1
|
Stys PK, Tsutsui S, Gafson AR, ‘t Hart BA, Belachew S, Geurts JJG. New views on the complex interplay between degeneration and autoimmunity in multiple sclerosis. Front Cell Neurosci 2024; 18:1426231. [PMID: 39161786 PMCID: PMC11330826 DOI: 10.3389/fncel.2024.1426231] [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: 05/01/2024] [Accepted: 06/14/2024] [Indexed: 08/21/2024] Open
Abstract
Multiple sclerosis (MS) is a frequently disabling neurological disorder characterized by symptoms, clinical signs and imaging abnormalities that typically fluctuate over time, affecting any level of the CNS. Prominent lymphocytic inflammation, many genetic susceptibility variants involving immune pathways, as well as potent responses of the neuroinflammatory component to immunomodulating drugs, have led to the natural conclusion that this disease is driven by a primary autoimmune process. In this Hypothesis and Theory article, we discuss emerging data that cast doubt on this assumption. After three decades of therapeutic experience, what has become clear is that potent immune modulators are highly effective at suppressing inflammatory relapses, yet exhibit very limited effects on the later progressive phase of MS. Moreover, neuropathological examination of MS tissue indicates that degeneration, CNS atrophy, and myelin loss are most prominent in the progressive stage, when lymphocytic inflammation paradoxically wanes. Finally, emerging clinical observations such as "progression independent of relapse activity" and "silent progression," now thought to take hold very early in the course, together argue that an underlying "cytodegenerative" process, likely targeting the myelinating unit, may in fact represent the most proximal step in a complex pathophysiological cascade exacerbated by an autoimmune inflammatory overlay. Parallels are drawn with more traditional neurodegenerative disorders, where a progressive proteopathy with prion-like propagation of toxic misfolded species is now known to play a key role. A potentially pivotal contribution of the Epstein-Barr virus and B cells in this process is also discussed.
Collapse
Affiliation(s)
- Peter K. Stys
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Shigeki Tsutsui
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Arie R. Gafson
- Biogen Digital Health, Biogen, Cambridge, MA, United States
| | - Bert A. ‘t Hart
- Department of Anatomy and Neurosciences, Amsterdam University Medical Centers (location VUmc), Amsterdam, Netherlands
| | - Shibeshih Belachew
- TheraPanacea, Paris, France
- Indivi (DBA of Healios AG), Basel, Switzerland
| | - Jeroen J. G. Geurts
- Department of Anatomy and Neurosciences, Amsterdam University Medical Centers (location VUmc), Amsterdam, Netherlands
| |
Collapse
|
2
|
Ciccarelli O, Barkhof F, Calabrese M, De Stefano N, Eshaghi A, Filippi M, Gasperini C, Granziera C, Kappos L, Rocca MA, Rovira À, Sastre-Garriga J, Sormani MP, Tur C, Toosy AT. Using the Progression Independent of Relapse Activity Framework to Unveil the Pathobiological Foundations of Multiple Sclerosis. Neurology 2024; 103:e209444. [PMID: 38889384 PMCID: PMC11226318 DOI: 10.1212/wnl.0000000000209444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 03/04/2024] [Indexed: 06/20/2024] Open
Abstract
Progression independent of relapse activity (PIRA), a recent concept to formalize disability accrual in multiple sclerosis (MS) independent of relapses, has gained popularity as a potential clinical trial outcome. We discuss its shortcomings and appraise the challenges of implementing it in clinical settings, experimental trials, and research. The current definition of PIRA assumes that acute inflammation, which can manifest as a relapse, and neurodegeneration, manifesting as progressive disability accrual, can be disentangled by introducing specific time windows between the onset of relapses and the observed increase in disability. The term PIRMA (progression independent of relapse and MRI activity) was recently introduced to indicate disability accrual in the absence of both clinical relapses and new brain and spinal cord MRI lesions. Assessing PIRMA in clinical practice is highly challenging because it necessitates frequent clinical assessments and brain and spinal cord MRI scans. PIRA is commonly assessed using Expanded Disability Status Scale, a scale heavily weighted toward motor disability, whereas a more granular assessment of disability deterioration, including cognitive decline, using composite measures or other tools, such as digital tools, would possess greater utility. Similarly, using PIRA as an outcome measure in randomized clinical trials is also challenging and requires methodological considerations. The underpinning pathobiology of disability accumulation, that is not associated with relapses, may encompass chronic active lesions (slowly expanding lesions and paramagnetic rim lesions), cortical lesions, brain and spinal cord atrophy, particularly in the gray matter, diffuse and focal microglial activation, persistent leptomeningeal enhancement, and white matter tract damage. We propose to use PIRA to understand the main determinant of disability accrual in observational, cohort studies, where regular MRI scans are not included, and introduce the term of "advanced-PIRMA" to investigate the contributions to disability accrual of the abovementioned processes, using conventional and advanced imaging. This is supported by the knowledge that MRI reflects the MS pathogenic mechanisms better than purely clinical descriptors. Any residual disability accrual, which remains unexplained after considering all these mechanisms with imaging, will highlight future research priorities to help complete our understanding of MS pathogenesis.
Collapse
Affiliation(s)
- Olga Ciccarelli
- From the Queen Square MS Centre (O.C., F.B., A.E., A.T.T.), Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London; National Institute for Health and Care Research (NIHR) (O.C.), University College London Hospitals (UCLH) Biomedical Research Centre; Centre for Medical Image Computing (F.B.), University College London, United Kingdom; Department of Radiology and Nuclear Medicine (F.B.), Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands; Department of Neurosciences, Biomedicine and Movement Sciences (M.C.), University of Verona; Department of Medicine, Surgery and Neuroscience (N.D.S.), University of Siena; Neuroimaging Research Unit (M.F., M.A.R.), Division of Neuroscience, and Neurology Unit (M.F., M.A.R.), Neurorehabilitation Unit, Neurophysiology Service, IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (M.F., M.A.R.), Milan; Department of Neuroscience (C. Gasperini), San Camillo Hospital, Rome, Italy; Translational Imaging in Neurology (ThINK) Basel (C. Granziera, L.K.), Department of Biomedical Engineering, Faculty of Medicine, University Hospital Basel and University of Basel; Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB) (C. Granziera, L.K.); University Hospital Basel and University of Basel (C. Granziera, L.K.), Switzerland; Section of Neuroradiology (À.R.), Department of Radiology, and Multiple Sclerosis Centre of Catalonia (J.S.-G., C.T.), Department of Neurology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Spain; Department of Health Sciences (M.P.S.), University of Genova; and IRCCS Ospedale Policlinico San Martino (M.P.S.), Genova, Italy
| | - Frederik Barkhof
- From the Queen Square MS Centre (O.C., F.B., A.E., A.T.T.), Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London; National Institute for Health and Care Research (NIHR) (O.C.), University College London Hospitals (UCLH) Biomedical Research Centre; Centre for Medical Image Computing (F.B.), University College London, United Kingdom; Department of Radiology and Nuclear Medicine (F.B.), Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands; Department of Neurosciences, Biomedicine and Movement Sciences (M.C.), University of Verona; Department of Medicine, Surgery and Neuroscience (N.D.S.), University of Siena; Neuroimaging Research Unit (M.F., M.A.R.), Division of Neuroscience, and Neurology Unit (M.F., M.A.R.), Neurorehabilitation Unit, Neurophysiology Service, IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (M.F., M.A.R.), Milan; Department of Neuroscience (C. Gasperini), San Camillo Hospital, Rome, Italy; Translational Imaging in Neurology (ThINK) Basel (C. Granziera, L.K.), Department of Biomedical Engineering, Faculty of Medicine, University Hospital Basel and University of Basel; Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB) (C. Granziera, L.K.); University Hospital Basel and University of Basel (C. Granziera, L.K.), Switzerland; Section of Neuroradiology (À.R.), Department of Radiology, and Multiple Sclerosis Centre of Catalonia (J.S.-G., C.T.), Department of Neurology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Spain; Department of Health Sciences (M.P.S.), University of Genova; and IRCCS Ospedale Policlinico San Martino (M.P.S.), Genova, Italy
| | - Massimiliano Calabrese
- From the Queen Square MS Centre (O.C., F.B., A.E., A.T.T.), Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London; National Institute for Health and Care Research (NIHR) (O.C.), University College London Hospitals (UCLH) Biomedical Research Centre; Centre for Medical Image Computing (F.B.), University College London, United Kingdom; Department of Radiology and Nuclear Medicine (F.B.), Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands; Department of Neurosciences, Biomedicine and Movement Sciences (M.C.), University of Verona; Department of Medicine, Surgery and Neuroscience (N.D.S.), University of Siena; Neuroimaging Research Unit (M.F., M.A.R.), Division of Neuroscience, and Neurology Unit (M.F., M.A.R.), Neurorehabilitation Unit, Neurophysiology Service, IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (M.F., M.A.R.), Milan; Department of Neuroscience (C. Gasperini), San Camillo Hospital, Rome, Italy; Translational Imaging in Neurology (ThINK) Basel (C. Granziera, L.K.), Department of Biomedical Engineering, Faculty of Medicine, University Hospital Basel and University of Basel; Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB) (C. Granziera, L.K.); University Hospital Basel and University of Basel (C. Granziera, L.K.), Switzerland; Section of Neuroradiology (À.R.), Department of Radiology, and Multiple Sclerosis Centre of Catalonia (J.S.-G., C.T.), Department of Neurology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Spain; Department of Health Sciences (M.P.S.), University of Genova; and IRCCS Ospedale Policlinico San Martino (M.P.S.), Genova, Italy
| | - Nicola De Stefano
- From the Queen Square MS Centre (O.C., F.B., A.E., A.T.T.), Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London; National Institute for Health and Care Research (NIHR) (O.C.), University College London Hospitals (UCLH) Biomedical Research Centre; Centre for Medical Image Computing (F.B.), University College London, United Kingdom; Department of Radiology and Nuclear Medicine (F.B.), Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands; Department of Neurosciences, Biomedicine and Movement Sciences (M.C.), University of Verona; Department of Medicine, Surgery and Neuroscience (N.D.S.), University of Siena; Neuroimaging Research Unit (M.F., M.A.R.), Division of Neuroscience, and Neurology Unit (M.F., M.A.R.), Neurorehabilitation Unit, Neurophysiology Service, IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (M.F., M.A.R.), Milan; Department of Neuroscience (C. Gasperini), San Camillo Hospital, Rome, Italy; Translational Imaging in Neurology (ThINK) Basel (C. Granziera, L.K.), Department of Biomedical Engineering, Faculty of Medicine, University Hospital Basel and University of Basel; Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB) (C. Granziera, L.K.); University Hospital Basel and University of Basel (C. Granziera, L.K.), Switzerland; Section of Neuroradiology (À.R.), Department of Radiology, and Multiple Sclerosis Centre of Catalonia (J.S.-G., C.T.), Department of Neurology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Spain; Department of Health Sciences (M.P.S.), University of Genova; and IRCCS Ospedale Policlinico San Martino (M.P.S.), Genova, Italy
| | - Arman Eshaghi
- From the Queen Square MS Centre (O.C., F.B., A.E., A.T.T.), Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London; National Institute for Health and Care Research (NIHR) (O.C.), University College London Hospitals (UCLH) Biomedical Research Centre; Centre for Medical Image Computing (F.B.), University College London, United Kingdom; Department of Radiology and Nuclear Medicine (F.B.), Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands; Department of Neurosciences, Biomedicine and Movement Sciences (M.C.), University of Verona; Department of Medicine, Surgery and Neuroscience (N.D.S.), University of Siena; Neuroimaging Research Unit (M.F., M.A.R.), Division of Neuroscience, and Neurology Unit (M.F., M.A.R.), Neurorehabilitation Unit, Neurophysiology Service, IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (M.F., M.A.R.), Milan; Department of Neuroscience (C. Gasperini), San Camillo Hospital, Rome, Italy; Translational Imaging in Neurology (ThINK) Basel (C. Granziera, L.K.), Department of Biomedical Engineering, Faculty of Medicine, University Hospital Basel and University of Basel; Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB) (C. Granziera, L.K.); University Hospital Basel and University of Basel (C. Granziera, L.K.), Switzerland; Section of Neuroradiology (À.R.), Department of Radiology, and Multiple Sclerosis Centre of Catalonia (J.S.-G., C.T.), Department of Neurology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Spain; Department of Health Sciences (M.P.S.), University of Genova; and IRCCS Ospedale Policlinico San Martino (M.P.S.), Genova, Italy
| | - Massimo Filippi
- From the Queen Square MS Centre (O.C., F.B., A.E., A.T.T.), Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London; National Institute for Health and Care Research (NIHR) (O.C.), University College London Hospitals (UCLH) Biomedical Research Centre; Centre for Medical Image Computing (F.B.), University College London, United Kingdom; Department of Radiology and Nuclear Medicine (F.B.), Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands; Department of Neurosciences, Biomedicine and Movement Sciences (M.C.), University of Verona; Department of Medicine, Surgery and Neuroscience (N.D.S.), University of Siena; Neuroimaging Research Unit (M.F., M.A.R.), Division of Neuroscience, and Neurology Unit (M.F., M.A.R.), Neurorehabilitation Unit, Neurophysiology Service, IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (M.F., M.A.R.), Milan; Department of Neuroscience (C. Gasperini), San Camillo Hospital, Rome, Italy; Translational Imaging in Neurology (ThINK) Basel (C. Granziera, L.K.), Department of Biomedical Engineering, Faculty of Medicine, University Hospital Basel and University of Basel; Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB) (C. Granziera, L.K.); University Hospital Basel and University of Basel (C. Granziera, L.K.), Switzerland; Section of Neuroradiology (À.R.), Department of Radiology, and Multiple Sclerosis Centre of Catalonia (J.S.-G., C.T.), Department of Neurology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Spain; Department of Health Sciences (M.P.S.), University of Genova; and IRCCS Ospedale Policlinico San Martino (M.P.S.), Genova, Italy
| | - Claudio Gasperini
