1
|
Beryozkin A, Byrne LC. In Vivo Imaging of Rodent Retina in Retinal Disease. Methods Mol Biol 2025; 2848:151-167. [PMID: 39240522 DOI: 10.1007/978-1-0716-4087-6_10] [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] [Indexed: 09/07/2024]
Abstract
High-quality imaging of the retina is crucial to the diagnosis and monitoring of disease, as well as for evaluating the success of therapeutics in human patients and in preclinical animal models. Here, we describe the basic principles and methods for in vivo retinal imaging in rodents, including fundus imaging, fluorescein angiography, optical coherence tomography, fundus autofluorescence, and infrared imaging. After providing a concise overview of each method and detailing the retinal diseases and conditions that can be visualized through them, we will proceed to discuss the advantages and disadvantages of each approach. These protocols will facilitate the acquisition of optimal images for subsequent quantification and analysis. Additionally, a brief explanation will be given regarding the potential results and the clinical significance of the detected abnormalities.
Collapse
Affiliation(s)
- Avigail Beryozkin
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Leah C Byrne
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
2
|
Raspe R, Günther R, Uecker R, Rakhymzhan A, Paul F, Radbruch H, Niesner RA, Hauser AE. Multimodal Longitudinal Optical Imaging Reveals Optic Neuritis Preceding Retinal Pathology in Experimental Autoimmune Encephalomyelitis. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2025; 12:e200338. [PMID: 39661921 PMCID: PMC11637508 DOI: 10.1212/nxi.0000000000200338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 09/18/2024] [Indexed: 12/13/2024]
Abstract
BACKGROUND AND OBJECTIVES Inflammatory demyelinating diseases of the CNS, chief among them multiple sclerosis (MS), are a major cause of disability in young adults. Early manifestations of MS commonly involve visual dysfunction, which is often caused by optic neuritis and is accompanied by quantifiable structural changes of the anterior visual pathway. Retinal optical coherence tomography (OCT) has emerged as an important tool for clinical assessment of these structural alterations, but the underlying pathobiological mechanisms and temporal dynamics are yet poorly understood at a cellular level. METHODS Using the experimental autoimmune encephalomyelitis (EAE) model of MS in fluorescent reporter mouse strains for neuronal function and innate immune cells, we use a unique combination of retinal intravital 2-photon microscopy (2PM) and OCT. In this fashion, we elucidate the spatiotemporal interplay of functional and structural retinal changes over the course of 1 month after EAE induction, with histopathologic imaging validating main results. RESULTS While all mice display histologic signs of optic neuritis early after EAE induction and independently of motor symptom severity, retinal signs of neuronal stress and parenchymal immune activation spike well after clinical peak of disease, with no signs of lasting structural damage appearing within 1 month after EAE induction. Thus, local retinal endpoints appear to be functions of downstream axonal damage rather than of immediate immune activation directed at the retina. However, as early as 1 week after EAE induction, retinal 2PM can detect recruitment of perivascular immune cells towards the optic nerve (ON), providing the earliest sign of disease activity in otherwise clinically inconspicuous mice. DISCUSSION Our work identifies the recruitment of CX3CR1+ cells to the ON as an early sign of disease underlining the importance of combined structural and functional retinal imaging for the spatiotemporal characterization of neuroinflammatory and neurodegenerative processes. It further proposes retinal phagocyte orientation, morphology, and abundance as potential surrogate markers for neurodegenerative activity.
Collapse
Affiliation(s)
- Raphael Raspe
- From the Departments of Rheumatology and Clinical Immunology (R.R., A.E.H., R.U.) and Neuropathology (R.R., H.R.), Charité - Universitätsmedizin Berlin; Deutsches Rheuma-Forschungszentrum, a Leibniz Institute, Immune Dynamics (A.E.H., R.G.) and Biophysical Analytics (A.R., R.A.N.), Berlin; NeuroCure Clinical Research Center, Charité - Universitätsmedizin Berlin (F.P.), Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max-Delbrueck Center for Molecular Medicine (F.P.); and Dynamic and Functional in vivo Imaging, Freie Universität (R.A.N.) Berlin, Germany
| | - Robert Günther
- From the Departments of Rheumatology and Clinical Immunology (R.R., A.E.H., R.U.) and Neuropathology (R.R., H.R.), Charité - Universitätsmedizin Berlin; Deutsches Rheuma-Forschungszentrum, a Leibniz Institute, Immune Dynamics (A.E.H., R.G.) and Biophysical Analytics (A.R., R.A.N.), Berlin; NeuroCure Clinical Research Center, Charité - Universitätsmedizin Berlin (F.P.), Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max-Delbrueck Center for Molecular Medicine (F.P.); and Dynamic and Functional in vivo Imaging, Freie Universität (R.A.N.) Berlin, Germany
| | - Ralf Uecker
- From the Departments of Rheumatology and Clinical Immunology (R.R., A.E.H., R.U.) and Neuropathology (R.R., H.R.), Charité - Universitätsmedizin Berlin; Deutsches Rheuma-Forschungszentrum, a Leibniz Institute, Immune Dynamics (A.E.H., R.G.) and Biophysical Analytics (A.R., R.A.N.), Berlin; NeuroCure Clinical Research Center, Charité - Universitätsmedizin Berlin (F.P.), Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max-Delbrueck Center for Molecular Medicine (F.P.); and Dynamic and Functional in vivo Imaging, Freie Universität (R.A.N.) Berlin, Germany
| | - Asylkhan Rakhymzhan
- From the Departments of Rheumatology and Clinical Immunology (R.R., A.E.H., R.U.) and Neuropathology (R.R., H.R.), Charité - Universitätsmedizin Berlin; Deutsches Rheuma-Forschungszentrum, a Leibniz Institute, Immune Dynamics (A.E.H., R.G.) and Biophysical Analytics (A.R., R.A.N.), Berlin; NeuroCure Clinical Research Center, Charité - Universitätsmedizin Berlin (F.P.), Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max-Delbrueck Center for Molecular Medicine (F.P.); and Dynamic and Functional in vivo Imaging, Freie Universität (R.A.N.) Berlin, Germany
| | - Friedemann Paul
- From the Departments of Rheumatology and Clinical Immunology (R.R., A.E.H., R.U.) and Neuropathology (R.R., H.R.), Charité - Universitätsmedizin Berlin; Deutsches Rheuma-Forschungszentrum, a Leibniz Institute, Immune Dynamics (A.E.H., R.G.) and Biophysical Analytics (A.R., R.A.N.), Berlin; NeuroCure Clinical Research Center, Charité - Universitätsmedizin Berlin (F.P.), Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max-Delbrueck Center for Molecular Medicine (F.P.); and Dynamic and Functional in vivo Imaging, Freie Universität (R.A.N.) Berlin, Germany
| | - Helena Radbruch
- From the Departments of Rheumatology and Clinical Immunology (R.R., A.E.H., R.U.) and Neuropathology (R.R., H.R.), Charité - Universitätsmedizin Berlin; Deutsches Rheuma-Forschungszentrum, a Leibniz Institute, Immune Dynamics (A.E.H., R.G.) and Biophysical Analytics (A.R., R.A.N.), Berlin; NeuroCure Clinical Research Center, Charité - Universitätsmedizin Berlin (F.P.), Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max-Delbrueck Center for Molecular Medicine (F.P.); and Dynamic and Functional in vivo Imaging, Freie Universität (R.A.N.) Berlin, Germany
| | - Raluca Aura Niesner
- From the Departments of Rheumatology and Clinical Immunology (R.R., A.E.H., R.U.) and Neuropathology (R.R., H.R.), Charité - Universitätsmedizin Berlin; Deutsches Rheuma-Forschungszentrum, a Leibniz Institute, Immune Dynamics (A.E.H., R.G.) and Biophysical Analytics (A.R., R.A.N.), Berlin; NeuroCure Clinical Research Center, Charité - Universitätsmedizin Berlin (F.P.), Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max-Delbrueck Center for Molecular Medicine (F.P.); and Dynamic and Functional in vivo Imaging, Freie Universität (R.A.N.) Berlin, Germany
| | - Anja Erika Hauser
- From the Departments of Rheumatology and Clinical Immunology (R.R., A.E.H., R.U.) and Neuropathology (R.R., H.R.), Charité - Universitätsmedizin Berlin; Deutsches Rheuma-Forschungszentrum, a Leibniz Institute, Immune Dynamics (A.E.H., R.G.) and Biophysical Analytics (A.R., R.A.N.), Berlin; NeuroCure Clinical Research Center, Charité - Universitätsmedizin Berlin (F.P.), Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max-Delbrueck Center for Molecular Medicine (F.P.); and Dynamic and Functional in vivo Imaging, Freie Universität (R.A.N.) Berlin, Germany
| |
Collapse
|
3
|
Elwood BW, Godwin CR, Anders JJ, Kardon RH, Gramlich OW. Correlation of Visual System Biomarkers With Motor Deficits in Experimental Autoimmune Encephalomyelitis-Optic Neuritis. Transl Vis Sci Technol 2024; 13:1. [PMID: 39087931 PMCID: PMC11305423 DOI: 10.1167/tvst.13.8.1] [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: 02/06/2024] [Accepted: 06/11/2024] [Indexed: 08/02/2024] Open
Abstract
Purpose Experimental autoimmune encephalomyelitis (EAE) scoring, the most commonly used primary outcome metric for an in vivo model of multiple sclerosis (MS), is highly variable and subjective. Here we explored the use of visual biomarkers in EAE as more objective and clinically relevant primary outcomes. Methods Motor impairment in myelin oligodendrocyte glycoprotein-immunized C57BL/6J mice was quantified using a five-point EAE grading scale. Pattern electroretinography (pERG) and retinal ganglion cell/inner plexiform layer (RGC/IPL) complex thickness were measured 60 days after induction. Optic nerve histopathology was analyzed at endpoint. Results EAE mice displayed motor impairments ranging from mild to severe. Significant correlations were seen between pERG amplitude and last EAE score, mean EAE score, and cumulative EAE score. Optical coherence tomography (OCT) analysis demonstrated a significant correlation between thinning of the RGC/IPL complex and both EAE score and pERG amplitude. Optic nerve histopathology showed significant correlations between demyelination and cumulative EAE score, pERG amplitude, and RGC/IPL complex thickness, as well as between immune cell infiltration and cumulative EAE score, pERG amplitude, and RGC/IPL complex thickness in EAE mice. Conclusions Unlike EAE scoring, pERG and OCT show direct measurement of retinal structure and function. Therefore we conclude that visual outcomes are well suited as a direct assessment of optic nerve involvement in this EAE model of MS while also being indicative of motor impairment. Translational Relevance Standardizing directly translatable measurements as primary outcome parameters in the murine EAE model could lead to more rapid and relevant testing of new therapeutic approaches for mitigating MS.
