | - Qi Li
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical GeneticsDepartment of Laboratory MedicineSichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduSichuan610072China
| | - Weiming Ren
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical GeneticsDepartment of Laboratory MedicineSichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduSichuan610072China
| | - Zimeng Ye
- School of MedicineUniversity of SydneyCamperdownNSW2050Australia
| | - Hongjing Li
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical GeneticsDepartment of Laboratory MedicineSichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduSichuan610072China
- Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026)Sichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalChengduSichuan610072China
| | - Zhenglin Yang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical GeneticsDepartment of Laboratory MedicineSichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduSichuan610072China
- Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026)Sichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalChengduSichuan610072China
- Jinfeng Laboratory, Chongqing, ChinaChongqing400000China
| | - Yi Shi
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical GeneticsDepartment of Laboratory MedicineSichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduSichuan610072China
- Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026)Sichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalChengduSichuan610072China
| |
Collapse
|
2
|
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
|
3
|
Seng C, Luo W, Földy C. Circuit formation in the adult brain. Eur J Neurosci 2022; 56:4187-4213. [PMID: 35724981 PMCID: PMC9546018 DOI: 10.1111/ejn.15742] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 06/08/2022] [Accepted: 06/09/2022] [Indexed: 11/30/2022]
Abstract
Neurons in the mammalian central nervous system display an enormous capacity for circuit formation during development but not later in life. In principle, new circuits could be also formed in adult brain, but the absence of the developmental milieu and the presence of growth inhibition and hundreds of working circuits are generally viewed as unsupportive for such a process. Here, we bring together evidence from different areas of neuroscience—such as neurological disorders, adult‐brain neurogenesis, innate behaviours, cell grafting, and in vivo cell reprogramming—which demonstrates robust circuit formation in adult brain. In some cases, adult‐brain rewiring is ongoing and required for certain types of behaviour and memory, while other cases show significant promise for brain repair in disease models. Together, these examples highlight that the adult brain has higher capacity for structural plasticity than previously recognized. Understanding the underlying mechanisms behind this retained plasticity has the potential to advance basic knowledge regarding the molecular organization of synaptic circuits and could herald a new era of neural circuit engineering for therapeutic repair.
Collapse
Affiliation(s)
- Charlotte Seng
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Science, University of Zurich, Zürich, Switzerland
| | - Wenshu Luo
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Science, University of Zurich, Zürich, Switzerland
| | - Csaba Földy
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Science, University of Zurich, Zürich, Switzerland
| |
Collapse
|
4
|
Joly S, Mdzomba JB, Rodriguez L, Morin F, Vallières L, Pernet V. B cell-dependent EAE induces visual deficits in the mouse with similarities to human autoimmune demyelinating diseases. J Neuroinflammation 2022; 19:54. [PMID: 35197067 PMCID: PMC8867627 DOI: 10.1186/s12974-022-02416-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 02/07/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In the field of autoimmune demyelinating diseases, visual impairments have extensively been studied using the experimental autoimmune encephalomyelitis (EAE) mouse model, which is classically induced by immunization with myelin oligodendrocyte glycoprotein peptide (MOG35-55). However, this model does not involve B cells like its human analogs. New antigens have thus been developed to induce a B cell-dependent form of EAE that better mimics human diseases. METHODS The present study aimed to characterize the visual symptoms of EAE induced with such an antigen called bMOG. After the induction of EAE with bMOG in C57BL/6J mice, visual function changes were studied by electroretinography and optomotor acuity tests. Motor deficits were assessed in parallel with a standard clinical scoring method. Histological examinations and Western blot analyses allowed to follow retinal neuron survival, gliosis, microglia activation, opsin photopigment expression in photoreceptors and optic nerve demyelination. Disease effects on retinal gene expression were established by RNA sequencing. RESULTS We observed that bMOG EAE mice exhibited persistent loss of visual acuity, despite partial recovery of electroretinogram and motor functions. This loss was likely due to retinal inflammation, gliosis and synaptic impairments, as evidenced by histological and transcriptomic data. Further analysis suggests that the M-cone photoreceptor pathway was also affected. CONCLUSION Therefore, by documenting visual changes induced by bMOG and showing similarities to those seen in diseases such as multiple sclerosis and neuromyelitis optica, this study offers a new approach to test protective or restorative ophthalmic treatments.