- From the Queen Square MS Centre (O.C., F.B., A.E., A.T.T.), Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London; National Institute for Health and Care Research (NIHR) (O.C.), University College London Hospitals (UCLH) Biomedical Research Centre; Centre for Medical Image Computing (F.B.), University College London, United Kingdom; Department of Radiology and Nuclear Medicine (F.B.), Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands; Department of Neurosciences, Biomedicine and Movement Sciences (M.C.), University of Verona; Department of Medicine, Surgery and Neuroscience (N.D.S.), University of Siena; Neuroimaging Research Unit (M.F., M.A.R.), Division of Neuroscience, and Neurology Unit (M.F., M.A.R.), Neurorehabilitation Unit, Neurophysiology Service, IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (M.F., M.A.R.), Milan; Department of Neuroscience (C. Gasperini), San Camillo Hospital, Rome, Italy; Translational Imaging in Neurology (ThINK) Basel (C. Granziera, L.K.), Department of Biomedical Engineering, Faculty of Medicine, University Hospital Basel and University of Basel; Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB) (C. Granziera, L.K.); University Hospital Basel and University of Basel (C. Granziera, L.K.), Switzerland; Section of Neuroradiology (À.R.), Department of Radiology, and Multiple Sclerosis Centre of Catalonia (J.S.-G., C.T.), Department of Neurology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Spain; Department of Health Sciences (M.P.S.), University of Genova; and IRCCS Ospedale Policlinico San Martino (M.P.S.), Genova, Italy
| | - Cristina Granziera
- From the Queen Square MS Centre (O.C., F.B., A.E., A.T.T.), Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London; National Institute for Health and Care Research (NIHR) (O.C.), University College London Hospitals (UCLH) Biomedical Research Centre; Centre for Medical Image Computing (F.B.), University College London, United Kingdom; Department of Radiology and Nuclear Medicine (F.B.), Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands; Department of Neurosciences, Biomedicine and Movement Sciences (M.C.), University of Verona; Department of Medicine, Surgery and Neuroscience (N.D.S.), University of Siena; Neuroimaging Research Unit (M.F., M.A.R.), Division of Neuroscience, and Neurology Unit (M.F., M.A.R.), Neurorehabilitation Unit, Neurophysiology Service, IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (M.F., M.A.R.), Milan; Department of Neuroscience (C. Gasperini), San Camillo Hospital, Rome, Italy; Translational Imaging in Neurology (ThINK) Basel (C. Granziera, L.K.), Department of Biomedical Engineering, Faculty of Medicine, University Hospital Basel and University of Basel; Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB) (C. Granziera, L.K.); University Hospital Basel and University of Basel (C. Granziera, L.K.), Switzerland; Section of Neuroradiology (À.R.), Department of Radiology, and Multiple Sclerosis Centre of Catalonia (J.S.-G., C.T.), Department of Neurology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Spain; Department of Health Sciences (M.P.S.), University of Genova; and IRCCS Ospedale Policlinico San Martino (M.P.S.), Genova, Italy
| | - Ludwig Kappos
- From the Queen Square MS Centre (O.C., F.B., A.E., A.T.T.), Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London; National Institute for Health and Care Research (NIHR) (O.C.), University College London Hospitals (UCLH) Biomedical Research Centre; Centre for Medical Image Computing (F.B.), University College London, United Kingdom; Department of Radiology and Nuclear Medicine (F.B.), Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands; Department of Neurosciences, Biomedicine and Movement Sciences (M.C.), University of Verona; Department of Medicine, Surgery and Neuroscience (N.D.S.), University of Siena; Neuroimaging Research Unit (M.F., M.A.R.), Division of Neuroscience, and Neurology Unit (M.F., M.A.R.), Neurorehabilitation Unit, Neurophysiology Service, IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (M.F., M.A.R.), Milan; Department of Neuroscience (C. Gasperini), San Camillo Hospital, Rome, Italy; Translational Imaging in Neurology (ThINK) Basel (C. Granziera, L.K.), Department of Biomedical Engineering, Faculty of Medicine, University Hospital Basel and University of Basel; Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB) (C. Granziera, L.K.); University Hospital Basel and University of Basel (C. Granziera, L.K.), Switzerland; Section of Neuroradiology (À.R.), Department of Radiology, and Multiple Sclerosis Centre of Catalonia (J.S.-G., C.T.), Department of Neurology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Spain; Department of Health Sciences (M.P.S.), University of Genova; and IRCCS Ospedale Policlinico San Martino (M.P.S.), Genova, Italy
| | - Maria A Rocca
- From the Queen Square MS Centre (O.C., F.B., A.E., A.T.T.), Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London; National Institute for Health and Care Research (NIHR) (O.C.), University College London Hospitals (UCLH) Biomedical Research Centre; Centre for Medical Image Computing (F.B.), University College London, United Kingdom; Department of Radiology and Nuclear Medicine (F.B.), Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands; Department of Neurosciences, Biomedicine and Movement Sciences (M.C.), University of Verona; Department of Medicine, Surgery and Neuroscience (N.D.S.), University of Siena; Neuroimaging Research Unit (M.F., M.A.R.), Division of Neuroscience, and Neurology Unit (M.F., M.A.R.), Neurorehabilitation Unit, Neurophysiology Service, IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (M.F., M.A.R.), Milan; Department of Neuroscience (C. Gasperini), San Camillo Hospital, Rome, Italy; Translational Imaging in Neurology (ThINK) Basel (C. Granziera, L.K.), Department of Biomedical Engineering, Faculty of Medicine, University Hospital Basel and University of Basel; Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB) (C. Granziera, L.K.); University Hospital Basel and University of Basel (C. Granziera, L.K.), Switzerland; Section of Neuroradiology (À.R.), Department of Radiology, and Multiple Sclerosis Centre of Catalonia (J.S.-G., C.T.), Department of Neurology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Spain; Department of Health Sciences (M.P.S.), University of Genova; and IRCCS Ospedale Policlinico San Martino (M.P.S.), Genova, Italy
| | - Àlex Rovira
- From the Queen Square MS Centre (O.C., F.B., A.E., A.T.T.), Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London; National Institute for Health and Care Research (NIHR) (O.C.), University College London Hospitals (UCLH) Biomedical Research Centre; Centre for Medical Image Computing (F.B.), University College London, United Kingdom; Department of Radiology and Nuclear Medicine (F.B.), Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands; Department of Neurosciences, Biomedicine and Movement Sciences (M.C.), University of Verona; Department of Medicine, Surgery and Neuroscience (N.D.S.), University of Siena; Neuroimaging Research Unit (M.F., M.A.R.), Division of Neuroscience, and Neurology Unit (M.F., M.A.R.), Neurorehabilitation Unit, Neurophysiology Service, IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (M.F., M.A.R.), Milan; Department of Neuroscience (C. Gasperini), San Camillo Hospital, Rome, Italy; Translational Imaging in Neurology (ThINK) Basel (C. Granziera, L.K.), Department of Biomedical Engineering, Faculty of Medicine, University Hospital Basel and University of Basel; Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB) (C. Granziera, L.K.); University Hospital Basel and University of Basel (C. Granziera, L.K.), Switzerland; Section of Neuroradiology (À.R.), Department of Radiology, and Multiple Sclerosis Centre of Catalonia (J.S.-G., C.T.), Department of Neurology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Spain; Department of Health Sciences (M.P.S.), University of Genova; and IRCCS Ospedale Policlinico San Martino (M.P.S.), Genova, Italy
| | - Jaume Sastre-Garriga
- From the Queen Square MS Centre (O.C., F.B., A.E., A.T.T.), Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London; National Institute for Health and Care Research (NIHR) (O.C.), University College London Hospitals (UCLH) Biomedical Research Centre; Centre for Medical Image Computing (F.B.), University College London, United Kingdom; Department of Radiology and Nuclear Medicine (F.B.), Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands; Department of Neurosciences, Biomedicine and Movement Sciences (M.C.), University of Verona; Department of Medicine, Surgery and Neuroscience (N.D.S.), University of Siena; Neuroimaging Research Unit (M.F., M.A.R.), Division of Neuroscience, and Neurology Unit (M.F., M.A.R.), Neurorehabilitation Unit, Neurophysiology Service, IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (M.F., M.A.R.), Milan; Department of Neuroscience (C. Gasperini), San Camillo Hospital, Rome, Italy; Translational Imaging in Neurology (ThINK) Basel (C. Granziera, L.K.), Department of Biomedical Engineering, Faculty of Medicine, University Hospital Basel and University of Basel; Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB) (C. Granziera, L.K.); University Hospital Basel and University of Basel (C. Granziera, L.K.), Switzerland; Section of Neuroradiology (À.R.), Department of Radiology, and Multiple Sclerosis Centre of Catalonia (J.S.-G., C.T.), Department of Neurology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Spain; Department of Health Sciences (M.P.S.), University of Genova; and IRCCS Ospedale Policlinico San Martino (M.P.S.), Genova, Italy
| | - Maria Pia Sormani
- From the Queen Square MS Centre (O.C., F.B., A.E., A.T.T.), Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London; National Institute for Health and Care Research (NIHR) (O.C.), University College London Hospitals (UCLH) Biomedical Research Centre; Centre for Medical Image Computing (F.B.), University College London, United Kingdom; Department of Radiology and Nuclear Medicine (F.B.), Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands; Department of Neurosciences, Biomedicine and Movement Sciences (M.C.), University of Verona; Department of Medicine, Surgery and Neuroscience (N.D.S.), University of Siena; Neuroimaging Research Unit (M.F., M.A.R.), Division of Neuroscience, and Neurology Unit (M.F., M.A.R.), Neurorehabilitation Unit, Neurophysiology Service, IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (M.F., M.A.R.), Milan; Department of Neuroscience (C. Gasperini), San Camillo Hospital, Rome, Italy; Translational Imaging in Neurology (ThINK) Basel (C. Granziera, L.K.), Department of Biomedical Engineering, Faculty of Medicine, University Hospital Basel and University of Basel; Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB) (C. Granziera, L.K.); University Hospital Basel and University of Basel (C. Granziera, L.K.), Switzerland; Section of Neuroradiology (À.R.), Department of Radiology, and Multiple Sclerosis Centre of Catalonia (J.S.-G., C.T.), Department of Neurology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Spain; Department of Health Sciences (M.P.S.), University of Genova; and IRCCS Ospedale Policlinico San Martino (M.P.S.), Genova, Italy
| | - Carmen Tur
- From the Queen Square MS Centre (O.C., F.B., A.E., A.T.T.), Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London; National Institute for Health and Care Research (NIHR) (O.C.), University College London Hospitals (UCLH) Biomedical Research Centre; Centre for Medical Image Computing (F.B.), University College London, United Kingdom; Department of Radiology and Nuclear Medicine (F.B.), Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands; Department of Neurosciences, Biomedicine and Movement Sciences (M.C.), University of Verona; Department of Medicine, Surgery and Neuroscience (N.D.S.), University of Siena; Neuroimaging Research Unit (M.F., M.A.R.), Division of Neuroscience, and Neurology Unit (M.F., M.A.R.), Neurorehabilitation Unit, Neurophysiology Service, IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (M.F., M.A.R.), Milan; Department of Neuroscience (C. Gasperini), San Camillo Hospital, Rome, Italy; Translational Imaging in Neurology (ThINK) Basel (C. Granziera, L.K.), Department of Biomedical Engineering, Faculty of Medicine, University Hospital Basel and University of Basel; Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB) (C. Granziera, L.K.); University Hospital Basel and University of Basel (C. Granziera, L.K.), Switzerland; Section of Neuroradiology (À.R.), Department of Radiology, and Multiple Sclerosis Centre of Catalonia (J.S.-G., C.T.), Department of Neurology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Spain; Department of Health Sciences (M.P.S.), University of Genova; and IRCCS Ospedale Policlinico San Martino (M.P.S.), Genova, Italy
| | - Ahmed T Toosy
- From the Queen Square MS Centre (O.C., F.B., A.E., A.T.T.), Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London; National Institute for Health and Care Research (NIHR) (O.C.), University College London Hospitals (UCLH) Biomedical Research Centre; Centre for Medical Image Computing (F.B.), University College London, United Kingdom; Department of Radiology and Nuclear Medicine (F.B.), Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands; Department of Neurosciences, Biomedicine and Movement Sciences (M.C.), University of Verona; Department of Medicine, Surgery and Neuroscience (N.D.S.), University of Siena; Neuroimaging Research Unit (M.F., M.A.R.), Division of Neuroscience, and Neurology Unit (M.F., M.A.R.), Neurorehabilitation Unit, Neurophysiology Service, IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (M.F., M.A.R.), Milan; Department of Neuroscience (C. Gasperini), San Camillo Hospital, Rome, Italy; Translational Imaging in Neurology (ThINK) Basel (C. Granziera, L.K.), Department of Biomedical Engineering, Faculty of Medicine, University Hospital Basel and University of Basel; Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB) (C. Granziera, L.K.); University Hospital Basel and University of Basel (C. Granziera, L.K.), Switzerland; Section of Neuroradiology (À.R.), Department of Radiology, and Multiple Sclerosis Centre of Catalonia (J.S.-G., C.T.), Department of Neurology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Spain; Department of Health Sciences (M.P.S.), University of Genova; and IRCCS Ospedale Policlinico San Martino (M.P.S.), Genova, Italy
| |
Collapse
|
3
|
Calabrese M, Preziosa P, Scalfari A, Colato E, Marastoni D, Absinta M, Battaglini M, De Stefano N, Di Filippo M, Hametner S, Howell OW, Inglese M, Lassmann H, Martin R, Nicholas R, Reynolds R, Rocca MA, Tamanti A, Vercellino M, Villar LM, Filippi M, Magliozzi R. Determinants and Biomarkers of Progression Independent of Relapses in Multiple Sclerosis. Ann Neurol 2024; 96:1-20. [PMID: 38568026 DOI: 10.1002/ana.26913] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 01/04/2024] [Accepted: 02/15/2024] [Indexed: 06/20/2024]
Abstract
Clinical, pathological, and imaging evidence in multiple sclerosis (MS) suggests that a smoldering inflammatory activity is present from the earliest stages of the disease and underlies the progression of disability, which proceeds relentlessly and independently of clinical and radiological relapses (PIRA). The complex system of pathological events driving "chronic" worsening is likely linked with the early accumulation of compartmentalized inflammation within the central nervous system as well as insufficient repair phenomena and mitochondrial failure. These mechanisms are partially lesion-independent and differ from those causing clinical relapses and the formation of new focal demyelinating lesions; they lead to neuroaxonal dysfunction and death, myelin loss, glia alterations, and finally, a neuronal network dysfunction outweighing central nervous system (CNS) compensatory mechanisms. This review aims to provide an overview of the state of the art of neuropathological, immunological, and imaging knowledge about the mechanisms underlying the smoldering disease activity, focusing on possible early biomarkers and their translation into clinical practice. ANN NEUROL 2024;96:1-20.