Collapse
Affiliation(s)
- Benjamin W. Elwood
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA, USA
- Center for the Prevention and Treatment of Visual Loss, Iowa City Veterans Affairs Health Care System, Iowa City, IA, USA
| | - Cheyanne R. Godwin
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA, USA
- Center for the Prevention and Treatment of Visual Loss, Iowa City Veterans Affairs Health Care System, Iowa City, IA, USA
| | - Jeffrey J. Anders
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA, USA
- Center for the Prevention and Treatment of Visual Loss, Iowa City Veterans Affairs Health Care System, Iowa City, IA, USA
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA
| | - Randy H. Kardon
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA, USA
- Center for the Prevention and Treatment of Visual Loss, Iowa City Veterans Affairs Health Care System, Iowa City, IA, USA
| | - Oliver W. Gramlich
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA, USA
- Center for the Prevention and Treatment of Visual Loss, Iowa City Veterans Affairs Health Care System, Iowa City, IA, USA
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
4
|
Cordano C, Werneburg S, Abdelhak A, Bennett DJ, Beaudry-Richard A, Duncan GJ, Oertel FC, Boscardin WJ, Yiu HH, Jabassini N, Merritt L, Nocera S, Sin JH, Samana IP, Condor Montes SY, Ananth K, Bischof A, Nourbakhsh B, Hauser SL, Cree BAC, Emery B, Schafer DP, Chan JR, Green AJ. Synaptic injury in the inner plexiform layer of the retina is associated with progression in multiple sclerosis. Cell Rep Med 2024; 5:101490. [PMID: 38574736 PMCID: PMC11031420 DOI: 10.1016/j.xcrm.2024.101490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 02/01/2024] [Accepted: 03/12/2024] [Indexed: 04/06/2024]
Abstract
While neurodegeneration underlies the pathological basis for permanent disability in multiple sclerosis (MS), predictive biomarkers for progression are lacking. Using an animal model of chronic MS, we find that synaptic injury precedes neuronal loss and identify thinning of the inner plexiform layer (IPL) as an early feature of inflammatory demyelination-prior to symptom onset. As neuronal domains are anatomically segregated in the retina and can be monitored longitudinally, we hypothesize that thinning of the IPL could represent a biomarker for progression in MS. Leveraging our dataset with over 800 participants enrolled for more than 12 years, we find that IPL atrophy directly precedes progression and propose that synaptic loss is predictive of functional decline. Using a blood proteome-wide analysis, we demonstrate a strong correlation between demyelination, glial activation, and synapse loss independent of neuroaxonal injury. In summary, monitoring synaptic injury is a biologically relevant approach that reflects a potential driver of progression.
Collapse
Affiliation(s)
- Christian Cordano
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA; Department of Neurology, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Sebastian Werneburg
- Department of Neurobiology, Brudnik Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, MA, USA; Department of Ophthalmology & Visual Sciences, Michigan Neuroscience Institute, University of Michigan - Michigan Medicine, Ann Arbor, MI, USA
| | - Ahmed Abdelhak
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Daniel J Bennett
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Alexandra Beaudry-Richard
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Greg J Duncan
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health & Science University, Portland, OR, USA
| | - Frederike C Oertel
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - W John Boscardin
- Department of Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | - Hao H Yiu
- Department of Biology, University of Maryland, College Park, MD, USA
| | - Nora Jabassini
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Lauren Merritt
- Department of Neurobiology, Brudnik Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Sonia Nocera
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Jung H Sin
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Isaac P Samana
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Shivany Y Condor Montes
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Kirtana Ananth
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Antje Bischof
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Bardia Nourbakhsh
- Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Stephen L Hauser
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Bruce A C Cree
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Ben Emery
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health & Science University, Portland, OR, USA
| | - Dorothy P Schafer
- Department of Neurobiology, Brudnik Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Jonah R Chan
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA.
| | - Ari J Green
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
5
|
Oertel FC, Hastermann M, Paul F. Delimiting MOGAD as a disease entity using translational imaging. Front Neurol 2023; 14:1216477. [PMID: 38333186 PMCID: PMC10851159 DOI: 10.3389/fneur.2023.1216477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 08/23/2023] [Indexed: 02/10/2024] Open
Abstract
The first formal consensus diagnostic criteria for myelin oligodendrocyte glycoprotein antibody-associated disease (MOGAD) were recently proposed. Yet, the distinction of MOGAD-defining characteristics from characteristics of its important differential diagnoses such as multiple sclerosis (MS) and aquaporin-4 antibody seropositive neuromyelitis optica spectrum disorder (NMOSD) is still obstructed. In preclinical research, MOG antibody-based animal models were used for decades to derive knowledge about MS. In clinical research, people with MOGAD have been combined into cohorts with other diagnoses. Thus, it remains unclear to which extent the generated knowledge is specifically applicable to MOGAD. Translational research can contribute to identifying MOGAD characteristic features by establishing imaging methods and outcome parameters on proven pathophysiological grounds. This article reviews suitable animal models for translational MOGAD research and the current state and prospect of translational imaging in MOGAD.
Collapse
Affiliation(s)
- Frederike Cosima Oertel
- Experimental and Clinical Research Center, Max-Delbrück-Centrum für Molekulare Medizin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Neuroscience Clinical Research Center, Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Department of Neurology, Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Maria Hastermann
- Experimental and Clinical Research Center, Max-Delbrück-Centrum für Molekulare Medizin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Neuroscience Clinical Research Center, Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Friedemann Paul
- Experimental and Clinical Research Center, Max-Delbrück-Centrum für Molekulare Medizin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Neuroscience Clinical Research Center, Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Department of Neurology, Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
6
|
Anders JJ, Elwood BW, Kardon RH, Gramlich OW. Acriflavine, a HIF-1 inhibitor, preserves vision in an experimental autoimmune encephalomyelitis model of optic neuritis. Front Immunol 2023; 14:1271118. [PMID: 37942317 PMCID: PMC10628762 DOI: 10.3389/fimmu.2023.1271118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 09/25/2023] [Indexed: 11/10/2023] Open
Abstract
Introduction Optic neuritis (ON) is often an early sign of multiple sclerosis (MS), and recent studies show a link between HIF-1 pathway activation and inflammation. This study aimed to determine if inhibition of the HIF-1 pathway using the HIF-1a antagonist acriflavine (ACF) can reduce clinical progression and rescue the ocular phenotype in an experimental autoimmune encephalomyelitis (EAE) ON model. Methods EAE-related ON was induced in 60 female C57BL/6J mice by immunization with MOG33-55, and 20 EAE mice received daily systemic injections of ACF at 5 mg/kg. Changes in the visual function and structure of ACF-treated EAE mice were compared to those of placebo-injected EAE mice and naïve control mice. Results ACF treatment improved motor-sensory impairment along with preserving visual acuity and optic nerve function. Analysis of retinal ganglion cell complex alsoshowed preserved thickness correlating with increased survival of retinal ganglion cells and their axons. Optic nerve cell infiltration and magnitude of demyelination were decreased in ACF-treated EAE mice. Subsequent in vitro studies revealed improvements not only attributed to the inhibition of HIF-1 butalso to previously unappreciated interaction with the eIF2a/ATF4 axis in the unfolded protein response pathway. Discussion This study suggests that ACF treatment is effective in an animal model of MS via its pleiotropic effects on the inhibition of HIF-1 and UPR signaling, and it may be a viable approach to promote rehabilitation in MS.
Collapse
Affiliation(s)
- Jeffrey J. Anders
- Department of Ophthalmology and Visual Science, The University of Iowa, Iowa City, IA, United States
- Center for the Prevention and Treatment of Visual Loss, Iowa City Veterans Affairs (VA) Health Care System, Iowa City, IA, United States
| | - Benjamin W. Elwood
- Department of Ophthalmology and Visual Science, The University of Iowa, Iowa City, IA, United States
- Center for the Prevention and Treatment of Visual Loss, Iowa City Veterans Affairs (VA) Health Care System, Iowa City, IA, United States
| | - Randy H. Kardon
- Department of Ophthalmology and Visual Science, The University of Iowa, Iowa City, IA, United States
- Center for the Prevention and Treatment of Visual Loss, Iowa City Veterans Affairs (VA) Health Care System, Iowa City, IA, United States
| | - Oliver W. Gramlich
- Department of Ophthalmology and Visual Science, The University of Iowa, Iowa City, IA, United States
- Center for the Prevention and Treatment of Visual Loss, Iowa City Veterans Affairs (VA) Health Care System, Iowa City, IA, United States
- Department of Neuroscience and Pharmacology, The University of Iowa, Iowa City, IA, United States
| |
Collapse
|
7
|
Remlinger J, Bagnoud M, Meli I, Massy M, Hoepner R, Linington C, Chan A, Bennett JL, Enzmann V, Salmen A. Modeling MOG Antibody-Associated Disorder and Neuromyelitis Optica Spectrum Disorder in Animal Models: Visual System Manifestations. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2023; 10:e200141. [PMID: 37429715 PMCID: PMC10691219 DOI: 10.1212/nxi.0000000000200141] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 05/15/2023] [Indexed: 07/12/2023]
Abstract
BACKGROUND AND OBJECTIVES Mechanisms of visual impairment in aquaporin 4 antibody (AQP4-IgG) seropositive neuromyelitis optica spectrum disorder (NMOSD) and myelin oligodendrocyte glycoprotein antibody (MOG-IgG)-associated disorder (MOGAD) are incompletely understood. The respective impact of optic nerve demyelination and primary and secondary retinal neurodegeneration are yet to be investigated in animal models. METHODS Active MOG35-55 experimental autoimmune encephalomyelitis (EAE) was induced in C57BL/6Jrj mice, and monoclonal MOG-IgG (8-18C5, murine), recombinant AQP4-IgG (rAb-53, human), or isotype-matched control IgG (Iso-IgG, human) was administered 10 days postimmunization. Mobility impairment was scored daily. Visual acuity by optomotor reflex and ganglion cell complex thickness (GCC, 3 innermost retinal layers) by optical coherence tomography (OCT) were longitudinally assessed. Histopathology of optic nerve and retina was investigated during presymptomatic, acute, and chronic disease phases for immune cells, demyelination, complement deposition, natural killer (NK) cell, AQP4, and astrocyte involvement, retinal ganglion cells (RGCs), and Müller cell activation. Groups were compared by nonparametric tests with a p value <0.05 indicating statistical significance. RESULTS Visual acuity decreased from baseline to chronic phase in MOG-IgG (mean ± standard error of the mean: 0.54 ± 0.01 to 0.46 ± 0.02 cycles/degree, p < 0.05) and AQP4-IgG EAE (0.54 ± 0.01 to 0.43 ± 0.02, cycles/degree, p < 0.05). Immune cell infiltration of optic nerves started in presymptomatic AQP4-IgG, but not in MOG-IgG EAE (5.85 ± 2.26 vs 0.13 ± 0.10 macrophages/region of interest [ROI] and 1.88 ± 0.63 vs 0.15 ± 0.06 T cells/ROI, both p < 0.05). Few NK cells, no complement deposition, and stable glial fibrillary acid protein and AQP4 fluorescence intensity characterized all EAE optic nerves. Lower GCC thickness (Spearman correlation coefficient r = -0.44, p < 0.05) and RGC counts (r = -0.47, p < 0.05) correlated with higher mobility impairment. RGCs decreased from presymptomatic to chronic disease phase in MOG-IgG (1,705 ± 51 vs 1,412 ± 45, p < 0.05) and AQP4-IgG EAE (1,758 ± 14 vs 1,526 ± 48, p < 0.01). Müller cell activation was not observed in either model. DISCUSSION In a multimodal longitudinal characterization of visual outcome in animal models of MOGAD and NMOSD, differential retinal injury and optic nerve involvement were not conclusively clarified. Yet optic nerve inflammation was earlier in AQP4-IgG-associated pathophysiology. Retinal atrophy determined by GCC thickness (OCT) and RGC counts correlating with mobility impairment in the chronic phase of MOG-IgG and AQP4-IgG EAE may serve as a generalizable marker of neurodegeneration.