Collapse
Affiliation(s)
- Sandrine Joly
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Regenerative Medicine Unit, University Hospital Center of Quebec, Laval University, Quebec City, QC Canada
| | - Julius Baya Mdzomba
- Regenerative Medicine Unit, University Hospital Center of Quebec, Laval University, Quebec City, QC Canada
- Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec City, QC Canada
| | - Léa Rodriguez
- Regenerative Medicine Unit, University Hospital Center of Quebec, Laval University, Quebec City, QC Canada
- Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec City, QC Canada
| | - Françoise Morin
- Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec City, QC Canada
- Neuroscience Unit, University Hospital Center of Quebec, Laval University, Quebec City, QC Canada
| | - Luc Vallières
- Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec City, QC Canada
- Neuroscience Unit, University Hospital Center of Quebec, Laval University, Quebec City, QC Canada
| | - Vincent Pernet
- Regenerative Medicine Unit, University Hospital Center of Quebec, Laval University, Quebec City, QC Canada
- Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec City, QC Canada
- Center for Experimental Neurology (ZEN), University of Bern, Bern, Switzerland
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Sahli Haus 1, UG Büro 1, Freiburgstrasse 14, 3010 Bern, Switzerland
| |
Collapse
|
5
|
Johnson MA, Mehrabian Z, Guo Y, Ghosh J, Brigell MG, Bernstein SL. Anti-NOGO Antibody Neuroprotection in a Rat Model of NAION. Transl Vis Sci Technol 2021; 10:12. [PMID: 34904998 PMCID: PMC8684297 DOI: 10.1167/tvst.10.14.12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Purpose To evaluate the efficacy and mechanisms of anti-NOGO receptor monoclonal antibody 11C7mAb in a rat model of nonarteritic anterior ischemic optic neuropathy (rNAION). Methods The rNAION was induced in one eye of 20 Long-Evans rats, which were studied in 10 groups of two rats, each group containing a sham rat receiving intravitreal injections of vehicle and a treatment rat receiving intravitreal injections of 11C7mAb. Fellow eyes served as naïve controls. The rats were tested using flash electroretinograms (ERGs), flash visual evoked potentials (VEPs), and optical coherence tomography (OCT). Thirty days after induction, they were euthanized, and the eyes were prepared for immunohistochemistry (two groups), hematoxylin and eosin staining (two groups) or transmission electron microscopy (TEM; six groups). Results Ninety-five percent of the VEP amplitude was preserved in eyes treated with 11C7mAb, versus 69% in the control eyes. Immunohistochemistry revealed a large reduction in microglia and extrinsic macrophages with axon sparing. In addition to axon preservation, TEM also showed reduced extracellular debris and only slight myelin damage compared with the vehicle-treated animals. There were no significant differences in retinal ganglion cell counts, nor was there a difference in optic nerve swelling as measured by OCT. ERGs were used to exclude eyes with retinal vascular occlusions, an occasional complication of the induction technique. Conclusions The 11C7mAb preserves optic nerve integrity and reduces inflammation in rNAION. Translational Relevance This study evaluates the efficacy of an anti-NOGO receptor antibody in a rat model of NAION, a disorder that currently has no universally-acknowledged treatment.
Collapse
Affiliation(s)
- Mary A Johnson
- Department of Ophthalmology & Visual Science, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Zara Mehrabian
- Department of Ophthalmology & Visual Science, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Yan Guo
- Department of Ophthalmology & Visual Science, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Joy Ghosh
- Eventide Asset Management, LLC, Boston, MA, USA
| | | | - Steven L Bernstein
- Department of Ophthalmology & Visual Science, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Anatomy and Pathology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
6
|
Johnson NP, Gregorich SM, Passaglia CL. Spatiotemporal Contrast Sensitivity of Brown-Norway Rats under Scotopic and Photopic Illumination. Neuroscience 2020; 449:63-73. [PMID: 33035619 DOI: 10.1016/j.neuroscience.2020.09.056] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/03/2020] [Accepted: 09/28/2020] [Indexed: 11/24/2022]
Abstract
Rats are a popular animal model for vision research and for investigating disorders of the visual system. The study aimed to quantify the spatiotemporal contrast sensitivity function (CSF) of healthy adult Brown-Norway rats under scotopic and photopic illumination. Animals were trained to jump onto the one of two adjacent platforms behind which was displayed a sinewave grating pattern. Contrast thresholds of light- and dark-adapted rats were determined using a staircase method of adjustment for gratings that varied in spatial frequency (sf) and temporal frequency (tf) and ranged several log-units in mean luminance. Photopic CSFs showed strong bandpass spatial tuning, consistent with prior measurements, and weak bandpass temporal tuning. CSFs were parameterized by a truncated log-parabola model, yielding a peak contrast sensitivity of 52 ± 9, peak sf of 0.17 ± 0.05 cycles/degree, sf limit of 1.6 ± 0.3 cycles/degree, low sf attenuation of 85 ± 9%, peak tf of 1.7 ± 1.1 Hz, extrapolated tf limit of 166 ± 44 Hz, and low tf attenuation of 55 ± 12%. CSFs became more lowpass and decreased systematically in contrast sensitivity and spatiotemporal acuity as mean luminance was reduced. CSFs were also measured via the visual head-tracking reflex. Photopic contrast sensitivity, spatial acuity, and temporal acuity were all markedly below that of the grating detection task and optomotor findings for other rat strains. The CSF data provide a comprehensive and quantitative description of rat spatial and temporal vision and a benchmark for evaluating effects of ocular diseases on their ability to see.