Collapse
Affiliation(s)
- Massimiliano Calabrese
- Department of Neurosciences and Biomedicine and Movement, The Multiple Sclerosis Center of University Hospital of Verona, Verona, Italy
| | - Paolo Preziosa
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Antonio Scalfari
- Centre of Neuroscience, Department of Medicine, Imperial College, London, UK
| | - Elisa Colato
- Department of Neurosciences and Biomedicine and Movement, The Multiple Sclerosis Center of University Hospital of Verona, Verona, Italy
| | - Damiano Marastoni
- Department of Neurosciences and Biomedicine and Movement, The Multiple Sclerosis Center of University Hospital of Verona, Verona, Italy
| | - Martina Absinta
- Translational Neuropathology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marco Battaglini
- Siena Imaging S.r.l., Siena, Italy
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Nicola De Stefano
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Massimiliano Di Filippo
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Simon Hametner
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Owain W Howell
- Institute of Life Sciences, Swansea University Medical School, Swansea, UK
| | - Matilde Inglese
- Dipartimento di neuroscienze, riabilitazione, oftalmologia, genetica e scienze materno-infantili - DINOGMI, University of Genova, Genoa, Italy
| | - Hans Lassmann
- Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Roland Martin
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
- Therapeutic Design Unit, Center for Molecular Medicine, Department of Clinical Neurosciences, Karolinska Institutet, Stockholm, Sweden
- Cellerys AG, Schlieren, Switzerland
| | - Richard Nicholas
- Department of Brain Sciences, Faculty of Medicine, Burlington Danes, Imperial College London, London, UK
| | - Richard Reynolds
- Division of Neuroscience, Department of Brain Sciences, Imperial College London, London, UK
| | - Maria A Rocca
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Agnese Tamanti
- Department of Neurosciences and Biomedicine and Movement, The Multiple Sclerosis Center of University Hospital of Verona, Verona, Italy
| | - Marco Vercellino
- Multiple Sclerosis Center & Neurologia I U, Department of Neuroscience, University Hospital AOU Città della Salute e della Scienza di Torino, Turin, Italy
| | - Luisa Maria Villar
- Department of Immunology, Ramon y Cajal University Hospital. IRYCIS. REI, Madrid, Spain
| | - Massimo Filippi
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
- Neurorehabilitation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurophysiology Service, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Roberta Magliozzi
- Department of Neurosciences and Biomedicine and Movement, The Multiple Sclerosis Center of University Hospital of Verona, Verona, Italy
| |
Collapse
|
4
|
Luchicchi A, Muñoz‐Gonzalez G, Halperin ST, Strijbis E, van Dijk LHM, Foutiadou C, Uriac F, Bouman PM, Schouten MAN, Plemel J, 't Hart BA, Geurts JJG, Schenk GJ. Micro-diffusely abnormal white matter: An early multiple sclerosis lesion phase with intensified myelin blistering. Ann Clin Transl Neurol 2024; 11:973-988. [PMID: 38425098 PMCID: PMC11021636 DOI: 10.1002/acn3.52015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/03/2024] [Accepted: 01/25/2024] [Indexed: 03/02/2024] Open
Abstract
OBJECTIVE Multiple sclerosis (MS) is a chronic central nervous system disease whose white matter lesion origin remains debated. Recently, we reported subtle changes in the MS normal appearing white matter (NAWM), presenting with an increase in myelin blisters and myelin protein citrullination, which may recapitulate some of the prodromal degenerative processes involved in MS pathogenesis. Here, to clarify the relevance of these changes for subsequent MS myelin degeneration we explored their prevalence in WM regions characterized by subtly reduced myelination (dubbed as micro-diffusely abnormal white matter, mDAWM). METHODS We used an in-depth (immuno)histochemistry approach in 27 MS donors with histological presence of mDAWM and 5 controls. An antibody panel against degenerative markers was combined and the presence of myelin/axonal aberrations was analyzed and compared with the NAWM from the same cases/slices/regions. RESULTS mDAWM-defined areas exhibit ill-defined borders, no signs of Wallerian degeneration, and they associate with visible veins. Remarkably, such areas present with augmented myelin blister frequency, enhanced prevalence of polar myelin phospholipids, citrullination, and degradation of myelin basic protein (MBP) when compared with the NAWM. Furthermore, enhanced reactivity of microglia/macrophages against citrullinated MBP was also observed in this tissue. INTERPRETATION We report a new histologically defined early phase in MS lesion formation, namely mDAWM, which lacks signs of Wallerian pathology. These results support the prelesional nature of the mDAWM. We conceptualize that evolution to pathologically evident lesions comprises the previously documented imbalance of axo-myelinic units (myelin blistering) leading to their degeneration and immune system activation by released myelin components.
Collapse
Affiliation(s)
- Antonio Luchicchi
- Department of Anatomy and NeurosciencesAmsterdam University Medical Centers, location VU Medical Center, Amsterdam NeuroscienceAmsterdamthe Netherlands
- MS Centrum Amsterdam, Amsterdam University Medical Centers, location VU Medical CenterAmsterdamthe Netherlands
| | - Gema Muñoz‐Gonzalez
- Department of Anatomy and NeurosciencesAmsterdam University Medical Centers, location VU Medical Center, Amsterdam NeuroscienceAmsterdamthe Netherlands
- MS Centrum Amsterdam, Amsterdam University Medical Centers, location VU Medical CenterAmsterdamthe Netherlands
| | - Saar T. Halperin
- Department of Anatomy and NeurosciencesAmsterdam University Medical Centers, location VU Medical Center, Amsterdam NeuroscienceAmsterdamthe Netherlands
- MS Centrum Amsterdam, Amsterdam University Medical Centers, location VU Medical CenterAmsterdamthe Netherlands
| | - Eva Strijbis
- MS Centrum Amsterdam, Amsterdam University Medical Centers, location VU Medical CenterAmsterdamthe Netherlands
- Department of NeurologyAmsterdam University Medical Centers, location VU Medical CenterAmsterdamthe Netherlands
| | - Laura H. M. van Dijk
- Department of Anatomy and NeurosciencesAmsterdam University Medical Centers, location VU Medical Center, Amsterdam NeuroscienceAmsterdamthe Netherlands
| | - Chrisa Foutiadou
- Department of Anatomy and NeurosciencesAmsterdam University Medical Centers, location VU Medical Center, Amsterdam NeuroscienceAmsterdamthe Netherlands
| | - Florence Uriac
- Department of Anatomy and NeurosciencesAmsterdam University Medical Centers, location VU Medical Center, Amsterdam NeuroscienceAmsterdamthe Netherlands
| | - Piet M. Bouman
- Department of Anatomy and NeurosciencesAmsterdam University Medical Centers, location VU Medical Center, Amsterdam NeuroscienceAmsterdamthe Netherlands
- MS Centrum Amsterdam, Amsterdam University Medical Centers, location VU Medical CenterAmsterdamthe Netherlands
| | - Maxime A. N. Schouten
- Department of Anatomy and NeurosciencesAmsterdam University Medical Centers, location VU Medical Center, Amsterdam NeuroscienceAmsterdamthe Netherlands
- MS Centrum Amsterdam, Amsterdam University Medical Centers, location VU Medical CenterAmsterdamthe Netherlands
| | - Jason Plemel
- Department of NeuroscienceUniversity of AlbertaEdmontonAlbertaCanada
| | - Bert A. 't Hart
- Department of Anatomy and NeurosciencesAmsterdam University Medical Centers, location VU Medical Center, Amsterdam NeuroscienceAmsterdamthe Netherlands
- MS Centrum Amsterdam, Amsterdam University Medical Centers, location VU Medical CenterAmsterdamthe Netherlands
| | - Jeroen J. G. Geurts
- Department of Anatomy and NeurosciencesAmsterdam University Medical Centers, location VU Medical Center, Amsterdam NeuroscienceAmsterdamthe Netherlands
- MS Centrum Amsterdam, Amsterdam University Medical Centers, location VU Medical CenterAmsterdamthe Netherlands
| | - Geert J. Schenk
- Department of Anatomy and NeurosciencesAmsterdam University Medical Centers, location VU Medical Center, Amsterdam NeuroscienceAmsterdamthe Netherlands
- MS Centrum Amsterdam, Amsterdam University Medical Centers, location VU Medical CenterAmsterdamthe Netherlands
| |
Collapse
|
5
|
Lee HH, Tian Q, Sheft M, Coronado-Leija R, Ramos-Llorden G, Abdollahzadeh A, Fieremans E, Novikov DS, Huang SY. The effects of axonal beading and undulation on axonal diameter estimation from diffusion MRI: Insights from simulations in human axons segmented from three-dimensional electron microscopy. NMR IN BIOMEDICINE 2024; 37:e5087. [PMID: 38168082 PMCID: PMC10942763 DOI: 10.1002/nbm.5087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 11/19/2023] [Accepted: 11/21/2023] [Indexed: 01/05/2024]
Abstract
The increasing availability of high-performance gradient systems in human MRI scanners has generated great interest in diffusion microstructural imaging applications such as axonal diameter mapping. Practically, sensitivity to axon diameter in diffusion MRI is attained at strong diffusion weightings b , where the deviation from the expected 1 / b scaling in white matter yields a finite transverse diffusivity, which is then translated into an axon diameter estimate. While axons are usually modeled as perfectly straight, impermeable cylinders, local variations in diameter (caliber variation or beading) and direction (undulation) are known to influence axonal diameter estimates and have been observed in microscopy data of human axons. In this study, we performed Monte Carlo simulations of diffusion in axons reconstructed from three-dimensional electron microscopy of a human temporal lobe specimen using simulated sequence parameters matched to the maximal gradient strength of the next-generation Connectome 2.0 human MRI scanner ( ≲ 500 mT/m). We show that axon diameter estimation is accurate for nonbeaded, nonundulating fibers; however, in fibers with caliber variations and undulations, the axon diameter is heavily underestimated due to caliber variations, and this effect overshadows the known overestimation of the axon diameter due to undulations. This unexpected underestimation may originate from variations in the coarse-grained axial diffusivity due to caliber variations. Given that increased axonal beading and undulations have been observed in pathological tissues, such as traumatic brain injury and ischemia, the interpretation of axon diameter alterations in pathology may be significantly confounded.
Collapse
Affiliation(s)
- Hong-Hsi Lee
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Qiyuan Tian
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Maxina Sheft
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts, USA
- Harvard–MIT Health Sciences and Technology, Cambridge, Massachusetts, USA
| | - Ricardo Coronado-Leija
- Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, New York, USA
- Center for Advanced Imaging Innovation and Research (CAI2R), New York University School of Medicine, New York, New York, USA
| | - Gabriel Ramos-Llorden
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Ali Abdollahzadeh
- Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, New York, USA
- Center for Advanced Imaging Innovation and Research (CAI2R), New York University School of Medicine, New York, New York, USA
| | - Els Fieremans
- Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, New York, USA
- Center for Advanced Imaging Innovation and Research (CAI2R), New York University School of Medicine, New York, New York, USA
| | - Dmitry S. Novikov
- Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, New York, USA
- Center for Advanced Imaging Innovation and Research (CAI2R), New York University School of Medicine, New York, New York, USA
| | - Susie Y. Huang
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
6
|
Motegi H, Kufukihara K, Kitagawa S, Sekiguchi K, Hata J, Fujiwara H, Jinzaki M, Okano H, Nakamura M, Iguchi Y, Nakahara J. Non-lesional white matter changes depicted by q-space diffusional MRI correlate with clinical disabilities in multiple sclerosis. J Neurol Sci 2024; 456:122851. [PMID: 38181653 DOI: 10.1016/j.jns.2023.122851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 11/20/2023] [Accepted: 12/17/2023] [Indexed: 01/07/2024]
Abstract
BACKGROUND We previously developed an optimized q-space diffusional MRI technique (normalized leptokurtic diffusion [NLD] map) to delineate the demyelinated lesions of multiple sclerosis (MS) patients. Herein, we evaluated the utility of NLD maps to discern the white matter abnormalities in normal-appearing white matter (NAWM) and the abnormalities' possible associations with physical and cognitive disabilities in MS. METHODS We conducted a retrospective observational study of MS patients treated at our hospital (Jan. 2012 to Dec. 2022). Clinical and MRI data were collected; Processing Speed Test (PST) data were obtained when possible. For a quantitative analysis of the NLD maps, we calculated the NLD index as GVROI/GVREF, where GV is a mean grayscale value in the regions of interest (ROIs) and the reference area (REF; cerebrospinal fluid). RESULTS One hundred-one individuals with MS were included. The lower corpus callosum and non-lesional WM NLD index were associated with worse Expanded Disability Status Scale (EDSS) and PST scores. The NLD indexes in the corpus callosum (p < 0.0001) and non-lesional white matter (p < 0.0001) were significantly reduced in progressive MS compared to relapsing-remitting MS. We categorized MS severity as moderate/severe (EDSS score ≥ 4 points) and mild (EDSS score < 4 points). The NLD indexes in the corpus callosum (p < 0.0001) and non-lesional white matter (p < 0.0001) were significantly lower in the moderate/severe MS group compared to the mild MS group. CONCLUSION The NLD map revealed abnormalities in the non-lesional white matter, providing valuable insights for evaluating manifestations in MS patients.
Collapse
Affiliation(s)
- Haruhiko Motegi
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan; Department of Neurology, The Jikei University School of Medicine, Tokyo, Japan.
| | - Kenji Kufukihara
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan; Department of Neurology, National Hospital Organization Tokyo Medical Center, Tokyo, Japan.
| | - Satoshi Kitagawa
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan.
| | - Koji Sekiguchi
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan.
| | - Junichi Hata
- Graduate School of Human Health Sciences, Tokyo Metropolitan University, Tokyo, Japan; Department of Physiology, Keio University School of Medicine, Tokyo, Japan; Laboratory for Marmoset Neural Architecture, RIKEN Center for Brain Science, Wako, Japan.
| | - Hirokazu Fujiwara
- Center of Preventive Medicine, Keio University School of Medicine, Tokyo, Japan.
| | - Masahiro Jinzaki
- Department of Radiology, Keio University School of Medicine, Tokyo, Japan.
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan; Laboratory for Marmoset Neural Architecture, RIKEN Center for Brain Science, Wako, Japan.
| | - Masaya Nakamura
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo, Japan.
| | - Yasuyuki Iguchi
- Department of Neurology, The Jikei University School of Medicine, Tokyo, Japan.
| | - Jin Nakahara
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan.
| |
Collapse
|
7
|
Cerdán Cerdá A, Toschi N, Treaba CA, Barletta V, Herranz E, Mehndiratta A, Gomez-Sanchez JA, Mainero C, De Santis S. A translational MRI approach to validate acute axonal damage detection as an early event in multiple sclerosis. eLife 2024; 13:e79169. [PMID: 38192199 PMCID: PMC10776086 DOI: 10.7554/elife.79169] [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: 04/01/2022] [Accepted: 12/05/2023] [Indexed: 01/10/2024] Open
Abstract
Axonal degeneration is a central pathological feature of multiple sclerosis and is closely associated with irreversible clinical disability. Current noninvasive methods to detect axonal damage in vivo are limited in their specificity and clinical applicability, and by the lack of proper validation. We aimed to validate an MRI framework based on multicompartment modeling of the diffusion signal (AxCaliber) in rats in the presence of axonal pathology, achieved through injection of a neurotoxin damaging the neuronal terminal of axons. We then applied the same MRI protocol to map axonal integrity in the brain of multiple sclerosis relapsing-remitting patients and age-matched healthy controls. AxCaliber is sensitive to acute axonal damage in rats, as demonstrated by a significant increase in the mean axonal caliber along the targeted tract, which correlated with neurofilament staining. Electron microscopy confirmed that increased mean axonal diameter is associated with acute axonal pathology. In humans with multiple sclerosis, we uncovered a diffuse increase in mean axonal caliber in most areas of the normal-appearing white matter, preferentially affecting patients with short disease duration. Our results demonstrate that MRI-based axonal diameter mapping is a sensitive and specific imaging biomarker that links noninvasive imaging contrasts with the underlying biological substrate, uncovering generalized axonal damage in multiple sclerosis as an early event.