Collapse
Affiliation(s)
- Jana Remlinger
- From the Department of Neurology (J.R., M.B., I.M., M.M., R.H., A.C., A.S.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences (J.R., M.M.), University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow, UK; Departments of Neurology and Ophthalmology (J.L.B.), Programs in Neuroscience and Immunology, University of Colorado Anschutz Medical Campus, Aurora; and Department of Ophthalmology (V.E.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland
| | - Maud Bagnoud
- From the Department of Neurology (J.R., M.B., I.M., M.M., R.H., A.C., A.S.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences (J.R., M.M.), University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow, UK; Departments of Neurology and Ophthalmology (J.L.B.), Programs in Neuroscience and Immunology, University of Colorado Anschutz Medical Campus, Aurora; and Department of Ophthalmology (V.E.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland
| | - Ivo Meli
- From the Department of Neurology (J.R., M.B., I.M., M.M., R.H., A.C., A.S.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences (J.R., M.M.), University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow, UK; Departments of Neurology and Ophthalmology (J.L.B.), Programs in Neuroscience and Immunology, University of Colorado Anschutz Medical Campus, Aurora; and Department of Ophthalmology (V.E.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland
| | - Marine Massy
- From the Department of Neurology (J.R., M.B., I.M., M.M., R.H., A.C., A.S.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences (J.R., M.M.), University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow, UK; Departments of Neurology and Ophthalmology (J.L.B.), Programs in Neuroscience and Immunology, University of Colorado Anschutz Medical Campus, Aurora; and Department of Ophthalmology (V.E.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland
| | - Robert Hoepner
- From the Department of Neurology (J.R., M.B., I.M., M.M., R.H., A.C., A.S.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences (J.R., M.M.), University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow, UK; Departments of Neurology and Ophthalmology (J.L.B.), Programs in Neuroscience and Immunology, University of Colorado Anschutz Medical Campus, Aurora; and Department of Ophthalmology (V.E.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland
| | - Christopher Linington
- From the Department of Neurology (J.R., M.B., I.M., M.M., R.H., A.C., A.S.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences (J.R., M.M.), University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow, UK; Departments of Neurology and Ophthalmology (J.L.B.), Programs in Neuroscience and Immunology, University of Colorado Anschutz Medical Campus, Aurora; and Department of Ophthalmology (V.E.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland
| | - Andrew Chan
- From the Department of Neurology (J.R., M.B., I.M., M.M., R.H., A.C., A.S.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences (J.R., M.M.), University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow, UK; Departments of Neurology and Ophthalmology (J.L.B.), Programs in Neuroscience and Immunology, University of Colorado Anschutz Medical Campus, Aurora; and Department of Ophthalmology (V.E.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland
| | - Jeffrey L Bennett
- From the Department of Neurology (J.R., M.B., I.M., M.M., R.H., A.C., A.S.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences (J.R., M.M.), University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow, UK; Departments of Neurology and Ophthalmology (J.L.B.), Programs in Neuroscience and Immunology, University of Colorado Anschutz Medical Campus, Aurora; and Department of Ophthalmology (V.E.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland
| | - Volker Enzmann
- From the Department of Neurology (J.R., M.B., I.M., M.M., R.H., A.C., A.S.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences (J.R., M.M.), University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow, UK; Departments of Neurology and Ophthalmology (J.L.B.), Programs in Neuroscience and Immunology, University of Colorado Anschutz Medical Campus, Aurora; and Department of Ophthalmology (V.E.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland
| | - Anke Salmen
- From the Department of Neurology (J.R., M.B., I.M., M.M., R.H., A.C., A.S.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences (J.R., M.M.), University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow, UK; Departments of Neurology and Ophthalmology (J.L.B.), Programs in Neuroscience and Immunology, University of Colorado Anschutz Medical Campus, Aurora; and Department of Ophthalmology (V.E.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland.
| |
Collapse
|
8
|
Kapell H, Fazio L, Dyckow J, Schwarz S, Cruz-Herranz A, Mayer C, Campos J, D’Este E, Möbius W, Cordano C, Pröbstel AK, Gharagozloo M, Zulji A, Narayanan Naik V, Delank A, Cerina M, Müntefering T, Lerma-Martin C, Sonner JK, Sin JH, Disse P, Rychlik N, Sabeur K, Chavali M, Srivastava R, Heidenreich M, Fitzgerald KC, Seebohm G, Stadelmann C, Hemmer B, Platten M, Jentsch TJ, Engelhardt M, Budde T, Nave KA, Calabresi PA, Friese MA, Green AJ, Acuna C, Rowitch DH, Meuth SG, Schirmer L. Neuron-oligodendrocyte potassium shuttling at nodes of Ranvier protects against inflammatory demyelination. J Clin Invest 2023; 133:e164223. [PMID: 36719741 PMCID: PMC10065072 DOI: 10.1172/jci164223] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 01/27/2023] [Indexed: 02/01/2023] Open
Abstract
Multiple sclerosis (MS) is a progressive inflammatory demyelinating disease of the CNS. Increasing evidence suggests that vulnerable neurons in MS exhibit fatal metabolic exhaustion over time, a phenomenon hypothesized to be caused by chronic hyperexcitability. Axonal Kv7 (outward-rectifying) and oligodendroglial Kir4.1 (inward-rectifying) potassium channels have important roles in regulating neuronal excitability at and around the nodes of Ranvier. Here, we studied the spatial and functional relationship between neuronal Kv7 and oligodendroglial Kir4.1 channels and assessed the transcriptional and functional signatures of cortical and retinal projection neurons under physiological and inflammatory demyelinating conditions. We found that both channels became dysregulated in MS and experimental autoimmune encephalomyelitis (EAE), with Kir4.1 channels being chronically downregulated and Kv7 channel subunits being transiently upregulated during inflammatory demyelination. Further, we observed that pharmacological Kv7 channel opening with retigabine reduced neuronal hyperexcitability in human and EAE neurons, improved clinical EAE signs, and rescued neuronal pathology in oligodendrocyte-Kir4.1-deficient (OL-Kir4.1-deficient) mice. In summary, our findings indicate that neuron-OL compensatory interactions promoted resilience through Kv7 and Kir4.1 channels and identify pharmacological activation of nodal Kv7 channels as a neuroprotective strategy against inflammatory demyelination.
Collapse
Affiliation(s)
- Hannah Kapell
- Department of Neurology, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Luca Fazio
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster (UKM), Münster, Germany
- Department of Neurology, University of Düsseldorf, Dusseldorf, Germany
| | - Julia Dyckow
- Department of Neurology, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Sophia Schwarz
- Department of Neurology, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Andrés Cruz-Herranz
- Weill Institute for Neurosciences, Department of Neurology, UCSF, San Francisco, California, USA
| | - Christina Mayer
- Institute of Neuroimmunology and Multiple Sclerosis, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Joaquin Campos
- Chica and Heinz Schaller Research Group, Institute of Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Elisa D’Este
- Optical Microscopy Facility, Max Planck Institute for Medical Research, Heidelberg, Germany
| | - Wiebke Möbius
- Electron Microscopy Core Unit, Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
- Cluster of Excellence, “Multiscale Bioimaging: from Molecular Machines to Network of Excitable Cells” (MBExC), University of Göttingen, Göttingen, Germany
| | - Christian Cordano
- Weill Institute for Neurosciences, Department of Neurology, UCSF, San Francisco, California, USA
| | - Anne-Katrin Pröbstel
- Weill Institute for Neurosciences, Department of Neurology, UCSF, San Francisco, California, USA
- Neurologic Clinic and Policlinic and Research Center for Clinical Neuroimmunology and Neuroscience Basel, Departments of Medicine, Biomedicine, and Clinical Research, University Hospital of Basel, University of Basel, Basel, Switzerland
| | - Marjan Gharagozloo
- Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Amel Zulji
- Department of Neurology, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Venu Narayanan Naik
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster (UKM), Münster, Germany
| | - Anna Delank
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster (UKM), Münster, Germany
| | - Manuela Cerina
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster (UKM), Münster, Germany
| | | | - Celia Lerma-Martin
- Department of Neurology, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Jana K. Sonner
- Chica and Heinz Schaller Research Group, Institute of Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Jung Hyung Sin
- Weill Institute for Neurosciences, Department of Neurology, UCSF, San Francisco, California, USA
| | - Paul Disse
- Institute for Genetics of Heart Diseases (IfGH), Cellular Electrophysiology and Molecular Biology, UKM, Münster, Germany
- University of Münster, Chembion, Münster, Germany
| | - Nicole Rychlik
- University of Münster, Chembion, Münster, Germany
- Institute of Physiology I, University of Münster, Münster, Germany
| | - Khalida Sabeur
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research and
- Department of Pediatrics, UCSF, San Francisco, California, USA
| | - Manideep Chavali
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research and
- Department of Pediatrics, UCSF, San Francisco, California, USA
| | - Rajneesh Srivastava
- Department of Neurology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Matthias Heidenreich
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP) and Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany
| | - Kathryn C. Fitzgerald
- Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Guiscard Seebohm
- Institute for Genetics of Heart Diseases (IfGH), Cellular Electrophysiology and Molecular Biology, UKM, Münster, Germany
| | - Christine Stadelmann
- Electron Microscopy Core Unit, Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
- Institute of Neuropathology, University Medical Center, Göttingen, Germany
| | - Bernhard Hemmer
- Department of Neurology, School of Medicine, Technical University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology, Munich, Germany
| | - Michael Platten
- Department of Neurology, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- DKTK Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), INF 280, Heidelberg, Germany
- Interdisciplinary Center for Neurosciences (IZN) and
- Mannheim Center for Translational Neuroscience and Institute for Innate Immunoscience, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Thomas J. Jentsch
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP) and Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany
- Neurocure Cluster of Excellence, Charité University Medicine Berlin, Berlin, Germany
| | - Maren Engelhardt
- Mannheim Center for Translational Neuroscience and Institute for Innate Immunoscience, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- Institute of Neuroanatomy, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- Institute of Anatomy and Cell Biology, Johannes Kepler University Linz, Linz, Austria
| | - Thomas Budde
- Institute of Physiology I, University of Münster, Münster, Germany
| | - Klaus-Armin Nave
- Electron Microscopy Core Unit, Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Peter A. Calabresi
- Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Manuel A. Friese
- Institute of Neuroimmunology and Multiple Sclerosis, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Ari J. Green
- Weill Institute for Neurosciences, Department of Neurology, UCSF, San Francisco, California, USA
- Department of Ophthalmology, UCSF, San Francisco, California, USA
| | - Claudio Acuna
- Chica and Heinz Schaller Research Group, Institute of Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - David H. Rowitch
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research and
- Department of Pediatrics, UCSF, San Francisco, California, USA
- Wellcome Trust–Medical Research Council Stem Cell Institute and
- Department of Paediatrics, University of Cambridge, Cambridge, United Kingdom
- Department of Neurosurgery, UCSF, San Francisco, California, USA
| | - Sven G. Meuth
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster (UKM), Münster, Germany
- Department of Neurology, University of Düsseldorf, Dusseldorf, Germany
| | - Lucas Schirmer
- Department of Neurology, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- Interdisciplinary Center for Neurosciences (IZN) and
- Mannheim Center for Translational Neuroscience and Institute for Innate Immunoscience, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
9
|
Mey GM, DeSilva TM. Utility of the visual system to monitor neurodegeneration in multiple sclerosis. Front Mol Neurosci 2023; 16:1125115. [PMID: 37063369 PMCID: PMC10090562 DOI: 10.3389/fnmol.2023.1125115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 03/14/2023] [Indexed: 03/31/2023] Open
Abstract
Neurodegeneration occurs early in the multiple sclerosis (MS) disease course and is an important driver of permanent disability. Current immunomodulatory therapies do not directly target neuronal health; thus, there is a critical need to develop neuroprotective strategies in MS. Outcome measures in clinical trials primarily evaluate disease activity and clinical disability scores rather than measures of neurodegeneration. The visual system provides a noninvasive correlate of brain atrophy and neuronal function through structural and functional exams. Furthermore, optic nerve axons and their respective neuronal cell bodies in the retina, in addition to their synaptic input to the thalamus, provide a distinct anatomy to investigate neurodegenerative processes. This review discusses the utility of the visual system as an early output measure of neurodegeneration in MS as well as an important platform to evaluate neuroprotective strategies in preclinical models.