Collapse
Affiliation(s)
- Nicholas P Johnson
- University of South Florida, Molecular Pharmacology & Physiology Department, 12901 Bruce B Downs Boulevard MDC 40, Tampa, FL 33612, United States
| | - Sarah M Gregorich
- University of South Florida, Medical Engineering Department, 4202 E Fowler Avenue, Tampa, FL 33620, United States
| | - Christopher L Passaglia
- University of South Florida, Molecular Pharmacology & Physiology Department, 12901 Bruce B Downs Boulevard MDC 40, Tampa, FL 33612, United States; University of South Florida, Medical Engineering Department, 4202 E Fowler Avenue, Tampa, FL 33620, United States; University of South Florida, Ophthalmology Department, 12901 Bruce B Downs Boulevard MDC21, Tampa, FL 33612, United States.
| |
Collapse
|
7
|
Tau modulates visual plasticity in adult and old mice. Neurobiol Aging 2020; 95:214-224. [DOI: 10.1016/j.neurobiolaging.2020.07.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 07/10/2020] [Accepted: 07/25/2020] [Indexed: 11/20/2022]
|
8
|
Hooks BM, Chen C. Circuitry Underlying Experience-Dependent Plasticity in the Mouse Visual System. Neuron 2020; 106:21-36. [PMID: 32272065 DOI: 10.1016/j.neuron.2020.01.031] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 01/13/2020] [Accepted: 01/23/2020] [Indexed: 12/15/2022]
Abstract
Since the discovery of ocular dominance plasticity, neuroscientists have understood that changes in visual experience during a discrete developmental time, the critical period, trigger robust changes in the visual cortex. State-of-the-art tools used to probe connectivity with cell-type-specific resolution have expanded the understanding of circuit changes underlying experience-dependent plasticity. Here, we review the visual circuitry of the mouse, describing projections from retina to thalamus, between thalamus and cortex, and within cortex. We discuss how visual circuit development leads to precise connectivity and identify synaptic loci, which can be altered by activity or experience. Plasticity extends to visual features beyond ocular dominance, involving subcortical and cortical regions, and connections between cortical inhibitory interneurons. Experience-dependent plasticity contributes to the alignment of networks spanning retina to thalamus to cortex. Disruption of this plasticity may underlie aberrant sensory processing in some neurodevelopmental disorders.
Collapse
Affiliation(s)
- Bryan M Hooks
- Department of Neurobiology, University of Pittsburgh School of Medicine, W1458 BSTWR, 203 Lothrop Street, Pittsburgh, PA 15213, USA.
| | - Chinfei Chen
- Department of Neurology, F.M. Kirby Neurobiology Center, Children's Hospital, Boston, 300 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
9
|
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
|
10
|
Shahidi SH, Kordi MR, Rajabi H, Malm C, Shah F, Quchan ASK. Exercise modulates the levels of growth inhibitor genes before and after multiple sclerosis. J Neuroimmunol 2020; 341:577172. [DOI: 10.1016/j.jneuroim.2020.577172] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 01/22/2020] [Accepted: 01/28/2020] [Indexed: 01/09/2023]
|
11
|
Nogo-A-targeting antibody promotes visual recovery and inhibits neuroinflammation after retinal injury. Cell Death Dis 2020; 11:101. [PMID: 32029703 PMCID: PMC7005317 DOI: 10.1038/s41419-020-2302-x] [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: 09/13/2019] [Revised: 01/21/2020] [Accepted: 01/23/2020] [Indexed: 12/11/2022]
Abstract
N-Methyl-D-aspartate (NMDA)-induced neuronal cell death is involved in a large spectrum of diseases affecting the brain and the retina such as Alzheimer’s disease and diabetic retinopathy. Associated neurological impairments may result from the inhibition of neuronal plasticity by Nogo-A. The objective of the current study was to determine the contribution of Nogo-A to NMDA excitotoxicity in the mouse retina. We observed that Nogo-A is upregulated in the mouse vitreous during NMDA-induced inflammation. Intraocular injection of a function-blocking antibody specific to Nogo-A (11C7) was carried out 2 days after NMDA-induced injury. This treatment significantly enhanced visual function recovery in injured animals. Strikingly, the expression of potent pro-inflammatory molecules was downregulated by 11C7, among which TNFα was the most durably decreased cytokine in microglia/macrophages. Additional analyses suggest that TNFα downregulation may stem from cofilin inactivation in microglia/macrophages. 11C7 also limited gliosis presumably via P.Stat3 downregulation. Diabetic retinopathy was associated with increased levels of Nogo-A in the eyes of donors. In summary, our results reveal that Nogo-A-targeting antibody can stimulate visual recovery after retinal injury and that Nogo-A is a potent modulator of excitotoxicity-induced neuroinflammation. These data may be used to design treatments against inflammatory eye diseases.