Collapse
Affiliation(s)
| | - Nicola Toschi
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical SchoolBostonUnited States
- Department of Biomedicine and Prevention, University of Rome Tor VergataRomeItaly
| | - Constantina A Treaba
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical SchoolBostonUnited States
| | - Valeria Barletta
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical SchoolBostonUnited States
| | - Elena Herranz
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical SchoolBostonUnited States
| | - Ambica Mehndiratta
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical SchoolBostonUnited States
| | - Jose A Gomez-Sanchez
- Instituto de Neurociencias de Alicante, CSIC-UMHSan Juan de AlicanteSpain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL)AlicanteSpain
- Millennium Nucleus for the Study of Pain (MiNuSPain)SantiagoChile
| | - Caterina Mainero
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical SchoolBostonUnited States
| | - Silvia De Santis
- Instituto de Neurociencias de Alicante, CSIC-UMHSan Juan de AlicanteSpain
| |
Collapse
|
8
|
Sarkar SK, Willson AML, Jordan MA. The Plasticity of Immune Cell Response Complicates Dissecting the Underlying Pathology of Multiple Sclerosis. J Immunol Res 2024; 2024:5383099. [PMID: 38213874 PMCID: PMC10783990 DOI: 10.1155/2024/5383099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 12/05/2023] [Accepted: 12/11/2023] [Indexed: 01/13/2024] Open
Abstract
Multiple sclerosis (MS) is a neurodegenerative autoimmune disease characterized by the destruction of the myelin sheath of the neuronal axon in the central nervous system. Many risk factors, including environmental, epigenetic, genetic, and lifestyle factors, are responsible for the development of MS. It has long been thought that only adaptive immune cells, especially autoreactive T cells, are responsible for the pathophysiology; however, recent evidence has indicated that innate immune cells are also highly involved in disease initiation and progression. Here, we compile the available data regarding the role immune cells play in MS, drawn from both human and animal research. While T and B lymphocytes, chiefly enhance MS pathology, regulatory T cells (Tregs) may serve a more protective role, as can B cells, depending on context and location. Cells chiefly involved in innate immunity, including macrophages, microglia, astrocytes, dendritic cells, natural killer (NK) cells, eosinophils, and mast cells, play varied roles. In addition, there is evidence regarding the involvement of innate-like immune cells, such as γδ T cells, NKT cells, MAIT cells, and innate-like B cells as crucial contributors to MS pathophysiology. It is unclear which of these cell subsets are involved in the onset or progression of disease or in protective mechanisms due to their plastic nature, which can change their properties and functions depending on microenvironmental exposure and the response of neural networks in damage control. This highlights the need for a multipronged approach, combining stringently designed clinical data with carefully controlled in vitro and in vivo research findings, to identify the underlying mechanisms so that more effective therapeutics can be developed.
Collapse
Affiliation(s)
- Sujan Kumar Sarkar
- Department of Anatomy, Histology and Physiology, Faculty of Animal Science and Veterinary Medicine, Sher-e-Bangla Agricultural University, Dhaka, Bangladesh
| | - Annie M. L. Willson
- Biomedical Sciences and Molecular Biology, CPHMVS, James Cook University, Townsville, Queensland 4811, Australia
| | - Margaret A. Jordan
- Biomedical Sciences and Molecular Biology, CPHMVS, James Cook University, Townsville, Queensland 4811, Australia
| |
Collapse
|
9
|
Chen X, Zhang J, Shen LS, Chen YP, Yang JQ, Tang WJ, Guo RM. Bibliometric analysis of myelin imaging studies of patients with multiple sclerosis (2000-2022). Quant Imaging Med Surg 2024; 14:837-851. [PMID: 38223029 PMCID: PMC10784065 DOI: 10.21037/qims-23-1157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 11/10/2023] [Indexed: 01/16/2024]
Abstract
Background Multiple sclerosis (MS) is a condition that can impact the central nervous system (CNS) and cause damage to the myelin, which is responsible for facilitating the normal transmission of electrical impulses along the nerves. We performed a bibliometric analysis of the scientific publications on myelin imaging in MS to reveal the development trends in this field and to evaluate research trends in myelin imaging in MS. Methods The Web of Science Core Collection was searched for articles related to myelin imaging in MS published between January 2000 and December 2022. CiteSpace, VOSviewer, and R language were used to evaluate and visualize contributions by and co-occurrence relationships among countries and institutions, authors, journals, citations, keywords, and so on. Results A total of 1,639 articles addressed the topic of myelin imaging in MS. The United States had the largest number of annual publications. The University of London was the institution with the highest number of publications (n=118) and citations (n=9,885). The top 3 productive authors were all from the University of British Columbia in Canada. An article published by Mackay et al. in 1994 had the most citations (n=272). Neuroimage [impact factor (IF) =7.40, Journal Citation Reports quartile 1 (Q1)] was the most productive journal in terms of the number of articles relating to myelin imaging in MS (n=149). In recent years, myelin water imaging, synthetic magnetic resonance imaging (SyMRI), inhomogeneous magnetization, positron emission tomography (PET) imaging, and aquaporin-4 (AQP4) have been researched hotspots of myelin imaging in MS. Conclusions With advancements in the pathophysiological research on myelin changes in MS, myelin imaging is playing an important role in the diagnosis and treatment of MS. In addition, the use of new sequences of myelin imaging to distinguish MS from other inflammatory demyelinating diseases is a future development trend in this field.
Collapse
Affiliation(s)
| | | | - Li-Shan Shen
- Department of Radiology, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yao-Ping Chen
- Department of Radiology, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jin-Quan Yang
- Department of Radiology, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | | | | |
Collapse
|
10
|
Dejbakht M, Akhzari M, Jalili S, Faraji F, Barazesh M. Multiple Sclerosis: New Insights into Molecular Pathogenesis and Novel Platforms for Disease Treatment. Curr Drug Res Rev 2024; 16:175-197. [PMID: 37724675 DOI: 10.2174/2589977516666230915103730] [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: 01/08/2023] [Revised: 06/23/2023] [Accepted: 07/03/2023] [Indexed: 09/21/2023]
Abstract
BACKGROUND Multiple sclerosis (MS), a chronic inflammatory disorder, affects the central nervous system via myelin degradation. The cause of MS is not fully known, but during recent years, our knowledge has deepened significantly regarding the different aspects of MS, including etiology, molecular pathophysiology, diagnosis and therapeutic options. Myelin basic protein (MBP) is the main myelin protein that accounts for maintaining the stability of the myelin sheath. Recent evidence has revealed that MBP citrullination or deamination, which is catalyzed by Ca2+ dependent peptidyl arginine deiminase (PAD) enzyme leads to the reduction of positive charge, and subsequently proteolytic cleavage of MBP. The overexpression of PAD2 in the brains of MS patients plays an essential role in new epitope formation and progression of the autoimmune disorder. Some drugs have recently entered phase III clinical trials with promising efficacy and will probably obtain approval in the near future. As different therapeutic platforms develop, finding an optimal treatment for each individual patient will be more challenging. AIMS This review provides a comprehensive insight into MS with a focus on its pathogenesis and recent advances in diagnostic methods and its present and upcoming treatment modalities. CONCLUSION MS therapy alters quickly as research findings and therapeutic options surrounding MS expand. McDonald's guidelines have created different criteria for MS diagnosis. In recent years, ever-growing interest in the development of PAD inhibitors has led to the generation of many reversible and irreversible PAD inhibitors against the disease with satisfactory therapeutic outcomes.
Collapse
Affiliation(s)
- Majid Dejbakht
- Department of Cellular and Molecular Research Center, Gerash University of Medical Sciences, Gerash, Iran
| | - Morteza Akhzari
- School of Nursing, Larestan University of Medical Sciences, Larestan, Iran
| | - Sajad Jalili
- Department of Orthopedics, School of Medicine, Ahvaz Jundishapour University of Medical Sciences, Ahvaz, Iran
| | - Fouziyeh Faraji
- Department of Cellular and Molecular Research Center, Gerash University of Medical Sciences, Gerash, Iran
| | - Mahdi Barazesh
- Department of Biotechnology, Cellular and Molecular Research Center, School of Paramedical, Gerash University of Medical Sciences, Gerash, Iran
| |
Collapse
|
11
|
Kessler W, Thomas C, Kuhlmann T. Microglia activation in periplaque white matter in multiple sclerosis depends on age and lesion type, but does not correlate with oligodendroglial loss. Acta Neuropathol 2023; 146:817-828. [PMID: 37897549 PMCID: PMC10628007 DOI: 10.1007/s00401-023-02645-2] [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: 04/14/2023] [Revised: 10/05/2023] [Accepted: 10/05/2023] [Indexed: 10/30/2023]
Abstract
Multiple sclerosis (MS) is the most frequent inflammatory and demyelinating disease of the CNS. The disease course in MS is highly variable and driven by a combination of relapse-driven disease activity and relapse-independent disease progression. The formation of new focal demyelinating lesions is associated with clinical relapses; however, the pathological mechanisms driving disease progression are less well understood. Current concepts suggest that ongoing focal and diffuse inflammation within the CNS in combination with an age-associated failure of compensatory and repair mechanisms contribute to disease progression. The aim of our study was to characterize the diffuse microglia activation in periplaque white matter (PPWM) of MS patients, to identify factors modulating its extent and to determine its potential correlation with loss or preservation of oligodendrocytes. We analyzed microglial and oligodendroglial numbers in PPWM in a cohort of 96 tissue blocks from 32 MS patients containing 100 lesions as well as a control cohort (n = 37). Microglia activation in PPWM was dependent on patient age, proximity to lesion, lesion type, and to a lesser degree on sex. Oligodendrocyte numbers were decreased in PPWM; however, increased microglia densities did not correlate with lower oligodendroglial cell counts, indicating that diffuse microglia activation is not sufficient to drive oligodendroglial loss in PPWM. In summary, our findings support the notion of the close relationship between focal and diffuse inflammation in MS and that age is an important modulator of MS pathology.
Collapse
Affiliation(s)
- Wiebke Kessler
- Institute of Neuropathology, University Hospital Münster, Pottkamp 2, 48149, Münster, Germany
| | - Christian Thomas
- Institute of Neuropathology, University Hospital Münster, Pottkamp 2, 48149, Münster, Germany
| | - Tanja Kuhlmann
- Institute of Neuropathology, University Hospital Münster, Pottkamp 2, 48149, Münster, Germany.
| |
Collapse
|
12
|
Galbusera R, Bahn E, Weigel M, Schaedelin S, Franz J, Lu P, Barakovic M, Melie‐Garcia L, Dechent P, Lutti A, Sati P, Reich DS, Nair G, Brück W, Kappos L, Stadelmann C, Granziera C. Postmortem quantitative MRI disentangles histological lesion types in multiple sclerosis. Brain Pathol 2023; 33:e13136. [PMID: 36480267 PMCID: PMC10580009 DOI: 10.1111/bpa.13136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 11/16/2022] [Indexed: 12/13/2022] Open
Abstract
Quantitative MRI (qMRI) probes the microstructural properties of the central nervous system (CNS) by providing biophysical measures of tissue characteristics. In this work, we aimed to (i) identify qMRI measures that distinguish histological lesion types in postmortem multiple sclerosis (MS) brains, especially the remyelinated ones; and to (ii) investigate the relationship between those measures and quantitative histological markers of myelin, axons, and astrocytes in the same experimental setting. Three fixed MS whole brains were imaged with qMRI at 3T to obtain magnetization transfer ratio (MTR), myelin water fraction (MWF), quantitative T1 (qT1), quantitative susceptibility mapping (QSM), fractional anisotropy (FA) and radial diffusivity (RD) maps. The identification of lesion types (active, inactive, chronic active, or remyelinated) and quantification of tissue components were performed using histological staining methods as well as immunohistochemistry and immunofluorescence. Pairwise logistic and LASSO regression models were used to identify the best qMRI discriminators of lesion types. The association between qMRI and quantitative histological measures was performed using Spearman's correlations and linear mixed-effect models. We identified a total of 65 lesions. MTR and MWF best predicted the chance of a lesion to be remyelinated, whereas RD and QSM were useful in the discrimination of active lesions. The measurement of microstructural properties through qMRI did not show any difference between chronic active and inactive lesions. MWF and RD were associated with myelin content in both lesions and normal-appearing white matter (NAWM), FA was the measure most associated with axon content in both locations, while MWF was associated with astrocyte immunoreactivity only in lesions. Moreover, we provided evidence of extensive astrogliosis in remyelinated lesions. Our study provides new information on the discriminative power of qMRI in differentiating MS lesions -especially remyelinated ones- as well as on the relative association between multiple qMRI measures and myelin, axon and astrocytes.
Collapse
Affiliation(s)
- Riccardo Galbusera
- Translational Imaging in Neurology (ThINk) Basel, Department of Biomedical Engineering, Faculty of MedicineUniversity Hospital Basel and University of BaselBaselSwitzerland
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB)University Hospital Basel and University of BaselBaselSwitzerland
| | - Erik Bahn
- Institute of NeuropathologyUniversity Medical CenterGöttingenGermany
| | - Matthias Weigel
- Translational Imaging in Neurology (ThINk) Basel, Department of Biomedical Engineering, Faculty of MedicineUniversity Hospital Basel and University of BaselBaselSwitzerland
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB)University Hospital Basel and University of BaselBaselSwitzerland
- Division of Radiological Physics, Department of RadiologyUniversity Hospital BaselBaselSwitzerland
| | - Sabine Schaedelin
- Clinical Trial Unit, Department of Clinical ResearchUniversity Hospital Basel, University of BaselBaselSwitzerland
| | - Jonas Franz
- Institute of NeuropathologyUniversity Medical CenterGöttingenGermany
- Campus Institute for Dynamics of Biological NetworksUniversity of GöttingenGöttingenGermany
- Max Planck Institute for Experimental MedicineGöttingenGermany
| | - Po‐Jui Lu
- Translational Imaging in Neurology (ThINk) Basel, Department of Biomedical Engineering, Faculty of MedicineUniversity Hospital Basel and University of BaselBaselSwitzerland
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB)University Hospital Basel and University of BaselBaselSwitzerland
| | - Muhamed Barakovic
- Translational Imaging in Neurology (ThINk) Basel, Department of Biomedical Engineering, Faculty of MedicineUniversity Hospital Basel and University of BaselBaselSwitzerland
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB)University Hospital Basel and University of BaselBaselSwitzerland
| | - Lester Melie‐Garcia
- Translational Imaging in Neurology (ThINk) Basel, Department of Biomedical Engineering, Faculty of MedicineUniversity Hospital Basel and University of BaselBaselSwitzerland
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB)University Hospital Basel and University of BaselBaselSwitzerland
| | - Peter Dechent
- Department of Cognitive NeurologyMR‐Research in Neurosciences, University Medical Center GöttingenGöttingenGermany
| | - Antoine Lutti
- Centre for Research in Neuroscience, Department of Clinical NeurosciencesLaboratoire de Recherche en Neuroimagerie (LREN) University Hospital and University of LausanneLausanneSwitzerland
| | - Pascal Sati
- Department of NeurologyCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Daniel S. Reich
- Translational Neuroradiology SectionNational Institute of Neurological Disorders and Stroke, National Institutes of HealthBethesdaMarylandUSA
| | - Govind Nair
- National Institute of Neurological Disorders and StrokeBethesdaMarylandUSA
| | - Wolfgang Brück
- Institute of NeuropathologyUniversity Medical CenterGöttingenGermany
| | - Ludwig Kappos
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB)University Hospital Basel and University of BaselBaselSwitzerland
| | - Christine Stadelmann
- Institute of NeuropathologyUniversity Medical CenterGöttingenGermany
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Network of Excitable Cells (MBExC) ”University of GoettingenGermany
| | - Cristina Granziera
- Translational Imaging in Neurology (ThINk) Basel, Department of Biomedical Engineering, Faculty of MedicineUniversity Hospital Basel and University of BaselBaselSwitzerland
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB)University Hospital Basel and University of BaselBaselSwitzerland
| |
Collapse
|
13
|
Drake SS, Mohammadnia A, Heale K, Groh AMR, Hua EML, Zaman A, Hintermayer MA, Zandee S, Gosselin D, Stratton JA, Sinclair DA, Fournier AE. Cellular rejuvenation protects neurons from inflammation mediated cell death. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.30.560301. [PMID: 37873446 PMCID: PMC10592844 DOI: 10.1101/2023.09.30.560301] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
In multiple sclerosis (MS), the invasion of the central nervous system by peripheral immune cells is followed by the activation of resident microglia and astrocytes. This cascade of events results in demyelination, which triggers neuronal damage and death. The molecular signals in neurons responsible for this damage are not yet fully characterized. In MS, retinal ganglion cell neurons (RGCs) of the central nervous system (CNS) undergo axonal injury and cell death. This phenomenon is mirrored in the experimental autoimmune encephalomyelitis (EAE) mouse model of MS. To understand the molecular landscape, we isolated RGCs from mice subjected to the EAE protocol. RNA-sequencing and ATAC-sequencing analyses were performed. Pathway analysis of the RNA-sequencing data revealed that RGCs displayed a molecular signature, similar to aged neurons, showcasing features of senescence. Single-nucleus RNA-sequencing analysis of neurons from human MS patients revealed a comparable senescence-like phenotype., which was supported by immunostaining RGCs in EAE mice. These changes include alterations to the nuclear envelope, modifications in chromatin marks, and accumulation of DNA damage. Transduction of RGCs with an Oct4 - Sox2 - Klf4 transgene to convert neurons in the EAE model to a more youthful epigenetic and transcriptomic state enhanced the survival of RGCs. Collectively, this research uncovers a previously unidentified senescent-like phenotype in neurons under pathological inflammation and neurons from MS patients. The rejuvenation of this aged transcriptome improved visual acuity and neuronal survival in the EAE model supporting the idea that age rejuvenation therapies and senotherapeutic agents could offer a direct means of neuroprotection in autoimmune disorders.