Collapse
Affiliation(s)
| | - Tara M. DeSilva
- Department of Neurosciences, Cleveland Clinic, Cleveland, OH, United States
| |
Collapse
|
10
|
Zhi JJ, Wu SL, Wu HQ, Ran Q, Gao X, Chen JF, Gu XM, Li T, Wang F, Xiao L, Ye J, Mei F. Insufficient Oligodendrocyte Turnover in Optic Nerve Contributes to Age-Related Axon Loss and Visual Deficits. J Neurosci 2023; 43:1859-1870. [PMID: 36725322 PMCID: PMC10027114 DOI: 10.1523/jneurosci.2130-22.2023] [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: 11/16/2022] [Revised: 01/16/2023] [Accepted: 01/27/2023] [Indexed: 02/03/2023] Open
Abstract
Age-related decline in visual functions is a prevalent health problem among elderly people, and no effective therapies are available up-to-date. Axon degeneration and myelin loss in optic nerves (ONs) are age-dependent and become evident in middle-aged (13-18 months) and old (20-22 months) mice of either sex compared with adult mice (3-8 months), accompanied by functional deficits. Oligodendrocyte (OL) turnover is actively going on in adult ONs. However, the longitudinal change and functional significance of OL turnover in aging ONs remain largely unknown. Here, using cell-lineage labeling and tracing, we reported that oligodendrogenesis displayed an age-dependent decrease in aging ONs. To understand whether active OL turnover is required for maintaining axons and visual function, we conditionally deleted the transcription factor Olig2 in the oligodendrocyte precursor cells of young mice. Genetically dampening OL turnover by Olig2 ablation resulted in accelerated axon loss and retinal degeneration, and subsequently impaired ON signal transmission, suggesting that OL turnover is an important mechanism to sustain axon survival and visual function. To test whether enhancing oligodendrogenesis can prevent age-related visual deficits, 12-month-old mice were treated with clemastine, a pro-myelination drug, or induced deletion of the muscarinic receptor 1 in oligodendrocyte precursor cells. The clemastine treatment or muscarinic receptor 1 deletion significantly increased new OL generation in the aged ONs and consequently preserved visual function and retinal integrity. Together, our data indicate that dynamic OL turnover in ONs is required for axon survival and visual function, and enhancing new OL generation represents a potential approach to reversing age-related declines of visual function.SIGNIFICANCE STATEMENT Oligodendrocyte (OL) turnover has been reported in adult optic nerves (ONs), but the longitudinal change and functional significance of OL turnover during aging remain largely unknown. Using cell-lineage tracing and oligodendroglia-specific manipulation, this study reported that OL generation was active in adult ONs and the efficiency decreased in an age-dependent manner. Genetically dampening OL generation by Olig2 ablation resulted in significant axon loss and retinal degeneration, along with delayed visual signal transmission. Conversely, pro-myelination approaches significantly increased new myelin generation in aging ONs, and consequently preserved retinal integrity and visual function. Our findings indicate that promoting OL generation might be a promising strategy to preserve visual function from age-related decline.
Collapse
Affiliation(s)
- Jun-Jie Zhi
- Department of Ophthalmology and Institute of Surgery Research, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Shuang-Ling Wu
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- School of Medicine, Chongqing University, Chongqing, 400030, China
| | - Hao-Qian Wu
- Department of Ophthalmology and Institute of Surgery Research, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Qi Ran
- Department of Ophthalmology and Institute of Surgery Research, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Xing Gao
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jing-Fei Chen
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Xing-Mei Gu
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Department of Medical English Teaching and Research, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Tao Li
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Fei Wang
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Lan Xiao
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jian Ye
- Department of Ophthalmology and Institute of Surgery Research, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
| | - Feng Mei
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- School of Medicine, Chongqing University, Chongqing, 400030, China
| |
Collapse
|
11
|
3-Dimensional Immunostaining and Automated Deep-Learning Based Analysis of Nerve Degeneration. Int J Mol Sci 2022; 23:ijms232314811. [PMID: 36499143 PMCID: PMC9739543 DOI: 10.3390/ijms232314811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/21/2022] [Accepted: 11/24/2022] [Indexed: 12/03/2022] Open
Abstract
Multiple sclerosis (MS) is an autoimmune and neurodegenerative disease driven by inflammation and demyelination in the brain, spinal cord, and optic nerve. Optic neuritis, characterized by inflammation and demyelination of the optic nerve, is a symptom in many patients with MS. The optic nerve is the highway for visual information transmitted from the retina to the brain. It contains axons from the retinal ganglion cells (RGCs) that reside in the retina, myelin forming oligodendrocytes and resident microglia and astrocytes. Inflammation, demyelination, and axonal degeneration are also present in the optic nerve of mice subjected to experimental autoimmune encephalomyelitis (EAE), a preclinical mouse model of MS. Monitoring the optic nerve in EAE is a useful strategy to study the presentation and progression of pathology in the visual system; however, current approaches have relied on sectioning, staining and manual quantification. Further, information regarding the spatial load of lesions and inflammation is dependent on the area of sectioning. To better characterize cellular pathology in the EAE model, we employed a tissue clearing and 3D immunolabelling and imaging protocol to observe patterns of immune cell infiltration and activation throughout the optic nerve. Increased density of TOPRO staining for nuclei captured immune cell infiltration and Iba1 immunostaining was employed to monitor microglia and macrophages. Axonal degeneration was monitored by neurofilament immunolabelling to reveal axonal swellings throughout the optic nerve. In parallel, we developed a convolutional neural network with a UNet architecture (CNN-UNet) called BlebNet for automated identification and quantification of axonal swellings in whole mount optic nerves. Together this constitutes a toolkit for 3-dimensional immunostaining to monitor general optic nerve pathology and fast automated quantification of axonal defects that could also be adapted to monitor axonal degeneration and inflammation in other neurodegenerative disease models.
Collapse
|
12
|
Mey GM, Evonuk KS, Chappell MK, Wolfe LM, Singh R, Batoki JC, Yu M, Peachey NS, Anand-Apte B, Bermel R, Ontaneda D, Nakamura K, Mahajan KR, DeSilva TM. Visual imaging as a predictor of neurodegeneration in experimental autoimmune demyelination and multiple sclerosis. Acta Neuropathol Commun 2022; 10:87. [PMID: 35706005 PMCID: PMC9199245 DOI: 10.1186/s40478-022-01391-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 05/28/2022] [Indexed: 11/10/2022] Open
Abstract
Thalamic volume is associated with clinical disability in multiple sclerosis (MS) and is vulnerable to secondary neurodegeneration due to its extensive connectivity throughout the central nervous system (CNS). Using a model of autoimmune demyelination that exhibits CNS-infiltrating immune cells in both spinal cord white matter and optic nerve, we sought to evaluate neurodegenerative changes due to lesions affecting the spino- and retino-thalamic pathways. We found comparable axonal loss in spinal cord white matter and optic nerve during the acute phase of disease consistent with synaptic loss, but not neuronal cell body loss in the thalamic nuclei that receive input from these discrete pathways. Loss of spinal cord neurons or retinal ganglion cells retrograde to their respective axons was not observed until the chronic phase of disease, where optical coherence tomography (OCT) documented reduced inner retinal thickness. In patients with relapsing-remitting MS without a history of optic neuritis, OCT measures of inner retinal volume correlated with retino-thalamic (lateral geniculate nucleus) and spino-thalamic (ventral posterior nucleus) volume as well as neuroperformance measures. These data suggest retinal imaging may serve as an important noninvasive predictor of neurodegeneration in MS.
Collapse
Affiliation(s)
- Gabrielle M Mey
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic Foundation, and Case Western Reserve University, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | - Kirsten S Evonuk
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic Foundation, and Case Western Reserve University, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
- Hooke Laboratories, Inc., Lawrence, MA, USA
| | - McKenzie K Chappell
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic Foundation, and Case Western Reserve University, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | - Laura M Wolfe
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic Foundation, and Case Western Reserve University, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | - Rupesh Singh
- Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Julia C Batoki
- Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Minzhong Yu
- Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
- Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Neal S Peachey
- Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
- Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
- Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| | - Bela Anand-Apte
- Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
- Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Robert Bermel
- Mellen Center for MS Treatment and Research, Neurological Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Daniel Ontaneda
- Mellen Center for MS Treatment and Research, Neurological Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Kunio Nakamura
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Kedar R Mahajan
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic Foundation, and Case Western Reserve University, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
- Mellen Center for MS Treatment and Research, Neurological Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Tara M DeSilva
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic Foundation, and Case Western Reserve University, 9500 Euclid Avenue, Cleveland, OH, 44195, USA.
| |
Collapse
|
13
|
Cordano C, Sin JH, Timmons G, Yiu HH, Stebbins K, Guglielmetti C, Cruz-Herranz A, Xin W, Lorrain D, Chan JR, Green AJ. Validating visual evoked potentials as a preclinical, quantitative biomarker for remyelination efficacy. Brain 2022; 145:3943-3952. [PMID: 35678509 DOI: 10.1093/brain/awac207] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 05/19/2022] [Accepted: 05/30/2022] [Indexed: 11/14/2022] Open
Abstract
Many biomarkers in clinical neuroscience lack pathological certification. This issue is potentially a significant contributor to the limited success of neuroprotective and neurorestorative therapies for human neurological disease - and is evident even in areas with therapeutic promise such as myelin repair. Despite the identification of promising remyelinating candidates, biologically validated methods to demonstrate therapeutic efficacy or provide robust preclinical evidence of remyelination in the central nervous system are lacking. Therapies with potential to remyelinate the central nervous system constitute one of the most promising and highly anticipated therapeutic developments in the pipeline to treat multiple sclerosis and other demyelinating diseases. The optic nerve has been proposed as an informative pathway to monitor remyelination in animals and human subjects. Recent clinical trials using visual evoked potential (VEP) have had promising results, but without unequivocal evidence about the cellular and molecular basis for signal changes on VEP, the interpretation of these trials is constrained. The VEP was originally developed and utilized in the clinic as a diagnostic tool but its use as a quantitative method for assessing therapeutic response requires certification of its biological specificity. Here, using the tools of experimental pathology we demonstrate that quantitative measurements of myelination using both histopathological measures of nodal structure and ultrastructural assessments correspond to VEP latency in both inflammatory and chemical models of demyelination. VEP latency improves after treatment with a tool remyelinating compound (clemastine), mirroring both quantitative and qualitative myelin assessment. Furthermore, clemastine does not improve VEP latency following demyelinating injury when administered to a transgenic animal incapable of forming new myelin. Therefore, using the capacity for therapeutic enhancement and biological loss of function we demonstrate conclusively that VEP measures myelin status and is thereby a validated tool for preclinical verification of remyelination.