Collapse
|
12
|
Joly S, Dejda A, Rodriguez L, Sapieha P, Pernet V. Nogo-A inhibits vascular regeneration in ischemic retinopathy. Glia 2018; 66:2079-2093. [PMID: 30051920 DOI: 10.1002/glia.23462] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 05/10/2018] [Accepted: 05/10/2018] [Indexed: 01/20/2023]
Abstract
Nogo-A is a potent glial-derived inhibitor of axon growth in the injured CNS and acts as a negative regulator of developmental angiogenesis by inhibiting vascular endothelial cell migration. However, its function in pathological angiogenesis has never been studied after ischemic injury in the CNS. Using the mouse model of oxygen-induced retinopathy (OIR) which yields defined zones of retinal ischemia, our goal was to investigate the role of Nogo-A in vascular regeneration. We demonstrate a marked upregulation of the Nogo-A receptor sphingosine 1-phosphate receptor 2 in blood vessels following OIR, while Nogo-A is abundantly expressed in surrounding glial cells. Acute inhibition of Nogo-A with function-blocking antibody 11C7 significantly improved vascular regeneration and consequently prevented pathological pre-retinal angiogenesis. Ultimately, inhibition of Nogo-A led to restoration of retinal function as determined by electrophysiological response of retinal cells to light stimulation. Our data suggest that anti-Nogo-A antibody may protect neuronal cells from ischemic damage by accelerating blood vessel repair in the CNS. Targeting Nogo-A by immunotherapy may improve CNS perfusion after vascular injuries.
Collapse
Affiliation(s)
- Sandrine Joly
- CUO-Recherche, Centre de recherche du CHU de Québec and Département d'ophtalmologie, Faculté de médecine, Université Laval, Quebec, Quebec, Canada
| | - Agnieszka Dejda
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec, Canada
| | - Léa Rodriguez
- CUO-Recherche, Centre de recherche du CHU de Québec and Département d'ophtalmologie, Faculté de médecine, Université Laval, Quebec, Quebec, Canada
| | - Przemyslaw Sapieha
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec, Canada
| | - Vincent Pernet
- CUO-Recherche, Centre de recherche du CHU de Québec and Département d'ophtalmologie, Faculté de médecine, Université Laval, Quebec, Quebec, Canada
| |
Collapse
|
13
|
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
|
14
|
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
|
15
|
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
|
16
|
Restoring GM1 ganglioside expression ameliorates axonal outgrowth inhibition and cognitive impairments induced by blast traumatic brain injury. Sci Rep 2017; 7:41269. [PMID: 28112258 PMCID: PMC5255550 DOI: 10.1038/srep41269] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 12/19/2016] [Indexed: 01/01/2023] Open
Abstract
Blast induced traumatic brain injury (B-TBI) may cause various degrees of cognitive and behavioral disturbances but the exact brain pathophysiology involved is poorly understood. It was previously suggested that ganglioside alteration on the axon surface as well as axonal regenerating inhibitors (ARIs) such as myelin associated glycoprotein (MAG) were involved in axonal outgrowth inhibition (AOI), leading to brain damage. GM1 ganglioside content in the brain was significantly reduced while GD1 ganglioside was not affected. The axonal regeneration was also reduced as seen by the phosphorylated NF-H expression. Moreover, B-TBI induced a significant elevation in MAG expression in the brains of the injured mice. The blast injured mice exhibited a significant decline in spatial memory as seen by the Y-maze test. In addition, the injured mice showed pronounced damage to the visual memory (as evaluated by the Novel object recognition test). A single low dose of GM1 (2 mg/kg; IP), shortly after the injury, prevented both the cognitive and the cellular changes in the brains of the injured mice. These results enlighten part of the complicated mechanism that underlies the damage induced by B-TBI and may also suggest a potential new treatment strategy for brain injuries.
Collapse
|