Collapse
|
14
|
Butruille L, Jubin P, Martin E, Aigrot MS, Lhomme M, Fini JB, Demeneix B, Stankoff B, Lubetzki C, Zalc B, Remaud S. Deleterious functional consequences of perfluoroalkyl substances accumulation into the myelin sheath. ENVIRONMENT INTERNATIONAL 2023; 180:108211. [PMID: 37751662 DOI: 10.1016/j.envint.2023.108211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 09/12/2023] [Accepted: 09/13/2023] [Indexed: 09/28/2023]
Abstract
Exposure to persistent organic pollutants during the perinatal period is of particular concern because of the potential increased risk of neurological disorders in adulthood. Here we questioned whether exposure to perfluorooctanoic acid (PFOA) and perfluorooctane sulfonate (PFOS) could alter myelin formation and regeneration. First, we show that PFOS, and to a lesser extent PFOA, accumulated into the myelin sheath of postnatal day 21 (p21) mice, whose mothers were exposed to either PFOA or PFOS (20 mg/L) via drinking water during late gestation and lactation, suggesting that accumulation of PFOS into the myelin could interfere with myelin formation and function. In fact, PFOS, but not PFOA, disrupted the generation of oligodendrocytes, the myelin-forming cells of the central nervous system, derived from neural stem cells localised in the subventricular zone of p21 exposed animals. Then, cerebellar slices were transiently demyelinated using lysophosphatidylcholine and remyelination was quantified in the presence of either PFOA or PFOS. Only PFOS impaired remyelination, a deleterious effect rescued by adding thyroid hormone (TH). Similarly to our observation in the mouse, we also showed that PFOS altered remyelination in Xenopus laevis using the Tg(Mbp:GFP-ntr) model of conditional demyelination and measuring, then, the number of oligodendrocytes. The functional consequences of PFOS-impaired remyelination were shown by its effects using a battery of behavioural tests. In sum, our data demonstrate that perinatal PFOS exposure disrupts oligodendrogenesis and myelin function through modulation of TH action. PFOS exposure may exacerbate genetic and environmental susceptibilities underlying myelin disorders, the most frequent being multiple sclerosis.
Collapse
Affiliation(s)
- L Butruille
- Sorbonne University, Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital, F-75013 Paris, France
| | - P Jubin
- Sorbonne University, Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital, F-75013 Paris, France
| | - E Martin
- Sorbonne University, Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital, F-75013 Paris, France
| | - M S Aigrot
- Sorbonne University, Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital, F-75013 Paris, France
| | - M Lhomme
- IHU ICAN (ICAN OMICS Lipidomics) Foundation for Innovation in Cardiometabolism and Nutrition, Pitié-Salpêtrière Hospital, F-75013 Paris, France
| | - J B Fini
- CNRS UMR 7221, Sorbonne University, Muséum National d'Histoire Naturelle, F-75005 Paris France
| | - B Demeneix
- CNRS UMR 7221, Sorbonne University, Muséum National d'Histoire Naturelle, F-75005 Paris France
| | - B Stankoff
- Sorbonne University, Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital, F-75013 Paris, France
| | - C Lubetzki
- Sorbonne University, Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital, F-75013 Paris, France
| | - B Zalc
- Sorbonne University, Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital, F-75013 Paris, France.
| | - S Remaud
- CNRS UMR 7221, Sorbonne University, Muséum National d'Histoire Naturelle, F-75005 Paris France.
| |
Collapse
|
15
|
Dustin E, McQuiston AR, Honke K, Palavicini JP, Han X, Dupree JL. Adult-onset depletion of sulfatide leads to axonal degeneration with relative myelin sparing. Glia 2023; 71:2285-2303. [PMID: 37283058 PMCID: PMC11007682 DOI: 10.1002/glia.24423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 05/10/2023] [Accepted: 05/12/2023] [Indexed: 06/08/2023]
Abstract
3-O-sulfogalactosylceramide (sulfatide) constitutes a class of sphingolipids that comprise about 4% of myelin lipids in the central nervous system. Previously, our group characterized a mouse with sulfatide's synthesizing enzyme, cerebroside sulfotransferase (CST), constitutively disrupted. Using these mice, we demonstrated that sulfatide is required for establishment and maintenance of myelin, axoglial junctions, and axonal domains and that sulfatide depletion results in structural pathologies commonly observed in Multiple Sclerosis (MS). Interestingly, sulfatide is reduced in regions of normal appearing white matter (NAWM) of MS patients. Sulfatide reduction in NAWM suggests depletion occurs early in disease development and consistent with functioning as a driving force of disease progression. To closely model MS, an adult-onset disease, our lab generated a "floxed" CST mouse and mated it against the PLP-creERT mouse, resulting in a double transgenic mouse that provides temporal and cell-type specific ablation of the Cst gene (Gal3st1). Using this mouse, we demonstrate adult-onset sulfatide depletion has limited effects on myelin structure but results in the loss of axonal integrity including deterioration of domain organization accompanied by axonal degeneration. Moreover, structurally preserved myelinated axons progressively lose the ability to function as myelinated axons, indicated by the loss of the N1 peak. Together, our findings indicate that sulfatide depletion, which occurs in the early stages of MS progression, is sufficient to drive the loss of axonal function independent of demyelination and that axonal pathology, which is responsible for the irreversible loss of neuronal function that is prevalent in MS, may occur earlier than previously recognized.
Collapse
Affiliation(s)
- E Dustin
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia, USA
- Research Service, Central Virginia Veterans Affairs Health Care Systems, Richmond, Virginia, USA
| | - A R McQuiston
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - K Honke
- Department of Biochemistry, Kochi University Medical School, Kochi, Japan
| | - J P Palavicini
- Department of Medicine, University of Texas Health San Antonio, San Antonio, Texas, USA
- Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - X Han
- Department of Medicine, University of Texas Health San Antonio, San Antonio, Texas, USA
- Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - J L Dupree
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia, USA
- Research Service, Central Virginia Veterans Affairs Health Care Systems, Richmond, Virginia, USA
| |
Collapse
|
16
|
Oost W, Huitema AJ, Kats K, Giepmans BNG, Kooistra SM, Eggen BJL, Baron W. Pathological ultrastructural alterations of myelinated axons in normal appearing white matter in progressive multiple sclerosis. Acta Neuropathol Commun 2023; 11:100. [PMID: 37340488 DOI: 10.1186/s40478-023-01598-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 06/02/2023] [Indexed: 06/22/2023] Open
Abstract
Multiple sclerosis (MS) pathophysiology includes inflammation, demyelination and neurodegeneration, but the exact mechanisms of disease initiation and progression are unknown. A major feature of lesions is lack of myelin, which increases axonal energy demand and requires adaptation in number and size of mitochondria. Outside lesions, subtle and diffuse alterations are observed in normal appearing white matter (NAWM) and normal appearing grey matter (NAGM), including increased oxidative stress, reduced axon density and changes in myelin composition and morphology. On an ultrastructural level, only limited data is available on alterations in myelinated axons. We generated large scale 2D scanning transmission electron microscopy images ('nanotomy') of non-demyelinated brain tissue of control and progressive MS donors, accessible via an open-access online repository. We observed a reduced density of myelinated axons in NAWM, without a decrease in cross-sectional axon area. Small myelinated axons were less frequently and large myelinated axons were more frequently present in NAWM, while the g-ratio was similar. The correlation between axonal mitochondrial radius and g-ratio was lost in NAWM, but not in NAGM. Myelinated axons in control GM and NAGM had a similar g-ratio and radius distribution. We hypothesize that axonal loss in NAWM is likely compensated by swelling of the remaining myelinated axons and subsequent adjustment of myelin thickness to maintain their g-ratio. Failure of axonal mitochondria to adjust their size and fine-tuning of myelin thickness may render NAWM axons and their myelin more susceptible to injury.
Collapse
Affiliation(s)
- Wendy Oost
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- MS Center Noord Nederland, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Allard J Huitema
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- MS Center Noord Nederland, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Kim Kats
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Ben N G Giepmans
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Susanne M Kooistra
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- MS Center Noord Nederland, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Bart J L Eggen
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- MS Center Noord Nederland, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Wia Baron
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
- MS Center Noord Nederland, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| |
Collapse
|
17
|
Doelman W, Reijnen RC, Dijksman N, Janssen APA, van Driel N, 't Hart BA, Philippens I, Araman C, Baron W, van Kasteren SI. Citrullinated human and murine MOG 35-55 display distinct biophysical and biochemical behavior. J Biol Chem 2023; 299:103065. [PMID: 36841486 PMCID: PMC10060747 DOI: 10.1016/j.jbc.2023.103065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 02/15/2023] [Accepted: 02/18/2023] [Indexed: 02/27/2023] Open
Abstract
The peptide spanning residues 35 to 55 of the protein myelin oligodendrocyte glycoprotein (MOG) has been studied extensively in its role as a key autoantigen in the neuroinflammatory autoimmune disease multiple sclerosis. Rodents and nonhuman primate species immunized with this peptide develop a neuroinflammatory condition called experimental autoimmune encephalomyelitis, often used as a model for multiple sclerosis. Over the last decade, the role of citrullination of this antigen in the disease onset and progression has come under increased scrutiny. We recently reported on the ability of these citrullinated MOG35-55 peptides to aggregate in an amyloid-like fashion, suggesting a new potential pathogenic mechanism underlying this disease. The immunodominant region of MOG is highly conserved between species, with the only difference between the murine and human protein, a polymorphism on position 42, which is serine in mice and proline for humans. Here, we show that the biophysical and biochemical behavior we previously observed for citrullinated murine MOG35-55 is fundamentally different for human and mouse MOG35-55. The citrullinated human peptides do not show amyloid-like behavior under the conditions where the murine peptides do. Moreover, we tested the ability of these peptides to stimulate lymphocytes derived from MOG immunized marmoset monkeys. While the citrullinated murine peptides did not produce a proliferative response, one of the citrullinated human peptides did. We postulate that this unexpected difference is caused by disparate antigen processing. Taken together, our results suggest that further study on the role of citrullination in MOG-induced experimental autoimmune encephalomyelitis is necessary.
Collapse
Affiliation(s)
- W Doelman
- Department of Bio-Organic Synthesis, Leiden Institute of Chemistry, Leiden, the Netherlands
| | - R C Reijnen
- Department of Bio-Organic Synthesis, Leiden Institute of Chemistry, Leiden, the Netherlands
| | - N Dijksman
- Section Molecular Neurobiology, Department Biomedical Sciences of Cells & Systems, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - A P A Janssen
- Department of Bio-Organic Synthesis, Leiden Institute of Chemistry, Leiden, the Netherlands
| | - N van Driel
- Department of Immunobiology, Biomedical Primate Research Center, Rijswijk, the Netherlands
| | - B A 't Hart
- Section Molecular Neurobiology, Department Biomedical Sciences of Cells & Systems, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - I Philippens
- Department of Immunobiology, Biomedical Primate Research Center, Rijswijk, the Netherlands
| | - C Araman
- Department of Bio-Organic Synthesis, Leiden Institute of Chemistry, Leiden, the Netherlands
| | - W Baron
- Section Molecular Neurobiology, Department Biomedical Sciences of Cells & Systems, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| | - S I van Kasteren
- Department of Bio-Organic Synthesis, Leiden Institute of Chemistry, Leiden, the Netherlands.
| |
Collapse
|
18
|
Schwarz K, Schmitz F. Synapse Dysfunctions in Multiple Sclerosis. Int J Mol Sci 2023; 24:ijms24021639. [PMID: 36675155 PMCID: PMC9862173 DOI: 10.3390/ijms24021639] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/11/2023] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic neuroinflammatory disease of the central nervous system (CNS) affecting nearly three million humans worldwide. In MS, cells of an auto-reactive immune system invade the brain and cause neuroinflammation. Neuroinflammation triggers a complex, multi-faceted harmful process not only in the white matter but also in the grey matter of the brain. In the grey matter, neuroinflammation causes synapse dysfunctions. Synapse dysfunctions in MS occur early and independent from white matter demyelination and are likely correlates of cognitive and mental symptoms in MS. Disturbed synapse/glia interactions and elevated neuroinflammatory signals play a central role. Glutamatergic excitotoxic synapse damage emerges as a major mechanism. We review synapse/glia communication under normal conditions and summarize how this communication becomes malfunctional during neuroinflammation in MS. We discuss mechanisms of how disturbed glia/synapse communication can lead to synapse dysfunctions, signaling dysbalance, and neurodegeneration in MS.
Collapse
|
19
|
van den Bosch AMR, Hümmert S, Steyer A, Ruhwedel T, Hamann J, Smolders J, Nave KA, Stadelmann C, Kole MHP, Möbius W, Huitinga I. Ultrastructural Axon-Myelin Unit Alterations in Multiple Sclerosis Correlate with Inflammation. Ann Neurol 2022; 93:856-870. [PMID: 36565265 DOI: 10.1002/ana.26585] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/13/2022] [Accepted: 12/17/2022] [Indexed: 12/25/2022]
Abstract
OBJECTIVE Changes in the normal-appearing white matter (NAWM) in multiple sclerosis (MS) may contribute to disease progression. Here, we systematically quantified ultrastructural and subcellular characteristics of the axon-myelin unit in MS NAWM and determined how this correlates with low-grade inflammation. METHODS Human brain tissue obtained with short postmortem delay and fixation at autopsy enables systematic quantification of ultrastructural characteristics. In this study, we performed high-resolution immunohis tochemistry and quantitative transmission electron microscopy to study inflammation and ultrastructural characteristics of the axon-myelin unit in MS NAWM (n = 8) and control white matter (WM) in the optic nerve. RESULTS In the MS NAWM, there were more activated and phagocytic microglia cells (HLA+ P2RY12- and Iba1+ CD68+ ) and more T cells (CD3+ ) compared to control WM, mainly located in the perivascular space. In MS NAWM compared to control WM, there were, as expected, longer paranodes and juxtaparanodes and larger overlap between paranodes and juxtaparanodes. There was less compact myelin wrapping, a lower g-ratio, and a higher frequency of axonal mitochondria. Changes in myelin and axonal mitochondrial frequency correlated positively with the number of active and phagocytic microglia and lymphocytes in the optic nerve. INTERPRETATION These data suggest that in MS NAWM myelin detachment and uncompact myelin wrapping occurs, potassium channels are unmasked at the nodes of Ranvier, and axonal energy demand is increased, or mitochondrial transport is stagnated, accompanied by increased presence of activated and phagocytic microglia and T cells. These subclinical alterations to the axon-myelin unit in MS NAWM may contribute to disease progression. ANN NEUROL 2023.