Collapse
Affiliation(s)
- Christian Cordano
- Department of Neurology, Weill Institute for Neuroscience, University of California, San Francisco; San Francisco, CA 94158, USA
| | - Jung H Sin
- Department of Neurology, Weill Institute for Neuroscience, University of California, San Francisco; San Francisco, CA 94158, USA
| | - Garrett Timmons
- Department of Neurology, Weill Institute for Neuroscience, University of California, San Francisco; San Francisco, CA 94158, USA
| | - Hao H Yiu
- Department of Neurology, Weill Institute for Neuroscience, University of California, San Francisco; San Francisco, CA 94158, USA
| | | | - Caroline Guglielmetti
- Department of Physical Therapy, University of California, San Francisco; San Francisco, CA 94158, USA
| | - Andres Cruz-Herranz
- Department of Neurology, Weill Institute for Neuroscience, University of California, San Francisco; San Francisco, CA 94158, USA
| | - Wendy Xin
- Department of Neurology, Weill Institute for Neuroscience, University of California, San Francisco; San Francisco, CA 94158, USA
| | | | - Jonah R Chan
- Department of Neurology, Weill Institute for Neuroscience, University of California, San Francisco; San Francisco, CA 94158, USA
| | - Ari J Green
- Department of Neurology, Weill Institute for Neuroscience, University of California, San Francisco; San Francisco, CA 94158, USA
| |
Collapse
|
14
|
Dietrich M, Hecker C, Martin E, Langui D, Gliem M, Stankoff B, Lubetzki C, Gruchot J, Göttle P, Issberner A, Nasiri M, Ramseier P, Beerli C, Tisserand S, Beckmann N, Shimshek D, Petzsch P, Akbar D, Levkau B, Stark H, Köhrer K, Hartung HP, Küry P, Meuth SG, Bigaud M, Zalc B, Albrecht P. Increased Remyelination and Proregenerative Microglia Under Siponimod Therapy in Mechanistic Models. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2022; 9:9/3/e1161. [PMID: 35354603 PMCID: PMC8969301 DOI: 10.1212/nxi.0000000000001161] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 01/27/2022] [Indexed: 12/11/2022]
Abstract
Background and Objectives Siponimod is an oral, selective sphingosine-1-phosphate receptor-1/5 modulator approved for treatment of multiple sclerosis. Methods Mouse MRI was used to investigate remyelination in the cuprizone model. We then used a conditional demyelination Xenopus laevis model to assess the dose-response of siponimod on remyelination. In experimental autoimmune encephalomyelitis–optic neuritis (EAEON) in C57Bl/6J mice, we monitored the retinal thickness and the visual acuity using optical coherence tomography and optomotor response. Optic nerve inflammatory infiltrates, demyelination, and microglial and oligodendroglial differentiation were assessed by immunohistochemistry, quantitative real-time PCR, and bulk RNA sequencing. Results An increased remyelination was observed in the cuprizone model. Siponimod treatment of demyelinated tadpoles improved remyelination in comparison to control in a bell-shaped dose-response curve. Siponimod in the EAEON model attenuated the clinical score, reduced the retinal degeneration, and improved the visual function after prophylactic and therapeutic treatment, also in a bell-shaped manner. Inflammatory infiltrates and demyelination of the optic nerve were reduced, the latter even after therapeutic treatment, which also shifted microglial differentiation to a promyelinating phenotype. Discussion These results confirm the immunomodulatory effects of siponimod and suggest additional regenerative and promyelinating effects, which follow the dynamics of a bell-shaped curve with high being less efficient than low concentrations.
Collapse
Affiliation(s)
- Michael Dietrich
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Christina Hecker
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Elodie Martin
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Dominique Langui
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Michael Gliem
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Bruno Stankoff
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Catherine Lubetzki
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Joel Gruchot
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Peter Göttle
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Andrea Issberner
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Milad Nasiri
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Pamela Ramseier
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Christian Beerli
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Sarah Tisserand
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Nicolau Beckmann
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Derya Shimshek
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Patrick Petzsch
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - David Akbar
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Bodo Levkau
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Holger Stark
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Karl Köhrer
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Hans-Peter Hartung
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Patrick Küry
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Sven Günther Meuth
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Marc Bigaud
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Bernard Zalc
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Philipp Albrecht
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| |
Collapse
|
15
|
Failed remyelination of the nonhuman primate optic nerve leads to axon degeneration, retinal damages, and visual dysfunction. Proc Natl Acad Sci U S A 2022; 119:e2115973119. [PMID: 35235463 PMCID: PMC8916013 DOI: 10.1073/pnas.2115973119] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Promotion of remyelination has become a new therapeutic avenue to prevent neuronal degeneration and promote recovery in white matter diseases, such as multiple sclerosis (MS). To date most of these strategies have been developed in short-lived rodent models of demyelination, which spontaneously repair. Well-defined nonhuman primate models closer to man would allow us to efficiently advance therapeutic approaches. Here we present a nonhuman primate model of optic nerve demyelination that recapitulates several features of MS lesions. The model leads to failed remyelination, associated with progressive axonal degeneration and visual dysfunction, thus providing the missing link to translate emerging preclinical therapies to the clinic for myelin disorders such as MS. White matter disorders of the central nervous system (CNS), such as multiple sclerosis (MS), lead to failure of nerve conduction and long-lasting neurological disabilities affecting a variety of sensory and motor systems, including vision. While most disease-modifying therapies target the immune and inflammatory response, the promotion of remyelination has become a new therapeutic avenue to prevent neuronal degeneration and promote recovery. Most of these strategies have been developed in short-lived rodent models of demyelination, which spontaneously repair and do not reflect the size, organization, and biology of the human CNS. Thus, well-defined nonhuman primate models are required to efficiently advance therapeutic approaches for patients. Here, we followed the consequence of long-term toxin-induced demyelination of the macaque optic nerve on remyelination and axon preservation, as well as its impact on visual functions. Findings from oculomotor behavior, ophthalmic examination, electrophysiology, and retinal imaging indicate visual impairment involving the optic nerve and retina. These visual dysfunctions fully correlated at the anatomical level, with sustained optic nerve demyelination, axonal degeneration, and alterations of the inner retinal layers. This nonhuman primate model of chronic optic nerve demyelination associated with axonal degeneration and visual dysfunction, recapitulates several key features of MS lesions and should be instrumental in providing the missing link to translate emerging repair promyelinating/neuroprotective therapies to the clinic for myelin disorders, such as MS.
Collapse
|
16
|
Jin J, Shneyderman M, Smith MD, Gharagozloo M, Sotirchos ES, Calabresi PA. Retinal pathology in spontaneous opticospinal experimental autoimmune encephalitis mice. J Neuroimmunol 2022; 367:577859. [DOI: 10.1016/j.jneuroim.2022.577859] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/20/2022] [Accepted: 03/28/2022] [Indexed: 11/30/2022]
|
17
|
Remlinger J, Madarasz A, Guse K, Hoepner R, Bagnoud M, Meli I, Feil M, Abegg M, Linington C, Shock A, Boroojerdi B, Kiessling P, Smith B, Enzmann V, Chan A, Salmen A. Antineonatal Fc Receptor Antibody Treatment Ameliorates MOG-IgG-Associated Experimental Autoimmune Encephalomyelitis. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2022; 9:9/2/e1134. [PMID: 35027475 PMCID: PMC8759074 DOI: 10.1212/nxi.0000000000001134] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 12/03/2021] [Indexed: 01/01/2023]
Abstract
BACKGROUND AND OBJECTIVES Myelin oligodendrocyte glycoprotein antibody-associated disorder (MOGAD) is a rare, autoimmune demyelinating CNS disorder, distinct from multiple sclerosis and neuromyelitis optica spectrum disorder. Characterized by pathogenic immunoglobulin G (IgG) antibodies against MOG, a potential treatment strategy for MOGAD is to reduce circulating IgG levels, e.g., by interference with the IgG recycling pathway mediated by the neonatal Fc receptor (FcRn). Although the optic nerve is often detrimentally involved in MOGAD, the effect of FcRn blockade on the visual pathway has not been assessed. Our objective was to investigate effects of a monoclonal anti-FcRn antibody in murine MOG-IgG-associated experimental autoimmune encephalomyelitis (EAE). METHODS We induced active MOG35-55 EAE in C57Bl/6 mice followed by the application of a monoclonal MOG-IgG (8-18C5) 10 days postimmunization (dpi). Animals were treated with either a specific monoclonal antibody against FcRn (α-FcRn, 4470) or an isotype-matched control IgG on 7, 10, and 13 dpi. Neurologic disability was scored daily on a 10-point scale. Visual acuity was assessed by optomotor reflex. Histopathologic hallmarks of disease were assessed in the spinal cord, optic nerve, and retina. Immune cell infiltration was visualized by immunohistochemistry, demyelination by Luxol fast blue staining and complement deposition and number of retinal ganglion cells by immunofluorescence. RESULTS In MOG-IgG-augmented MOG35-55 EAE, anti-FcRn treatment significantly attenuated neurologic disability over the course of disease (mean area under the curve and 95% confidence intervals (CIs): α-FcRn [n = 27], 46.02 [37.89-54.15]; isotype IgG [n = 24], 66.75 [59.54-73.96], 3 independent experiments), correlating with reduced amounts of demyelination and macrophage infiltration into the spinal cord. T- and B-cell infiltration and complement deposition remained unchanged. Compared with isotype, anti-FcRn treatment prevented reduction of visual acuity over the course of disease (median cycles/degree and interquartile range: α-FcRn [n = 16], 0.50 [0.48-0.55] to 0.50 [0.48-0.58]; isotype IgG [n = 17], 0.50 [0.49-0.54] to 0.45 [0.39-0.51]). DISCUSSION We show preserved optomotor response and ameliorated course of disease after anti-FcRn treatment in an experimental model using a monoclonal MOG-IgG to mimic MOGAD. Selectively targeting FcRn might represent a promising therapeutic approach in MOGAD.
Collapse
Affiliation(s)
- Jana Remlinger
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany
| | - Adrian Madarasz
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany
| | - Kirsten Guse
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany
| | - Robert Hoepner
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany
| | - Maud Bagnoud
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany
| | - Ivo Meli
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany
| | - Moritz Feil
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany
| | - Mathias Abegg
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany
| | - Christopher Linington
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany
| | - Anthony Shock
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany
| | - Babak Boroojerdi
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany
| | - Peter Kiessling
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany
| | - Bryan Smith
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany
| | - Volker Enzmann
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany
| | - Andrew Chan
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany
| | - Anke Salmen
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany.
| |
Collapse
|
18
|
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: 1.3] [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
|
19
|
Petrikowski L, Reinehr S, Haupeltshofer S, Deppe L, Graz F, Kleiter I, Dick HB, Gold R, Faissner S, Joachim SC. Progressive Retinal and Optic Nerve Damage in a Mouse Model of Spontaneous Opticospinal Encephalomyelitis. Front Immunol 2022; 12:759389. [PMID: 35140707 PMCID: PMC8818777 DOI: 10.3389/fimmu.2021.759389] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 12/29/2021] [Indexed: 11/13/2022] Open
Abstract
Neuromyelitis optica spectrum disorder (NMOSD) and myelin oligodendrocyte glycoprotein-antibody-associated disease (MOGAD) are antibody mediated CNS disorders mostly affecting the optic nerve and spinal cord with potential severe impact on the visual pathway. Here, we investigated inflammation and degeneration of the visual system in a spontaneous encephalomyelitis animal model. We used double-transgenic (2D2/Th) mice which develop a spontaneous opticospinal encephalomyelitis (OSE). Retinal morphology and its function were evaluated via spectral domain optical coherence tomography (SD-OCT) and electroretinography (ERG) in 6- and 8-week-old mice. Immunohistochemistry of retina and optic nerve and examination of the retina via RT-qPCR were performed using markers for inflammation, immune cells and the complement pathway. OSE mice showed clinical signs of encephalomyelitis with an incidence of 75% at day 38. A progressive retinal thinning was detected in OSE mice via SD-OCT. An impairment in photoreceptor signal transmission occurred. This was accompanied by cellular infiltration and demyelination of optic nerves. The number of microglia/macrophages was increased in OSE optic nerves and retinas. Analysis of the retina revealed a reduced retinal ganglion cell number and downregulated Pou4f1 mRNA expression in OSE retinas. RT-qPCR revealed an elevation of microglia markers and the cytokines Tnfa and Tgfb. We also documented an upregulation of the complement system via the classical pathway. In summary, we describe characteristics of inflammation and degeneration of the visual system in a spontaneous encephalomyelitis model, characterized by coinciding inflammatory and degenerative mechanisms in both retina and optic nerve with involvement of the complement system.
Collapse
Affiliation(s)
- Laura Petrikowski
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
- Department of Neurology, Ruhr-University Bochum, St. Josef-Hospital, Bochum, Germany
| | - Sabrina Reinehr
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Steffen Haupeltshofer
- Department of Neurology, Ruhr-University Bochum, St. Josef-Hospital, Bochum, Germany
| | - Leonie Deppe
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Florian Graz
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
- Department of Neurology, Ruhr-University Bochum, St. Josef-Hospital, Bochum, Germany
| | - Ingo Kleiter
- Department of Neurology, Ruhr-University Bochum, St. Josef-Hospital, Bochum, Germany
| | - H. Burkhard Dick
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Ralf Gold
- Department of Neurology, Ruhr-University Bochum, St. Josef-Hospital, Bochum, Germany
| | - Simon Faissner
- Department of Neurology, Ruhr-University Bochum, St. Josef-Hospital, Bochum, Germany
- *Correspondence: Simon Faissner, ; Stephanie C. Joachim,
| | - Stephanie C. Joachim
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
- *Correspondence: Simon Faissner, ; Stephanie C. Joachim,
| |
Collapse
|
20
|
Frenger MJ, Hecker C, Sindi M, Issberner A, Hartung HP, Meuth SG, Dietrich M, Albrecht P. Semi-Automated Live Tracking of Microglial Activation in CX3CR1 GFP Mice During Experimental Autoimmune Encephalomyelitis by Confocal Scanning Laser Ophthalmoscopy. Front Immunol 2021; 12:761776. [PMID: 34745138 PMCID: PMC8567040 DOI: 10.3389/fimmu.2021.761776] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 10/07/2021] [Indexed: 11/13/2022] Open
Abstract
Confocal scanning laser ophthalmoscopy (cSLO) is a non-invasive technique for real-time imaging of the retina. We developed a step-by-step protocol for the semi-automatic evaluation of myeloid cells in cSLO images from CX3CR1GFP mice, expressing green fluorescent protein (GFP) under control of the endogenous CX3C chemokine receptor 1 locus. We identified cSLO parameters allowing us to distinguish animals with experimental autoimmune encephalomyelitis (EAE) from sham-treated/naïve animals. Especially cell count (CC) and the total microglial area (SuA) turned out to be reliable parameters. Comparing the cSLO results with clinical parameters, we found significant correlations between the clinical EAE score and the SuA and of the inner retinal layer thickness, measured by optical coherence tomography, with the CC as well as the SuA. As a final step, we performed immunohistochemistry to confirm that the GFP-expressing cells visualized by the cSLO are Iba1 positive and validated the step-by-step protocol against manual counting. We present a semi-automatic step-by-step protocol with a balance between fast data evaluation and adequate accuracy, which is optimized by the option to manually adapt the contrast threshold. This protocol may be useful for numerous research questions on the role of microglial polarization in models of inflammatory and degenerating CNS diseases involving the retina.
Collapse
Affiliation(s)
- Moritz J. Frenger
- Department of Neurology, Heinrich-Heine University Düsseldorf, Medical Faculty, Düsseldorf, Germany
| | - Christina Hecker
- Department of Neurology, Heinrich-Heine University Düsseldorf, Medical Faculty, Düsseldorf, Germany
| | - Mustafa Sindi
- Department of Neurology, Heinrich-Heine University Düsseldorf, Medical Faculty, Düsseldorf, Germany
| | - Andrea Issberner
- Department of Neurology, Heinrich-Heine University Düsseldorf, Medical Faculty, Düsseldorf, Germany
| | - Hans-Peter Hartung
- Department of Neurology, Heinrich-Heine University Düsseldorf, Medical Faculty, Düsseldorf, Germany
- Brain and Mind Center, University of Sydney, Sydney, NSW, Australia
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Sven G. Meuth
- Department of Neurology, Heinrich-Heine University Düsseldorf, Medical Faculty, Düsseldorf, Germany
| | - Michael Dietrich
- Department of Neurology, Heinrich-Heine University Düsseldorf, Medical Faculty, Düsseldorf, Germany
| | - Philipp Albrecht
- Department of Neurology, Heinrich-Heine University Düsseldorf, Medical Faculty, Düsseldorf, Germany
| |
Collapse
|
21
|
Ingwersen J, Graf J, Kluge J, Weise M, Dietrich M, Lee JI, Harmel J, Hartung HP, Ruck T, Meuth SG, Albrecht P, Aktas O, Ringelstein M. CNS Involvement in Chronic Inflammatory Demyelinating Polyneuropathy: Subtle Retinal Changes in Optical Coherence Tomography. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2021; 9:9/1/e1099. [PMID: 34667130 PMCID: PMC8529418 DOI: 10.1212/nxi.0000000000001099] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 08/30/2021] [Indexed: 11/30/2022]
Abstract
Background and Objectives Chronic inflammatory demyelinating polyneuropathy (CIDP) is an autoimmune disease primarily affecting the peripheral nervous system. However, several noncontrolled studies have suggested concomitant inflammatory CNS demyelination similar to multiple sclerosis. The aim of this study was to investigate an involvement of the visual pathway in patients with CIDP. Methods In this prospective cross-sectional study, we used high-resolution spectral-domain optical coherence tomography to compare the thickness of the peripapillary retinal nerve fiber layer and the deeper macular retinal layers as well as the total macular volume (TMV) in 22 patients with CIDP and 22 age-matched and sex-matched healthy control (HC) individuals. Retinal layers were semiautomatically segmented by the provided software and were correlated with clinical measures and nerve conduction studies. Results In patients with CIDP compared with healthy age-matched and sex-matched controls, we found slight but significant volume reductions of the ganglion cell/inner plexiform layer complex (CIDP 1.86 vs HC 1.95 mm3, p = 0.015), the retinal pigment epithelium (CIDP 0.38 vs HC 0.40 mm3, p = 0.02), and the TMV (CIDP 8.48 vs HC 8.75 mm3, p = 0.018). The ganglion cell layer volume and motor nerve conduction velocity were positively associated (B = 0.002, p = 0.02). Discussion Our data reveal subtle retinal neurodegeneration in patients with CIDP, providing evidence for visual pathway involvement, detectable by OCT. The results need corroboration in independent, larger cohorts.
Collapse
Affiliation(s)
- Jens Ingwersen
- From the Department of Neurology (J.A., J.G., J.K., M.W., M.D., J.-I.L., J.H., H.-P.H., T.R., S.G.M., P.A., O.A., M.R.), Medical Faculty, Heinrich-Heine-University Düsseldorf, Germany; Brain and Mind Centre (H.-P.H.), University of Sydney; Department of Neurology (H.-P.H.), Medical University of Vienna, Austria; Department of Neurology (H.-P.H., M.R.), Center for Neurology and Neuropsychiatry, LVR-Klinikum Düsseldorf, Germany; and Department of Neurology (H.-P.H.), Palacky University in Olomouc, Olomouc, Czech Republic
| | - Jonas Graf
- From the Department of Neurology (J.A., J.G., J.K., M.W., M.D., J.-I.L., J.H., H.-P.H., T.R., S.G.M., P.A., O.A., M.R.), Medical Faculty, Heinrich-Heine-University Düsseldorf, Germany; Brain and Mind Centre (H.-P.H.), University of Sydney; Department of Neurology (H.-P.H.), Medical University of Vienna, Austria; Department of Neurology (H.-P.H., M.R.), Center for Neurology and Neuropsychiatry, LVR-Klinikum Düsseldorf, Germany; and Department of Neurology (H.-P.H.), Palacky University in Olomouc, Olomouc, Czech Republic
| | - Julia Kluge
- From the Department of Neurology (J.A., J.G., J.K., M.W., M.D., J.-I.L., J.H., H.-P.H., T.R., S.G.M., P.A., O.A., M.R.), Medical Faculty, Heinrich-Heine-University Düsseldorf, Germany; Brain and Mind Centre (H.-P.H.), University of Sydney; Department of Neurology (H.-P.H.), Medical University of Vienna, Austria; Department of Neurology (H.-P.H., M.R.), Center for Neurology and Neuropsychiatry, LVR-Klinikum Düsseldorf, Germany; and Department of Neurology (H.-P.H.), Palacky University in Olomouc, Olomouc, Czech Republic
| | - Margit Weise
- From the Department of Neurology (J.A., J.G., J.K., M.W., M.D., J.-I.L., J.H., H.-P.H., T.R., S.G.M., P.A., O.A., M.R.), Medical Faculty, Heinrich-Heine-University Düsseldorf, Germany; Brain and Mind Centre (H.-P.H.), University of Sydney; Department of Neurology (H.-P.H.), Medical University of Vienna, Austria; Department of Neurology (H.-P.H., M.R.), Center for Neurology and Neuropsychiatry, LVR-Klinikum Düsseldorf, Germany; and Department of Neurology (H.-P.H.), Palacky University in Olomouc, Olomouc, Czech Republic
| | - Michael Dietrich
- From the Department of Neurology (J.A., J.G., J.K., M.W., M.D., J.-I.L., J.H., H.-P.H., T.R., S.G.M., P.A., O.A., M.R.), Medical Faculty, Heinrich-Heine-University Düsseldorf, Germany; Brain and Mind Centre (H.-P.H.), University of Sydney; Department of Neurology (H.-P.H.), Medical University of Vienna, Austria; Department of Neurology (H.-P.H., M.R.), Center for Neurology and Neuropsychiatry, LVR-Klinikum Düsseldorf, Germany; and Department of Neurology (H.-P.H.), Palacky University in Olomouc, Olomouc, Czech Republic
| | - John-Ih Lee
- From the Department of Neurology (J.A., J.G., J.K., M.W., M.D., J.-I.L., J.H., H.-P.H., T.R., S.G.M., P.A., O.A., M.R.), Medical Faculty, Heinrich-Heine-University Düsseldorf, Germany; Brain and Mind Centre (H.-P.H.), University of Sydney; Department of Neurology (H.-P.H.), Medical University of Vienna, Austria; Department of Neurology (H.-P.H., M.R.), Center for Neurology and Neuropsychiatry, LVR-Klinikum Düsseldorf, Germany; and Department of Neurology (H.-P.H.), Palacky University in Olomouc, Olomouc, Czech Republic
| | - Jens Harmel
- From the Department of Neurology (J.A., J.G., J.K., M.W., M.D., J.-I.L., J.H., H.-P.H., T.R., S.G.M., P.A., O.A., M.R.), Medical Faculty, Heinrich-Heine-University Düsseldorf, Germany; Brain and Mind Centre (H.-P.H.), University of Sydney; Department of Neurology (H.-P.H.), Medical University of Vienna, Austria; Department of Neurology (H.-P.H., M.R.), Center for Neurology and Neuropsychiatry, LVR-Klinikum Düsseldorf, Germany; and Department of Neurology (H.-P.H.), Palacky University in Olomouc, Olomouc, Czech Republic
| | - Hans-Peter Hartung
- From the Department of Neurology (J.A., J.G., J.K., M.W., M.D., J.-I.L., J.H., H.-P.H., T.R., S.G.M., P.A., O.A., M.R.), Medical Faculty, Heinrich-Heine-University Düsseldorf, Germany; Brain and Mind Centre (H.-P.H.), University of Sydney; Department of Neurology (H.-P.H.), Medical University of Vienna, Austria; Department of Neurology (H.-P.H., M.R.), Center for Neurology and Neuropsychiatry, LVR-Klinikum Düsseldorf, Germany; and Department of Neurology (H.-P.H.), Palacky University in Olomouc, Olomouc, Czech Republic
| | - Tobias Ruck
- From the Department of Neurology (J.A., J.G., J.K., M.W., M.D., J.-I.L., J.H., H.-P.H., T.R., S.G.M., P.A., O.A., M.R.), Medical Faculty, Heinrich-Heine-University Düsseldorf, Germany; Brain and Mind Centre (H.-P.H.), University of Sydney; Department of Neurology (H.-P.H.), Medical University of Vienna, Austria; Department of Neurology (H.-P.H., M.R.), Center for Neurology and Neuropsychiatry, LVR-Klinikum Düsseldorf, Germany; and Department of Neurology (H.-P.H.), Palacky University in Olomouc, Olomouc, Czech Republic
| | - Sven G Meuth
- From the Department of Neurology (J.A., J.G., J.K., M.W., M.D., J.-I.L., J.H., H.-P.H., T.R., S.G.M., P.A., O.A., M.R.), Medical Faculty, Heinrich-Heine-University Düsseldorf, Germany; Brain and Mind Centre (H.-P.H.), University of Sydney; Department of Neurology (H.-P.H.), Medical University of Vienna, Austria; Department of Neurology (H.-P.H., M.R.), Center for Neurology and Neuropsychiatry, LVR-Klinikum Düsseldorf, Germany; and Department of Neurology (H.-P.H.), Palacky University in Olomouc, Olomouc, Czech Republic
| | - Philipp Albrecht
- From the Department of Neurology (J.A., J.G., J.K., M.W., M.D., J.-I.L., J.H., H.-P.H., T.R., S.G.M., P.A., O.A., M.R.), Medical Faculty, Heinrich-Heine-University Düsseldorf, Germany; Brain and Mind Centre (H.-P.H.), University of Sydney; Department of Neurology (H.-P.H.), Medical University of Vienna, Austria; Department of Neurology (H.-P.H., M.R.), Center for Neurology and Neuropsychiatry, LVR-Klinikum Düsseldorf, Germany; and Department of Neurology (H.-P.H.), Palacky University in Olomouc, Olomouc, Czech Republic
| | - Orhan Aktas
- From the Department of Neurology (J.A., J.G., J.K., M.W., M.D., J.-I.L., J.H., H.-P.H., T.R., S.G.M., P.A., O.A., M.R.), Medical Faculty, Heinrich-Heine-University Düsseldorf, Germany; Brain and Mind Centre (H.-P.H.), University of Sydney; Department of Neurology (H.-P.H.), Medical University of Vienna, Austria; Department of Neurology (H.-P.H., M.R.), Center for Neurology and Neuropsychiatry, LVR-Klinikum Düsseldorf, Germany; and Department of Neurology (H.-P.H.), Palacky University in Olomouc, Olomouc, Czech Republic
| | - Marius Ringelstein
- From the Department of Neurology (J.A., J.G., J.K., M.W., M.D., J.-I.L., J.H., H.-P.H., T.R., S.G.M., P.A., O.A., M.R.), Medical Faculty, Heinrich-Heine-University Düsseldorf, Germany; Brain and Mind Centre (H.-P.H.), University of Sydney; Department of Neurology (H.-P.H.), Medical University of Vienna, Austria; Department of Neurology (H.-P.H., M.R.), Center for Neurology and Neuropsychiatry, LVR-Klinikum Düsseldorf, Germany; and Department of Neurology (H.-P.H.), Palacky University in Olomouc, Olomouc, Czech Republic.