Collapse
Affiliation(s)
- Aletta M R van den Bosch
- Department of Neuroimmunology, Netherlands Institute for Neuroscience, Royal Netherlands Academy for Arts and Sciences, Amsterdam, the Netherlands
| | - Sophie Hümmert
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Anna Steyer
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Torben Ruhwedel
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Jörg Hamann
- Department of Neuroimmunology, Netherlands Institute for Neuroscience, Royal Netherlands Academy for Arts and Sciences, Amsterdam, the Netherlands.,Department of Experimental Immunology, Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Joost Smolders
- Department of Neuroimmunology, Netherlands Institute for Neuroscience, Royal Netherlands Academy for Arts and Sciences, Amsterdam, the Netherlands.,Department of Neurology and Immunology, Multiple Sclerosis Center ErasMS, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Christine Stadelmann
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Maarten H P Kole
- Department of Axonal Signaling, Netherlands Institute for Neuroscience, Royal Netherlands Academy for Arts and Sciences, Amsterdam, the Netherlands.,Cell Biology, Neurobiology, and Biophysics, Department of Biology, Faculty of Science, University of Utrecht, Utrecht, the Netherlands
| | - Wiebke Möbius
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Inge Huitinga
- Department of Neuroimmunology, Netherlands Institute for Neuroscience, Royal Netherlands Academy for Arts and Sciences, Amsterdam, the Netherlands.,Center for Neuroscience, Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
20
|
Sabet MF, Barman S, Beller M, Meuth SG, Melzer N, Aktas O, Goebels N, Prozorovski T. Myelinating Co-Culture as a Model to Study Anti-NMDAR Neurotoxicity. Int J Mol Sci 2022; 24:ijms24010248. [PMID: 36613687 PMCID: PMC9820503 DOI: 10.3390/ijms24010248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/06/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022] Open
Abstract
Anti-NMDA receptor (NMDAR) encephalitis is frequently associated with demyelinating disorders (e.g., multiple sclerosis (MS), neuromyelitis optica spectrum disorder (NMOSD), myelin oligodendrocyte glycoprotein-associated disease (MOGAD)) with regard to clinical presentation, neuropathological and cerebrospinal fluid findings. Indeed, autoantibodies (AABs) against the GluN1 (NR1) subunit of the NMDAR diminish glutamatergic transmission in both neurons and oligodendrocytes, leading to a state of NMDAR hypofunction. Considering the vital role of oligodendroglial NMDAR signaling in neuron-glia communication and, in particular, in tightly regulated trophic support to neurons, the influence of GluN1 targeting on the physiology of myelinated axon may be of importance. We applied a myelinating spinal cord cell culture model that contains all major CNS cell types, to evaluate the effects of a patient-derived GluN1-specific monoclonal antibody (SSM5) on neuronal and myelin integrity. A non-brain reactive (12D7) antibody was used as the corresponding isotype control. We show that in cultures at the late stage of myelination, prolonged treatment with SSM5, but not 12D7, leads to neuronal damage. This is characterized by neurite blebbing and fragmentation, and a reduction in the number of myelinated axons. However, this significant toxic effect of SSM5 was not observed in earlier cultures at the beginning of myelination. Anti-GluN1 AABs induce neurodegenerative changes and associated myelin loss in myelinated spinal cord cultures. These findings may point to the higher vulnerability of myelinated neurons towards interference in glutamatergic communication, and may refer to the disturbance of the NMDAR-mediated oligodendrocyte metabolic supply. Our work contributes to the understanding of the emerging association of NMDAR encephalitis with demyelinating disorders.
Collapse
Affiliation(s)
- Mercedeh Farhat Sabet
- Department of Neurology, Medical Faculty, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany
| | - Sumanta Barman
- Department of Neurology, Medical Faculty, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany
| | - Mathias Beller
- Institut für Mathematische Modellierung Biologischer Systeme, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany
| | - Sven G. Meuth
- Department of Neurology, Medical Faculty, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany
| | - Nico Melzer
- Department of Neurology, Medical Faculty, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany
| | - Orhan Aktas
- Department of Neurology, Medical Faculty, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany
| | - Norbert Goebels
- Department of Neurology, Medical Faculty, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany
- Correspondence: (N.G.); (T.P.); Tel.: +49-211-81-04594 (N.G.); +49-211-81-05146 (T.P.)
| | - Tim Prozorovski
- Department of Neurology, Medical Faculty, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany
- Correspondence: (N.G.); (T.P.); Tel.: +49-211-81-04594 (N.G.); +49-211-81-05146 (T.P.)
| |
Collapse
|
21
|
Fathy YY, Jonkman LE, Bol JJ, Timmermans E, Jonker AJ, Rozemuller AJM, van de Berg WDJ. Axonal degeneration in the anterior insular cortex is associated with Alzheimer's co-pathology in Parkinson's disease and dementia with Lewy bodies. Transl Neurodegener 2022; 11:52. [PMID: 36474289 PMCID: PMC9728006 DOI: 10.1186/s40035-022-00325-x] [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: 06/17/2022] [Accepted: 11/08/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Axons, crucial for impulse transmission and cellular trafficking, are thought to be primary targets of neurodegeneration in Parkinson's disease (PD) and dementia with Lewy bodies (DLB). Axonal degeneration occurs early, preceeding and exceeding neuronal loss, and contributes to the spread of pathology, yet is poorly described outside the nigrostriatal circuitry. The insula, a cortical brain hub, was recently discovered to be highly vulnerable to pathology and plays a role in cognitive deficits in PD and DLB. The aim of this study was to evaluate morphological features as well as burden of proteinopathy and axonal degeneration in the anterior insular sub-regions in PD, PD with dementia (PDD), and DLB. METHODS α-Synuclein, phosphorylated (p-)tau, and amyloid-β pathology load were evaluated in the anterior insular (agranular and dysgranular) subregions of post-mortem human brains (n = 27). Axonal loss was evaluated using modified Bielschowsky silver staining and quantified using stereology. Cytoskeletal damage was comprehensively studied using immunofluorescent multi-labelling and 3D confocal laser-scanning microscopy. RESULTS Compared to PD and PDD, DLB showed significantly higher α-synuclein and p-tau pathology load, argyrophilic grains, and more severe axonal loss, particularly in the anterior agranular insula. Alternatively, the dysgranular insula showed a significantly higher load of amyloid-β pathology and its axonal density correlated with cognitive performance. p-Tau contributed most to axonal loss in the DLB group, was highest in the anterior agranular insula and significantly correlated with CDR global scores for dementia. Neurofilament and myelin showed degenerative changes including swellings, demyelination, and detachment of the axon-myelin unit. CONCLUSIONS Our results highlight the selective vulnerability of the anterior insular sub-regions to various converging pathologies, leading to impaired axonal integrity in PD, PDD and DLB, disrupting their functional properties and potentially contributing to cognitive, emotional, and autonomic deficits.
Collapse
Affiliation(s)
- Yasmine Y. Fathy
- grid.12380.380000 0004 1754 9227Amsterdam UMC, Department of Anatomy and Neurosciences, Section Clinical Neuroanatomy and Biobanking, Amsterdam Neuroscience, Vrije University Amsterdam, O
- 2 Life Sciences building, De Boelelaan 1108, 1081 HZ Amsterdam, Netherlands ,grid.484519.5Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, the Netherlands ,grid.5645.2000000040459992XDepartment of Neurology, Erasmus Medical Center, Postbus 2040, 3000 CA Rotterdam, Netherlands
| | - Laura E. Jonkman
- grid.12380.380000 0004 1754 9227Amsterdam UMC, Department of Anatomy and Neurosciences, Section Clinical Neuroanatomy and Biobanking, Amsterdam Neuroscience, Vrije University Amsterdam, O
- 2 Life Sciences building, De Boelelaan 1108, 1081 HZ Amsterdam, Netherlands ,grid.484519.5Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, the Netherlands
| | - John J. Bol
- grid.12380.380000 0004 1754 9227Amsterdam UMC, Department of Anatomy and Neurosciences, Section Clinical Neuroanatomy and Biobanking, Amsterdam Neuroscience, Vrije University Amsterdam, O
- 2 Life Sciences building, De Boelelaan 1108, 1081 HZ Amsterdam, Netherlands ,grid.484519.5Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, the Netherlands
| | - Evelien Timmermans
- grid.12380.380000 0004 1754 9227Amsterdam UMC, Department of Anatomy and Neurosciences, Section Clinical Neuroanatomy and Biobanking, Amsterdam Neuroscience, Vrije University Amsterdam, O
- 2 Life Sciences building, De Boelelaan 1108, 1081 HZ Amsterdam, Netherlands ,grid.484519.5Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, the Netherlands
| | - Allert J. Jonker
- grid.12380.380000 0004 1754 9227Amsterdam UMC, Department of Anatomy and Neurosciences, Section Clinical Neuroanatomy and Biobanking, Amsterdam Neuroscience, Vrije University Amsterdam, O
- 2 Life Sciences building, De Boelelaan 1108, 1081 HZ Amsterdam, Netherlands ,grid.484519.5Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, the Netherlands
| | - Annemieke J. M. Rozemuller
- grid.484519.5Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, the Netherlands ,grid.12380.380000 0004 1754 9227Amsterdam UMC, Department of Pathology, Amsterdam Neuroscience, Vrije University Amsterdam, De Boelelaan, Amsterdam, Netherlands
| | - Wilma D. J. van de Berg
- grid.12380.380000 0004 1754 9227Amsterdam UMC, Department of Anatomy and Neurosciences, Section Clinical Neuroanatomy and Biobanking, Amsterdam Neuroscience, Vrije University Amsterdam, O
- 2 Life Sciences building, De Boelelaan 1108, 1081 HZ Amsterdam, Netherlands ,grid.484519.5Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, the Netherlands
| |
Collapse
|
22
|
Collongues N, Becker G, Jolivel V, Ayme-Dietrich E, de Seze J, Binamé F, Patte-Mensah C, Monassier L, Mensah-Nyagan AG. A Narrative Review on Axonal Neuroprotection in Multiple Sclerosis. Neurol Ther 2022; 11:981-1042. [PMID: 35610531 PMCID: PMC9338208 DOI: 10.1007/s40120-022-00363-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 05/03/2022] [Indexed: 01/08/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) resulting in demyelination and neurodegeneration. The therapeutic strategy is now largely based on reducing inflammation with immunosuppressive drugs. Unfortunately, when disease progression is observed, no drug offers neuroprotection apart from its anti-inflammatory effect. In this review, we explore current knowledge on the assessment of neurodegeneration in MS and look at putative targets that might prove useful in protecting the axon from degeneration. Among them, Bruton's tyrosine kinase inhibitors, anti-apoptotic and antioxidant agents, sex hormones, statins, channel blockers, growth factors, and molecules preventing glutamate excitotoxicity have already been studied. Some of them have reached phase III clinical trials and carry a great message of hope for our patients with MS.
Collapse
Affiliation(s)
- Nicolas Collongues
- Department of Neurology, University Hospital of Strasbourg, Strasbourg, France. .,Center for Clinical Investigation, INSERM U1434, Strasbourg, France. .,Biopathology of Myelin, Neuroprotection and Therapeutic Strategy, INSERM U1119, Strasbourg, France. .,University Department of Pharmacology, Addictology, Toxicology and Therapeutic, Strasbourg University, Strasbourg, France.
| | - Guillaume Becker
- University Department of Pharmacology, Addictology, Toxicology and Therapeutic, Strasbourg University, Strasbourg, France.,NeuroCardiovascular Pharmacology and Toxicology Laboratory, UR7296, University Hospital of Strasbourg, Strasbourg, France
| | - Valérie Jolivel
- Biopathology of Myelin, Neuroprotection and Therapeutic Strategy, INSERM U1119, Strasbourg, France
| | - Estelle Ayme-Dietrich
- University Department of Pharmacology, Addictology, Toxicology and Therapeutic, Strasbourg University, Strasbourg, France.,NeuroCardiovascular Pharmacology and Toxicology Laboratory, UR7296, University Hospital of Strasbourg, Strasbourg, France
| | - Jérôme de Seze
- Department of Neurology, University Hospital of Strasbourg, Strasbourg, France.,Center for Clinical Investigation, INSERM U1434, Strasbourg, France.,Biopathology of Myelin, Neuroprotection and Therapeutic Strategy, INSERM U1119, Strasbourg, France
| | - Fabien Binamé
- Biopathology of Myelin, Neuroprotection and Therapeutic Strategy, INSERM U1119, Strasbourg, France
| | - Christine Patte-Mensah
- Biopathology of Myelin, Neuroprotection and Therapeutic Strategy, INSERM U1119, Strasbourg, France
| | - Laurent Monassier
- University Department of Pharmacology, Addictology, Toxicology and Therapeutic, Strasbourg University, Strasbourg, France.,NeuroCardiovascular Pharmacology and Toxicology Laboratory, UR7296, University Hospital of Strasbourg, Strasbourg, France
| | - Ayikoé Guy Mensah-Nyagan
- Biopathology of Myelin, Neuroprotection and Therapeutic Strategy, INSERM U1119, Strasbourg, France
| |
Collapse
|
23
|
Sobel RA, Albertelli M, Hinojoza JR, Eaton MJ, Grimes KV, Rubenstein E. Azetidine-2-Carboxylic Acid-Induced Oligodendrogliopathy: Relevance to the Pathogenesis of Multiple Sclerosis. J Neuropathol Exp Neurol 2022; 81:414-433. [PMID: 35521963 PMCID: PMC9123080 DOI: 10.1093/jnen/nlac028] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The naturally occurring imino acid azetidine-2-carboxylic acid (Aze) is consumed by humans and can be misincorporated in place of proline in myelin basic protein (MBP) in vitro. To determine Aze effects on the mammalian CNS in vivo, adult CD1 mice were given Aze orally or intraperitoneally. Clinical signs reminiscent of MBP-mutant mice occurred with 600 mg/kg Aze exposure. Aze induced oligodendrocyte (OL) nucleomegaly and nucleoplasm clearing, dilated endoplasmic reticulum, cytoplasmic vacuolation, abnormal mitochondria, and Aze dose-dependent apoptosis. Immunohistochemistry demonstrated myelin blistering and nuclear translocation of unfolded protein response (UPR)/proinflammatory molecules (ATF3, ATF4, ATF6, eIF2α, GADD153, NFκB, PERK, XBP1), MHC I expression, and MBP cytoplasmic aggregation in OL. There were scattered microglial nodules in CNS white matter (WM); other CNS cells appeared unaffected. Mice given Aze in utero and postnatally showed more marked effects than their dams. These OL, myelin, and microglial alterations are found in normal-appearing WM (NAWM) in multiple sclerosis (MS) patients. Thus, Aze induces a distinct oligodendrogliopathy in mice that recapitulates MS NAWM pathology without leukocyte infiltration. Because myelin proteins are relatively stable throughout life, we hypothesize that Aze misincorporation in myelin proteins during myelinogenesis in humans results in a progressive UPR that may be a primary process in MS pathogenesis.