| |
Collapse
|
22
|
Mehmood A, Ali W, Song S, Din ZU, Guo RY, Shah W, Ilahi I, Yin B, Yan H, Zhang L, Khan M, Ali W, Zeb L, Safari H, Li B. Optical coherence tomography monitoring and diagnosing retinal changes in multiple sclerosis. Brain Behav 2021; 11:e2302. [PMID: 34520634 PMCID: PMC8553325 DOI: 10.1002/brb3.2302] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 06/22/2021] [Accepted: 07/12/2021] [Indexed: 12/14/2022] Open
Abstract
This study explores the use of optical coherence tomography (OCT) to monitor and diagnose multiple sclerosis (MS). The analysis of reduced total macular volume and peripapillary retinal nerve fiber layer thinning are shown. The severity of these defects increases as MS progresses, reflecting the progressive degeneration of nerve fibers and retinal ganglion cells. The OCT parameters are noninvasive, sensitive indicators that can be used to assess the progression of neurodegeneration and inflammation in MS.
Collapse
Affiliation(s)
- Arshad Mehmood
- Department of Neurology, The Second Hospital of Hebei Medical University, City Shijiazhuang, Hebei Province, P. R. China.,Key Laboratory of Neurology of Hebei Province, City Shijiazhuang, Hebei Province, P. R. China
| | - Wajid Ali
- Key Laboratory of Functional Inorganic Materials Chemistry, School of Chemistry and Materials Science, Heilongjiang University, Harbin, P. R. China
| | - Shuang Song
- Department of Neurology, The Second Hospital of Hebei Medical University, City Shijiazhuang, Hebei Province, P. R. China.,Key Laboratory of Neurology of Hebei Province, City Shijiazhuang, Hebei Province, P. R. China
| | - Zaheer Ud Din
- Institute of Cancer Stem Cell, Dalian Medical University, Liaoning Province, P. R. China
| | - Ruo-Yi Guo
- Department of Neurology, The Second Hospital of Hebei Medical University, City Shijiazhuang, Hebei Province, P. R. China.,Key Laboratory of Neurology of Hebei Province, City Shijiazhuang, Hebei Province, P. R. China
| | - Wahid Shah
- Department of Physiology, Hebei Medical University, Shijiazhuang, Hebei, P. R. China
| | - Ikram Ilahi
- Department of Zoology, University of Malakand, Chakdara, Khyber Pakhtunkhwa, Pakistan
| | - Bowen Yin
- Department of Neurology, The Second Hospital of Hebei Medical University, City Shijiazhuang, Hebei Province, P. R. China.,Key Laboratory of Neurology of Hebei Province, City Shijiazhuang, Hebei Province, P. R. China.,Department of Neurology, The First Hospital of Qinhuangdao, Qinhuangdao, Hebei, P. R. China
| | - Hongjing Yan
- Department of Neurology, The Second Hospital of Hebei Medical University, City Shijiazhuang, Hebei Province, P. R. China.,Key Laboratory of Neurology of Hebei Province, City Shijiazhuang, Hebei Province, P. R. China
| | - Lu Zhang
- Department of Neurology, The Second Hospital of Hebei Medical University, City Shijiazhuang, Hebei Province, P. R. China.,Key Laboratory of Neurology of Hebei Province, City Shijiazhuang, Hebei Province, P. R. China
| | - Murad Khan
- Department of Genetics, Hebei Key Lab of Laboratory Animal, Hebei Medical University, Shijiazhuang, Hebei Province, P. R. China
| | - Wajid Ali
- Green and Environmental Chemistry, Ecotoxicology and Ecology Laboratory, Department of Zoology, University of Malakand, Chakdara, Khyber Pakhtunkhwa, Pakistan
| | - Liaqat Zeb
- School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, P.R. China
| | - Hamidreza Safari
- Department of Immunology, Torbat Jam Faculty of Medical Sciences, Torbat Jam, Iran
| | - Bin Li
- Department of Neurology, The Second Hospital of Hebei Medical University, City Shijiazhuang, Hebei Province, P. R. China.,Key Laboratory of Neurology of Hebei Province, City Shijiazhuang, Hebei Province, P. R. China
| |
Collapse
|
23
|
Cruz-Herranz A, Oertel FC, Kim K, Cantó E, Timmons G, Sin JH, Devereux M, Baker N, Michel B, Schubert RD, Rani L, Cordano C, Baranzini SE, Green AJ. Distinctive waves of innate immune response in the retina in experimental autoimmune encephalomyelitis. JCI Insight 2021; 6:e149228. [PMID: 34100385 PMCID: PMC8262300 DOI: 10.1172/jci.insight.149228] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 04/28/2021] [Indexed: 12/27/2022] Open
Abstract
Neurodegeneration mediates neurological disability in inflammatory demyelinating diseases of the CNS. The role of innate immune cells in mediating this damage has remained controversial with evidence for destructive and protective effects. This has complicated efforts to develop treatment. The time sequence and dynamic evolution of the opposing functions are especially unclear. Given limits of in vivo monitoring in human diseases such as multiple sclerosis (MS), animal models are warranted to investigate the association and timing of innate immune activation with neurodegeneration. Using noninvasive in vivo retinal imaging of experimental autoimmune encephalitis (EAE) in CX3CR1GFP/+–knock-in mice followed by transcriptional profiling, we are able to show 2 distinct waves separated by a marked reduction in the number of innate immune cells and change in cell morphology. The first wave is characterized by an inflammatory phagocytic phenotype preceding the onset of EAE, whereas the second wave is characterized by a regulatory, antiinflammatory phenotype during the chronic stage. Additionally, the magnitude of the first wave is associated with neuronal loss. Two transcripts identified — growth arrest–specific protein 6 (GAS6) and suppressor of cytokine signaling 3 (SOCS3) — might be promising targets for enhancing protective effects of microglia in the chronic phase after initial injury.
Collapse
Affiliation(s)
- Andrés Cruz-Herranz
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Frederike C Oertel
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, California, USA.,Experimental and Clinical Research Center (ECRC), Max-Delbrück-Centrum for Molecular Medicine, and.,NeuroCure Clinical Research Center (NCRC), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Kicheol Kim
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Ester Cantó
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Garrett Timmons
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Jung H Sin
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Michael Devereux
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Nicholas Baker
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Brady Michel
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Ryan D Schubert
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Lakshmisahithi Rani
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Christian Cordano
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Sergio E Baranzini
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Ari J Green
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, California, USA.,Department of Ophthalmology, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
24
|
Reduced macular thickness and macular vessel density in early-treated adult patients with PKU. Mol Genet Metab Rep 2021; 27:100767. [PMID: 34026550 PMCID: PMC8121983 DOI: 10.1016/j.ymgmr.2021.100767] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 04/27/2021] [Accepted: 04/28/2021] [Indexed: 11/21/2022] Open
Abstract
Purpose Macular structure is poorly evaluated in early-treated phenylketonuria (ETPKU). To evaluate potential changes, we aimed to examine retinas of PKU patients using optical coherence tomography (OCT) with additional OCT angiography (OCTA) and compare the results to healthy controls. Methods A total of 100 adults were recruited in this monocentric, case-control study: 50 patients with ETPKU (mean age: 30.66 ± 8.00 years) and 50 healthy controls (mean age: 30.45 ± 7.18 years). Macular thickness, vessel density and flow area of the right eye was assessed with spectral domain OCT angiography SD-OCT(A). Macular microstructural data between the ETPKU and control group was compared. In the ETPKU group, the relationship between visual functional parameters (best corrected visual acuity [VA], spherical equivalent [SE], contrast sensitivity [CS] and near stereoacuity) and microstructural alterations was examined. The dependency of OCT(A) values on serum phenylalanine (Phe) level was analysed. Results There was significant average parafoveal and perifoveal total retinal layer thinning in ETPKU patients compared to healthy controls (p < 0.016 and p < 0.001, respectively), while the foveal region remained unchanged in the ETPKU group. Whole macular and parafoveal superficial capillary plexus density was significantly decreased in ETPKU compared to controls (p < 0.001). There were no significant differences in the foveal avascular zone, nonflow area, macular superficial and deep capillary plexus between the groups. The temporal parafoveal inner retinal layer thickness was found to negatively correlate with individual Phe levels (r = -0.35, p = 0.042). There was no difference in vascular density and retinal thickness in the subgroup analysis of patients with good therapy adherence compared to patients on a relaxed diet. Conclusions Durable elevation in Phe levels are only partially associated with macular retinal structural changes. However, therapy adherence might not influence these ophthalmological complications.