Collapse
Affiliation(s)
- Raymond A Sobel
- From the Laboratory Service, Veterans Affairs Health Care System, Palo Alto, California, USA.,Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| | - Megan Albertelli
- Department of Comparative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Julian R Hinojoza
- From the Laboratory Service, Veterans Affairs Health Care System, Palo Alto, California, USA.,Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| | - Mary Jane Eaton
- From the Laboratory Service, Veterans Affairs Health Care System, Palo Alto, California, USA.,Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| | - Kevin V Grimes
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, California, USA
| | - Edward Rubenstein
- Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
24
|
Halperin ST, ’t Hart BA, Luchicchi A, Schenk GJ. The Forgotten Brother: The Innate-like B1 Cell in Multiple Sclerosis. Biomedicines 2022; 10:606. [PMID: 35327408 PMCID: PMC8945227 DOI: 10.3390/biomedicines10030606] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/21/2022] [Accepted: 03/01/2022] [Indexed: 02/04/2023] Open
Abstract
Multiple sclerosis (MS) is a neurodegenerative disease of the central nervous system (CNS), traditionally considered a chronic autoimmune attack against the insulating myelin sheaths around axons. However, the exact etiology has not been identified and is likely multi-factorial. Recently, evidence has been accumulating that implies that autoimmune processes underlying MS may, in fact, be triggered by pathological processes initiated within the CNS. This review focuses on a relatively unexplored immune cell-the "innate-like" B1 lymphocyte. The B1 cell is a primary-natural-antibody- and anti-inflammatory-cytokine-producing cell present in the healthy brain. It has been recently shown that its frequency and function may differ between MS patients and healthy controls, but its exact involvement in the MS pathogenic process remains obscure. In this review, we propose that this enigmatic cell may play a more prominent role in MS pathology than ever imagined. We aim to shed light on the human B1 cell in health and disease, and how dysregulation in its delicate homeostatic role could impact MS. Furthermore, novel therapeutic avenues to restore B1 cells' beneficial functions will be proposed.
Collapse
Affiliation(s)
| | | | - Antonio Luchicchi
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, MS Center Amsterdam, Amsterdam UMC, Vrije Universiteit, 1081 HZ Amsterdam, The Netherlands; (S.T.H.); (B.A.’t.H.)
| | - Geert J. Schenk
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, MS Center Amsterdam, Amsterdam UMC, Vrije Universiteit, 1081 HZ Amsterdam, The Netherlands; (S.T.H.); (B.A.’t.H.)
| |
Collapse
|
25
|
Joost S, Schweiger F, Pfeiffer F, Ertl C, Keiler J, Frank M, Kipp M. Cuprizone Intoxication Results in Myelin Vacuole Formation. Front Cell Neurosci 2022; 16:709596. [PMID: 35250482 PMCID: PMC8895267 DOI: 10.3389/fncel.2022.709596] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 01/28/2022] [Indexed: 11/17/2022] Open
Abstract
Myelin damage is a histopathological hallmark of multiple sclerosis lesions. Results of post mortem studies suggest that impaired myelin-axon interaction characterized by focal myelin detachments is an early event during lesion genesis. In this study, we investigated the ultrastructural changes of the axon-myelin interface in the cuprizone model using serial block face scanning electron microscopy and immunohistochemistry. We show that non-inflammatory injury of oligodendrocytes by cuprizone intoxication results in myelin vacuole formation and axonal swellings, paralleled by early alterations of the node of Ranvier cytoarchitecture. This remarkable resemblance of ultrastructural myelin characteristics in multiple sclerosis and the cuprizone animal model suggests that the cuprizone model is a valuable tool to study early pathologies during lesion formation.
Collapse
Affiliation(s)
- Sarah Joost
- Institute of Anatomy, Rostock University Medical Center, Rostock, Germany
| | - Felix Schweiger
- Institute of Anatomy, Rostock University Medical Center, Rostock, Germany
| | - Friederike Pfeiffer
- Werner Reichardt Centre for Integrative Neuroscience, University of Tübingen, Tübingen, Germany
| | - Carolin Ertl
- Institute of Anatomy, Rostock University Medical Center, Rostock, Germany
| | - Jonas Keiler
- Institute of Anatomy, Rostock University Medical Center, Rostock, Germany
| | - Marcus Frank
- Medical Biology and Electron Microscopy Center, Rostock University Medical Center, Rostock, Germany
- Department of Life, Light and Matter, University of Rostock, Rostock, Germany
| | - Markus Kipp
- Institute of Anatomy, Rostock University Medical Center, Rostock, Germany
- Center for Transdisciplinary Neurosciences Rostock, Rostock University Medical Center, Rostock, Germany
- *Correspondence: Markus Kipp,
| |
Collapse
|
26
|
Kular L, Ewing E, Needhamsen M, Pahlevan Kakhki M, Covacu R, Gomez-Cabrero D, Brundin L, Jagodic M. DNA methylation changes in glial cells of the normal-appearing white matter in Multiple Sclerosis patients. Epigenetics 2022; 17:1311-1330. [PMID: 35094644 PMCID: PMC9586622 DOI: 10.1080/15592294.2021.2020436] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Multiple Sclerosis (MS), the leading cause of non-traumatic neurological disability in young adults, is a chronic inflammatory and neurodegenerative disease of the central nervous system (CNS). Due to the poor accessibility to the target organ, CNS-confined processes underpinning the later progressive form of MS remain elusive thereby limiting treatment options. We aimed to examine DNA methylation, a stable epigenetic mark of genome activity, in glial cells to capture relevant molecular changes underlying MS neuropathology. We profiled DNA methylation in nuclei of non-neuronal cells, isolated from 38 post-mortem normal-appearing white matter (NAWM) specimens of MS patients (n = 8) in comparison to white matter of control individuals (n = 14), using Infinium MethylationEPIC BeadChip. We identified 1,226 significant (genome-wide adjusted P-value < 0.05) differentially methylated positions (DMPs) between MS patients and controls. Functional annotation of the altered DMP-genes uncovered alterations of processes related to cellular motility, cytoskeleton dynamics, metabolic processes, synaptic support, neuroinflammation and signaling, such as Wnt and TGF-β pathways. A fraction of the affected genes displayed transcriptional differences in the brain of MS patients, as reported by publically available transcriptomic data. Cell type-restricted annotation of DMP-genes attributed alterations of cytoskeleton rearrangement and extracellular matrix remodelling to all glial cell types, while some processes, including ion transport, Wnt/TGF-β signaling and immune processes were more specifically linked to oligodendrocytes, astrocytes and microglial cells, respectively. Our findings strongly suggest that NAWM glial cells are highly altered, even in the absence of lesional insult, collectively exhibiting a multicellular reaction in response to diffuse inflammation.
Collapse
Affiliation(s)
- Lara Kular
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Ewoud Ewing
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Maria Needhamsen
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Majid Pahlevan Kakhki
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Ruxandra Covacu
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - David Gomez-Cabrero
- Department of Medicine, Unit of Computational Medicine, Center for Molecular Medicine, Karolinska Institutet, Solna, Sweden
- Mucosal and Salivary Biology Division, King’s College London Dental Institute, London, UK
- Translational Bioinformatics Unit, Navarrabiomed, Complejo Hospitalario de Navarra (Chn), Universidad Pública de Navarra (Upna), IdiSNA, Pamplona, Spain
- Biological & Environmental Sciences & Engineering Division, King Abdullah University of Science & Technology, Thuwal, Kingdom of Saudi Arabia
| | - Lou Brundin
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Maja Jagodic
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
27
|
Miedema A, Gerrits E, Brouwer N, Jiang Q, Kracht L, Meijer M, Nutma E, Peferoen-Baert R, Pijnacker ATE, Wesseling EM, Wijering MHC, Gabius HJ, Amor S, Eggen BJL, Kooistra SM. Brain macrophages acquire distinct transcriptomes in multiple sclerosis lesions and normal appearing white matter. Acta Neuropathol Commun 2022; 10:8. [PMID: 35090578 PMCID: PMC8796391 DOI: 10.1186/s40478-021-01306-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/15/2021] [Indexed: 12/13/2022] Open
Abstract
Multiple sclerosis (MS) is a disease of the central nervous system that is characterized by inflammation and focal areas of demyelination, ultimately resulting in axonal degradation and neuronal loss. Several lines of evidence point towards a role for microglia and other brain macrophages in disease initiation and progression, but exactly how lesion formation is triggered is currently unknown. Here, we characterized early changes in MS brain tissue through transcriptomic analysis of normal appearing white matter (NAWM). We found that NAWM was characterized by enriched expression of genes associated with inflammation and cellular stress derived from brain macrophages. Single cell RNA sequencing confirmed a stress response in brain macrophages in NAWM and identified specific microglia and macrophage subsets at different stages of demyelinating lesions. We identified both phagocytic/activated microglia and CAM clusters that were associated with various MS lesion types. These overall changes in microglia and macrophages associated with lesion development in MS brain tissue may provide therapeutic targets to limit lesion progression and demyelination.
Collapse
|
28
|
Morgan ML, Teo W, Hernandez Y, Brideau C, Cummins K, Kuipers HF, Stys PK. Cuprizone-induced Demyelination in Mouse Brain is not due to Depletion of Copper. ASN Neuro 2022; 14:17590914221126367. [PMID: 36114624 PMCID: PMC9483969 DOI: 10.1177/17590914221126367] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
The cuprizone (CPZ) model allows the study of the biochemical processes underlying
nonautoimmune-mediated demyelination, remyelination, and chronic white matter disease
progression. CPZ is a copper (Cu) chelator that chiefly causes oligodendrocyte apoptosis
in the corpus callosum and cerebellum when administered in the mouse diet. While
disruption of Cu homeostasis is known to cause neurodegeneration (as is observed in
Wilson’s and Menkes disease), no consensus exists to date as to CPZ’s mechanism of action.
We sought to determine whether CPZ-induced pathology is due to Cu depletion as is
generally believed. Cu supplementation in chow, in stoichiometric excess to the added CPZ,
did not reduce CPZ-induced demyelination in C57Bl/6 mice. Moreover, equivalent doses of
other known Cu chelators neocuproine and D-penicillamine (D-Pen) failed to induce central
nervous system (CNS) demyelination. Since administration of D-Pen in the treatment of
Wilson’s disease can induce hypocupremia, we next sought to recreate penicillamine-induced
Cu deficiency to compare with purported CPZ-induced Cu deficiency. The resulting clinical
phenotype and histopathology were unlike that of CPZ. D-Pen-treated mice exhibited digit
paralysis, tail flaccidity, subcutaneous hemorrhaging, and optic and sciatic neuropathy,
all of which were prevented with Cu supplementation. No demyelination of the corpus
callosum or cerebellum was observed, even with D-Pen doses tenfold higher than CPZ.
Intriguingly, addition of D-Pen to the CPZ diet paradoxically prevented demyelination in a
dose-dependent manner.
Collapse
Affiliation(s)
- Megan L. Morgan
- Cumming School of Medicine, Department of Clinical Neurosciences, University of Calgary, Hotchkiss Brain Institute, Calgary, 3330 Hospital Drive N.W. HRIC 1B37A, Calgary, AB, T2N 4N1, Canada
| | - Wulin Teo
- Cumming School of Medicine, Department of Clinical Neurosciences, University of Calgary, Hotchkiss Brain Institute, Calgary, 3330 Hospital Drive N.W. HRIC 1B37A, Calgary, AB, T2N 4N1, Canada
| | - Yda Hernandez
- Cumming School of Medicine, Department of Clinical Neurosciences, University of Calgary, Hotchkiss Brain Institute, Calgary, 3330 Hospital Drive N.W. HRIC 1B37A, Calgary, AB, T2N 4N1, Canada
| | - Craig Brideau
- Cumming School of Medicine, Department of Clinical Neurosciences, University of Calgary, Hotchkiss Brain Institute, Calgary, 3330 Hospital Drive N.W. HRIC 1B37A, Calgary, AB, T2N 4N1, Canada
| | - Karen Cummins
- Cumming School of Medicine, Department of Clinical Neurosciences, University of Calgary, Hotchkiss Brain Institute, Calgary, 3330 Hospital Drive N.W. HRIC 1B37A, Calgary, AB, T2N 4N1, Canada
| | - Hedwich F. Kuipers
- Cumming School of Medicine, Department of Clinical Neurosciences, University of Calgary, Hotchkiss Brain Institute, Calgary, 3330 Hospital Drive N.W. HRIC 1B37A, Calgary, AB, T2N 4N1, Canada
| | - Peter K. Stys
- Cumming School of Medicine, Department of Clinical Neurosciences, University of Calgary, Hotchkiss Brain Institute, Calgary, 3330 Hospital Drive N.W. HRIC 1B37A, Calgary, AB, T2N 4N1, Canada
| |
Collapse
|
29
|
Huitema MJD, Strijbis EMM, Luchicchi A, Bol JGJM, Plemel JR, Geurts JJG, Schenk GJ. Myelin Quantification in White Matter Pathology of Progressive Multiple Sclerosis Post-Mortem Brain Samples: A New Approach for Quantifying Remyelination. Int J Mol Sci 2021; 22:ijms222312634. [PMID: 34884445 PMCID: PMC8657470 DOI: 10.3390/ijms222312634] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/16/2021] [Accepted: 11/18/2021] [Indexed: 01/14/2023] Open
Abstract
Multiple sclerosis (MS) is a demyelinating and neurodegenerative disease of the central nervous system (CNS). Repair through remyelination can be extensive, but quantification of remyelination remains challenging. To date, no method for standardized digital quantification of remyelination of MS lesions exists. This methodological study aims to present and validate a novel standardized method for myelin quantification in progressive MS brains to study myelin content more precisely. Fifty-five MS lesions in 32 tissue blocks from 14 progressive MS cases and five tissue blocks from 5 non-neurological controls were sampled. MS lesions were selected by macroscopic investigation of WM by standard histopathological methods. Tissue sections were stained for myelin with luxol fast blue (LFB) and histological assessment of de- or remyelination was performed by light microscopy. The myelin quantity was estimated with a novel myelin quantification method (MQM) in ImageJ. Three independent raters applied the MQM and the inter-rater reliability was calculated. We extended the method to diffusely appearing white matter (DAWM) and encephalitis to test potential wider applicability of the method. Inter-rater agreement was excellent (ICC = 0.96) and there was a high reliability with a lower- and upper limit of agreement up to −5.93% to 18.43% variation in myelin quantity. This study builds on the established concepts of histopathological semi-quantitative assessment of myelin and adds a novel, reliable and accurate quantitative measurement tool for the assessment of myelination in human post-mortem samples.