Collapse
Key Words
- Dopamine
- Macular retinal vessel density
- Macular thickness
- Optical Coherence Tomography Angiography, (OCTA)
- Optical Coherence Tomography, (OCT)
- Optical coherence tomography angiography
- Parkinson's disease, (PD)
- Phenylalanine level
- Phenylalanine, (Phe)
- Phenylketonuria
- Tyrosine, (Tyr)
- aminoacid supplements, (AAS)
- axial length, (AL)
- contrast sensitivity, (CS)
- deep capillary plexus, (DCP)
- diopters, (D)
- dopamine, (DA)
- early treated phenylketonuria, (ETPKU)
- external limiting membrane/inner segment of photoreceptors/outer segment of photoreceptors, (ELM/IS/OS)
- ganglion cell complex, (GCC)
- ganglion cell layer, (GCL)
- inner limiting membrane, (ILM)
- inner nuclear layer, (INL)
- inner plexiform layer, (IPL)
- inner retinal layer, (IRL)
- intraocular pressure, (IOP)
- outer nuclear layer, (ONL)
- outer plexiform layer, (OPL)
- outer retinal layer, (ORL)
- phenylalanine hydroxylase gene, (PAH)
- phenylketonuria, (PKU)
- retinal nerve fiber layer, (RNFL)
- retinal pigment epithelium, (RPE)
- signal strength index, (SSI)
- sine-wave contrast test, (SWCT)
- spectral domain, (SD)
- spherical equivalent, (SE)
- split-spectrum amplitude-decorrelation angiography, (SSADA)
- superficial capillary plexus, (SCP)
- tetrahydrobiopterin, (BH4)
- total retinal layer thickness, (TRLT)
- vessel density, (VD)
- visual acuity, (VA)
- visual evoked potential, (VEP)
Collapse
|
25
|
Sekyi MT, Lauderdale K, Atkinson KC, Golestany B, Karim H, Feri M, Soto JS, Diaz C, Kim SH, Cilluffo M, Nusinowitz S, Katzenellenbogen JA, Tiwari‐Woodruff SK. Alleviation of extensive visual pathway dysfunction by a remyelinating drug in a chronic mouse model of multiple sclerosis. Brain Pathol 2021; 31:312-332. [PMID: 33368801 PMCID: PMC8018057 DOI: 10.1111/bpa.12930] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 12/09/2020] [Accepted: 12/11/2020] [Indexed: 11/30/2022] Open
Abstract
Visual deficits are among the most prevalent symptoms in patients with multiple sclerosis (MS). To understand deficits in the visual pathway during MS and potential treatment effects, we used experimental autoimmune encephalomyelitis (EAE), the most commonly used animal model of MS. The afferent visual pathway was assessed in vivo using optical coherence tomography (OCT), electroretinography (ERG), and visually evoked cortical potentials (VEPs). Inflammation, demyelination, and neurodegeneration were examined by immunohistochemistry ex vivo. In addition, an immunomodulatory, remyelinating agent, the estrogen receptor β ligand chloroindazole (IndCl), was tested for its therapeutic potential in the visual pathway. EAE produced functional deficits in visual system electrophysiology, including suppression of ERG and VEP waveform amplitudes and increased signal latencies. Therapeutic IndCl rescued overall visual system latency by VEP but had little impact on amplitude or ERG findings relative to vehicle. Faster VEP conduction in IndCl-treated mice was associated with enhanced myelin basic protein signal in all visual system structures examined. IndCl preserved retinal ganglion cells (RGCs) and oligodendrocyte density in the prechiasmatic white matter, but similar retinal nerve fiber layer thinning by OCT was noted in vehicle and IndCl-treated mice. Although IndCl differentially attenuated leukocyte and astrocyte staining signal throughout the structures analyzed, axolemmal varicosities were observed in all visual fiber tracts of mice with EAE irrespective of treatment, suggesting impaired axonal energy homeostasis. These data support incomplete functional recovery of VEP amplitude with IndCl, as fiber tracts displayed persistent axon pathology despite remyelination-induced decreases in latencies, evidenced by reduced optic nerve g-ratio in IndCl-treated mice. Although additional studies are required, these findings demonstrate the dynamics of visual pathway dysfunction and disability during EAE, along with the importance of early treatment to mitigate EAE-induced axon damage.
Collapse
Affiliation(s)
- Maria T. Sekyi
- Division of Biomedical SciencesRiverside School of MedicineUniversity of CaliforniaRiversideCAUSA
- Department of BioengineeringRiverside Bourns School of EngineeringUniversity of CaliforniaRiversideCAUSA
| | - Kelli Lauderdale
- Division of Biomedical SciencesRiverside School of MedicineUniversity of CaliforniaRiversideCAUSA
| | - Kelley C. Atkinson
- Division of Biomedical SciencesRiverside School of MedicineUniversity of CaliforniaRiversideCAUSA
| | - Batis Golestany
- Division of Biomedical SciencesRiverside School of MedicineUniversity of CaliforniaRiversideCAUSA
| | - Hawra Karim
- Division of Biomedical SciencesRiverside School of MedicineUniversity of CaliforniaRiversideCAUSA
| | - Micah Feri
- Division of Biomedical SciencesRiverside School of MedicineUniversity of CaliforniaRiversideCAUSA
| | - Joselyn S. Soto
- Division of Biomedical SciencesRiverside School of MedicineUniversity of CaliforniaRiversideCAUSA
| | - Cobi Diaz
- Division of Biomedical SciencesRiverside School of MedicineUniversity of CaliforniaRiversideCAUSA
| | - Sung Hoon Kim
- Department of Chemistry and Cancer CenterUniversity of Illinois at Urbana‐ChampaignUrbanaILUSA
| | - Marianne Cilluffo
- BRI Electron Microscopy LaboratoryLos Angeles School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Steven Nusinowitz
- Stein Eye InstituteLos Angeles School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | | | - Seema K. Tiwari‐Woodruff
- Division of Biomedical SciencesRiverside School of MedicineUniversity of CaliforniaRiversideCAUSA
| |
Collapse
|
26
|
Dietrich M, Hecker C, Nasiri M, Samsam S, Issberner A, Kohne Z, Hartung HP, Albrecht P. Neuroprotective Properties of Dimethyl Fumarate Measured by Optical Coherence Tomography in Non-inflammatory Animal Models. Front Neurol 2021; 11:601628. [PMID: 33519681 PMCID: PMC7838501 DOI: 10.3389/fneur.2020.601628] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 12/15/2020] [Indexed: 12/26/2022] Open
Abstract
While great advances have been made in the immunomodulatory treatment of multiple sclerosis (MS), there is still an unmet need for drugs with neuroprotective potential. Dimethyl fumarate (DMF) has been suggested to exert both immunomodulatory and neuroprotective effects in MS. To investigate if DMF has neuroprotective effects independent of immunomodulation we evaluated its effects in the non-inflammatory animal models of light-induced photoreceptor loss and optic nerve crush. This might also reveal applications for DMF besides MS, such as age related macular degeneration. Retinal neurodegeneration was longitudinally assessed by in vivo retinal imaging using optical coherence tomography (OCT), and glutathione (GSH) measurements as well as histological investigations were performed to clarify the mode of action. For light-induced photoreceptor loss, one eye of C57BL/6J mice was irradiated with a LED cold light lamp while for optic nerve crush the optic nerve was clamped behind the eye bulb. The other eye served as control. GSH was measured in the optic nerve, choroid and retina and immunohistological staining of retinal microglia (Iba1) was performed. Mice were treated with 15 or 30 mg DMF/kg bodyweight or vehicle. While no protective effects were observed in optic nerve crush, in the light-induced retinal degeneration model DMF treatment significantly reduced retinal degeneration. In these mice, GSH levels in the retina and surrounding choroid were increased and histological investigations revealed less microglial activation in the outer retinal layers, suggesting both antioxidant and anti-inflammatory effects.
Collapse
Affiliation(s)
- Michael Dietrich
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Christina Hecker
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Milad Nasiri
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Sogol Samsam
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Andrea Issberner
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Zippora Kohne
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Philipp Albrecht
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
27
|
Abstract
Optic neuritis (ON) is an inflammatory attack of the optic nerve that leads to visual disability. It is the most common optic neuropathy affecting healthy young adults, most commonly women aged 20-45 years. It can be idiopathic and monophasic or as part of a neurologic disease such as multiple sclerosis with recurrence and cumulative damage. Currently, there is no therapy to repair the damage from optic neuritis. Animal models are an essential tool for the understanding of the pathogenesis of optic neuritis and for the development of potential treatment strategies. Experimental autoimmune encephalomyelitis (EAE) is the most commonly used experimental rodent model for human autoimmune inflammatory demyelinating diseases of the central nervous system (CNS). In this review, we discuss the latest rodent models regarding optic neuritis, focusing on EAE model, and on its recent achievements and developments.
Collapse
Affiliation(s)
- Yael Redler
- Department of Neuro-Ophthalmology, Massachusetts Eye & Ear Infirmary/Harvard Medical School, Boston, MA, United States
| | - Michael Levy
- Department of Neurology, Massachusetts General Hospital/Harvard Medical School, Boston, MA, United States
| |
Collapse
|
28
|
Hecker C, Dietrich M, Issberner A, Hartung HP, Albrecht P. Comparison of different optomotor response readouts for visual testing in experimental autoimmune encephalomyelitis-optic neuritis. J Neuroinflammation 2020; 17:216. [PMID: 32682447 PMCID: PMC7368714 DOI: 10.1186/s12974-020-01889-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 07/06/2020] [Indexed: 12/12/2022] Open
Abstract
Optomotor response is increasingly used in preclinical research for evaluating the visual function in rodents. However, the most suitable measuring protocol for specific scientific questions is not always established. We aimed to determine the optimal parameters for visual function analysis in experimental autoimmune encephalomyelitis optic neuritis (EAEON), an animal model for multiple sclerosis. Contrast sensitivity as well as spatial frequency both had a low variance and a good test-retest reliability. Also, both parameters were able to differentiate between the EAEON and the control group. Correlations with the retinal degeneration, assessed by optical coherence tomography, the infiltration of immune cells, and the clinical disability score revealed that spatial frequency was superior to contrast sensitivity analysis. We therefore conclude that spatial frequency testing is better suited as visual acuity assessment in C57Bl/6 J EAEON mice. Furthermore, contrast sensitivity measurements are more time consuming, possibly leading to more stress for the animals.
Collapse
Affiliation(s)
- Christina Hecker
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Michael Dietrich
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Andrea Issberner
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Philipp Albrecht
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany.
| |
Collapse
|
29
|
Esmael A, Elsherif M, Abdelsalam M, Sabry D, Mamdouh M, Belal T. Retinal thickness as a potential biomarker of neurodegeneration and a predictor of early cognitive impairment in patients with multiple sclerosis. Neurol Res 2020; 42:564-574. [PMID: 32370626 DOI: 10.1080/01616412.2020.1761174] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
OBJECTIVES The purpose of this research is to predict the cognitive impairment and to determine its correlation with retinal thickness, mainly (RFNL and GCIPL) in cases of multiple sclerosis. METHODS 60 multiple sclerosis patients and 30 age and sex-matched healthy controls were included in this study. Cognitive functions were evaluated in all study participants by the Montreal Cognitive Assessment (MoCA). OCT imaging was done to determine the thickness. The correlation between the cognitive domains of MoCA and the thickness of the retinal nerve fiber layers was analyzed by Spearman correlation. ROC curve was constructed to determine the cut-off points for retinal thickness, and a binary logistic regression was performed to determine the independent predictive capacity of established cut-off points. RESULTS Impaired cognition was found in 26 MS patients (43.3%). Cognitively impaired patients were significantly older (P < 0.05), had significantly longer disease duration (P < 0.05), had higher average EDSS scores (4.3 ± 1.22 vs 3.1 ± 1.45, P < 0.001), and occurred more in progressive types of MS (P < 0.001). A significant positive correlation was found between cognitive function and RNFL thickness and GCIPL (P < 0.001). The retinal thickness (RNFL and GCIPL) cut-off points established for the prediction of cognitive impairment in MS patients were 79 μm and 76 μm, respectively. CONCLUSION The clear correlation between cognitive impairment and atrophy of inner retinal layers (RNFL and GCIPL) proposes that OCT is valuable in evaluating the neurodegeneration and prediction of early cognitive impairment in MS. ABBREVIATIONS EDSS: Expanded Disability Status Scale; HCs: Healthy controls; GCIPL: Ganglion cell-inner plexiform layer; ILM: Internal limiting membrane; INL: Inner nuclear layer; MoCA: Montreal Cognitive Assessment; MS: Multiple sclerosis; PPMS: Primary progressive multiple sclerosis; RNFL: Retinal nerve fiber layer; RRMS: Relapsing-remitting multiple sclerosis; SD: Standard deviations; SPMS: Secondary progressive multiple sclerosis; SPSS: Statistical Package for the Social Sciences.
Collapse
Affiliation(s)
- Ahmed Esmael
- Neurology Department, Mansoura University Hospital
| | | | | | - Dalia Sabry
- Ophthalmic Center, Mansoura University , Mansoura, Egypt
| | | | - Tamer Belal
- Neurology Department, Mansoura University Hospital
| |
Collapse
|