Collapse
Affiliation(s)
- Marije J. D. Huitema
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, MS Center Amsterdam, Amsterdam UMC, VU University Medical Center, De Boelelaan 1108, 1081 HV Amsterdam, The Netherlands; (M.J.D.H.); (A.L.); (J.G.J.M.B.); (J.J.G.G.)
| | - Eva M. M. Strijbis
- Department of Neurology, MS Center Amsterdam, Amsterdam UMC, VU University Medical Center, 1081 HZ Amsterdam, The Netherlands;
| | - Antonio Luchicchi
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, MS Center Amsterdam, Amsterdam UMC, VU University Medical Center, De Boelelaan 1108, 1081 HV Amsterdam, The Netherlands; (M.J.D.H.); (A.L.); (J.G.J.M.B.); (J.J.G.G.)
| | - John G. J. M. Bol
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, MS Center Amsterdam, Amsterdam UMC, VU University Medical Center, De Boelelaan 1108, 1081 HV Amsterdam, The Netherlands; (M.J.D.H.); (A.L.); (J.G.J.M.B.); (J.J.G.G.)
| | - Jason R. Plemel
- Department of Medicine, Division of Neurology, Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2S2, Canada;
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, AB T6G 2S2, Canada
| | - Jeroen J. G. Geurts
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, MS Center Amsterdam, Amsterdam UMC, VU University Medical Center, De Boelelaan 1108, 1081 HV Amsterdam, The Netherlands; (M.J.D.H.); (A.L.); (J.G.J.M.B.); (J.J.G.G.)
| | - Geert J. Schenk
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, MS Center Amsterdam, Amsterdam UMC, VU University Medical Center, De Boelelaan 1108, 1081 HV Amsterdam, The Netherlands; (M.J.D.H.); (A.L.); (J.G.J.M.B.); (J.J.G.G.)
- Correspondence:
| |
Collapse
|
30
|
Mechanism-based criteria to improve therapeutic outcomes in progressive multiple sclerosis. Nat Rev Neurol 2021; 18:40-55. [PMID: 34732831 DOI: 10.1038/s41582-021-00581-x] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2021] [Indexed: 02/07/2023]
Abstract
In contrast to the multiple disease-modifying therapies that are available for relapsing-remitting multiple sclerosis (MS), the therapeutic options for progressive MS (PMS) are limited. Recent advances in our understanding of the neuroimmunology of PMS, including the mechanisms that drive slowly expanding lesions, have fuelled optimism for improved treatment of this condition. In this Review, we highlight the commonly observed neuropathology of PMS and discuss the associated mechanisms of CNS injury. We then apply this knowledge to formulate criteria for therapeutic efficacy in PMS, beginning with the need for early treatment owing to the substantial neuropathology that is already present at the initial clinical presentation. Other requirements include: antagonism of neuroaxonal injury mediators such as pro-inflammatory microglia and lymphocytes; remediation of oxidative stress resulting from iron deposition and mitochondrial dysfunction; and promotion of neuroprotection through remyelination. We consider whether current disease-modifying therapies for relapsing-remitting MS meet the criteria for successful therapeutics in PMS and suggest that the evidence favours the early introduction of sphingosine 1-phosphate receptor modulators. Finally, we weigh up emerging medications, including repurposed generic medications and Bruton's tyrosine kinase inhibitors, against these fundamental criteria. In this new therapeutic era in PMS, success depends collectively on understanding disease mechanisms, drug characteristics (including brain penetration) and rational use.
Collapse
|
31
|
Elliott C, Momayyezsiahkal P, Arnold DL, Liu D, Ke J, Zhu L, Zhu B, George IC, Bradley DP, Fisher E, Cahir-McFarland E, Stys PK, Geurts JJG, Franchimont N, Gafson A, Belachew S. Abnormalities in normal-appearing white matter from which multiple sclerosis lesions arise. Brain Commun 2021; 3:fcab176. [PMID: 34557664 PMCID: PMC8453433 DOI: 10.1093/braincomms/fcab176] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 06/08/2021] [Accepted: 06/09/2021] [Indexed: 11/24/2022] Open
Abstract
Normal-appearing white matter is far from normal in multiple sclerosis; little is known about the precise pathology or spatial pattern of this alteration and its relation to subsequent lesion formation. This study was undertaken to evaluate normal-appearing white matter abnormalities in brain areas where multiple sclerosis lesions subsequently form, and to investigate the spatial distribution of normal-appearing white matter abnormalities in persons with multiple sclerosis. Brain MRIs of pre-lesion normal-appearing white matter were analysed in participants with new T2 lesions, pooled from three clinical trials: SYNERGY (NCT01864148; n = 85 with relapsing multiple sclerosis) was the test data set; ASCEND (NCT01416181; n = 154 with secondary progressive multiple sclerosis) and ADVANCE (NCT00906399; n = 261 with relapsing-remitting multiple sclerosis) were used as validation data sets. Focal normal-appearing white matter tissue state was analysed prior to lesion formation in areas where new T2 lesions later formed (pre-lesion normal-appearing white matter) using normalized magnetization transfer ratio and T2-weighted (nT2) intensities, and compared with overall normal-appearing white matter and spatially matched contralateral normal-appearing white matter. Each outcome was analysed using linear mixed-effects models. Follow-up time (as a categorical variable), patient-level characteristics (including treatment group) and other baseline variables were treated as fixed effects. In SYNERGY, nT2 intensity was significantly higher, and normalized magnetization transfer ratio was lower in pre-lesion normal-appearing white matter versus overall and contralateral normal-appearing white matter at all time points up to 24 weeks before new T2 lesion onset. In ASCEND and ADVANCE (for which normalized magnetization transfer ratio was not available), nT2 intensity in pre-lesion normal-appearing white matter was significantly higher compared to both overall and contralateral normal-appearing white matter at all pre-lesion time points extending up to 2 years prior to lesion formation. In all trials, nT2 intensity in the contralateral normal-appearing white matter was also significantly higher at all pre-lesion time points compared to overall normal-appearing white matter. Brain atlases of normal-appearing white matter abnormalities were generated using measures of voxel-wise differences in normalized magnetization transfer ratio of normal-appearing white matter in persons with multiple sclerosis compared to scanner-matched healthy controls. We observed that overall spatial distribution of normal-appearing white matter abnormalities in persons with multiple sclerosis largely recapitulated the anatomical distribution of probabilities of T2 hyperintense lesions. Overall, these findings suggest that intrinsic spatial properties and/or longstanding precursory abnormalities of normal-appearing white matter tissue may contribute to the risk of autoimmune acute demyelination in multiple sclerosis.
Collapse
Affiliation(s)
| | - Parya Momayyezsiahkal
- NeuroRx Research, Montreal, QC H2X 3P9, Canada.,McGill University, Montreal, QC H3A 0G4, Canada
| | - Douglas L Arnold
- NeuroRx Research, Montreal, QC H2X 3P9, Canada.,McGill University, Montreal, QC H3A 0G4, Canada
| | - Dawei Liu
- Biogen Digital Health, Biogen, Cambridge, MA 02142, USA
| | - Jun Ke
- Biogen, Cambridge, MA 02142, USA
| | - Li Zhu
- Biogen, Cambridge, MA 02142, USA
| | - Bing Zhu
- Biogen, Cambridge, MA 02142, USA
| | - Ilena C George
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
| | | | | | | | - Peter K Stys
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Jeroen J G Geurts
- Department of Anatomy and Neurosciences, Amsterdam UMC, 1081 HV Amsterdam, Netherlands
| | | | - Arie Gafson
- Biogen Digital Health, Biogen, Cambridge, MA 02142, USA
| | | |
Collapse
|
32
|
Beaudoin AM, Rheault F, Theaud G, Laberge F, Whittingstall K, Lamontagne A, Descoteaux M. Modern Technology in Multi-Shell Diffusion MRI Reveals Diffuse White Matter Changes in Young Adults With Relapsing-Remitting Multiple Sclerosis. Front Neurosci 2021; 15:665017. [PMID: 34447292 PMCID: PMC8383891 DOI: 10.3389/fnins.2021.665017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 07/20/2021] [Indexed: 11/13/2022] Open
Abstract
Objective To characterize microstructural white matter changes related to relapsing-remitting multiple sclerosis using advanced diffusion MRI modeling and tractography. The association between imaging data and patient’s cognitive performance, fatigue severity and depressive symptoms is also explored. Methods In this cross-sectional study, 24 relapsing-remitting multiple sclerosis patients and 11 healthy controls were compared using high angular resolution diffusion imaging (HARDI). The imaging method includes a multi-shell scheme, free water correction to obtain tissue-specific measurements, probabilistic tracking algorithm robust to crossing fibers and white matter lesions, automatic streamlines and bundle dissection and tract-profiling with tractometry. The neuropsychological evaluation included the Symbol Digit Modalities Test, Paced Auditory Serial Addition Test, Modified Fatigue Impact Scale and Beck Depression Inventory-II. Results Bundle-wise analysis by tractometry revealed a difference between patients and controls for 11 of the 14 preselected white matter bundles. In patients, free water corrected fractional anisotropy was significantly reduced while radial and mean diffusivities were increased, consistent with diffuse demyelination. The fornix and left inferior fronto-occipital fasciculus exhibited a higher free water fraction. Eight bundles showed an increase in total apparent fiber density and four bundles had a higher number of fiber orientations, suggesting axonal swelling and increased organization complexity, respectively. In the association study, depressive symptoms were associated with diffusion abnormalities in the right superior longitudinal fasciculus. Conclusion Tissue-specific diffusion measures showed abnormalities along multiple cerebral white matter bundles in patients with relapsing-remitting multiple sclerosis. The proposed methodology combines free-water imaging, advanced bundle dissection and tractometry, which is a novel approach to investigate cerebral pathology in multiple sclerosis. It opens a new window of use for HARDI-derived measures and free water corrected diffusion measures. Advanced diffusion MRI provides a better insight into cerebral white matter changes in relapsing-remitting multiple sclerosis, namely diffuse demyelination, edema and increased fiber density and complexity.
Collapse
Affiliation(s)
- Ann-Marie Beaudoin
- Department of Neurology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - François Rheault
- Sherbrooke Connectivity Imaging Laboratory (SCIL), Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Guillaume Theaud
- Sherbrooke Connectivity Imaging Laboratory (SCIL), Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Frédéric Laberge
- Sherbrooke Connectivity Imaging Laboratory (SCIL), Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Kevin Whittingstall
- Department of Radiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Albert Lamontagne
- Department of Neurology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Maxime Descoteaux
- Sherbrooke Connectivity Imaging Laboratory (SCIL), Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
33
|
Killestein J, Liguori M. Loss of Neurologic Reserve in Progressive Multiple Sclerosis: A Paradigm Shift? Neurol Clin Pract 2021; 11:271-272. [PMID: 34484925 PMCID: PMC8382419 DOI: 10.1212/cpj.0000000000001106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Joep Killestein
- Amsterdam UMC (JK), Vrije Universiteit Amsterdam, Department of Neurology, MS Center Amsterdam, Amsterdam Neuroscience, the Netherlands; and National Research Council (CNR) (ML), Institute of Biomedical Technologies, Bari Unit, Italy
| | - Maria Liguori
- Amsterdam UMC (JK), Vrije Universiteit Amsterdam, Department of Neurology, MS Center Amsterdam, Amsterdam Neuroscience, the Netherlands; and National Research Council (CNR) (ML), Institute of Biomedical Technologies, Bari Unit, Italy
| |
Collapse
|
34
|
't Hart BA, Luchicchi A, Schenk GJ, Stys PK, Geurts JJG. Mechanistic underpinning of an inside-out concept for autoimmunity in multiple sclerosis. Ann Clin Transl Neurol 2021; 8:1709-1719. [PMID: 34156169 PMCID: PMC8351380 DOI: 10.1002/acn3.51401] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/27/2021] [Accepted: 05/20/2021] [Indexed: 12/16/2022] Open
Abstract
The neuroinflammatory disease multiple sclerosis is driven by autoimmune pathology in the central nervous system. However, the trigger of the autoimmune pathogenic process is unknown. MS models in immunologically naïve, specific‐pathogen‐free bred rodents support an exogenous trigger, such as an infection. The validity of this outside–in pathogenic concept for MS has been frequently challenged by the difficulty to translate pathogenic concepts developed in these models into effective therapies for the MS patient. Studies in well‐validated non‐human primate multiple sclerosis models where, just like in humans, the autoimmune pathogenic process develops from an experienced immune system trained by prior infections, rather support an endogenous trigger. Data reviewed here corroborate the validity of this inside–out pathogenic concept for multiple sclerosis. They also provide a plausible sequence of events reminiscent of Wilkin’s primary lesion theory: (i) that autoimmunity is a physiological response of the immune system against excess antigen turnover in diseased tissue (the primary lesion) and (ii) that individuals developing autoimmune disease are (genetically predisposed) high responders against critical antigens. Data obtained in multiple sclerosis brains reveal the presence in normally appearing white matter of myelinated axons where myelin sheaths have locally dissociated from their enwrapped axon (i.e., blistering). The ensuing disintegration of axon–myelin units potentially causes the excess systemic release of post‐translationally modified myelin. Data obtained in a unique primate multiple sclerosis model revealed a core pathogenic role of T cells present in the normal repertoire, which hyper‐react to post‐translationally modified (citrullinated) myelin–oligodendrocyte glycoprotein and evoke clinical and pathological aspects of multiple sclerosis.
Collapse
Affiliation(s)
- Bert A 't Hart
- Department Anatomy and Neuroscience, University Medical Center Amsterdam, Amsterdam, The Netherlands.,Department Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center, Groningen, The Netherlands
| | - Antonio Luchicchi
- Department Anatomy and Neuroscience, University Medical Center Amsterdam, Amsterdam, The Netherlands
| | - Geert J Schenk
- Department Anatomy and Neuroscience, University Medical Center Amsterdam, Amsterdam, The Netherlands
| | - Peter K Stys
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, University of Calgary Cumming School of Medicine, Calgary, Canada
| | - Jeroen J G Geurts
- Department Anatomy and Neuroscience, University Medical Center Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
35
|
Multiple sclerosis and drug discovery: A work of translation. EBioMedicine 2021; 68:103392. [PMID: 34044219 PMCID: PMC8245896 DOI: 10.1016/j.ebiom.2021.103392] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 03/26/2021] [Accepted: 04/22/2021] [Indexed: 02/08/2023] Open
Abstract
Multiple sclerosis (MS) is after trauma the most important neurological disease in young adults, affecting 1 per 1000 individuals. With currently available medications, most of these targeting the immune system, satisfactory results have been obtained in patients with relapsing MS, but these can have serious adverse effects. Moreover, despite some promising developments, such as with B cell targeting therapies or sphingosine-1-phosphate modulating drugs, there still is a high unmet need of safe drugs with broad efficacy in patients with progressive MS. Despite substantial investments and intensive preclinical research, the proportion of promising lead compounds that reaches the approved drug status remains disappointingly low. One cause lies in the poor predictive validity of MS animal models used in the translation of pathogenic mechanisms into safe and effective treatments for the patient. This disturbing situation has raised criticism against the relevance of animal models used in preclinical research and calls for improvement of these models. This publication presents a potentially useful strategy to enhance the predictive validity of MS animal models, namely, to analyze the causes of failure in forward translation (lab to clinic) via reverse translation (clinic to lab). Through this strategy new insights can be gained that can help generate more valid MS models.
Collapse
|
36
|
Ten Bosch GJA, Bolk J, 't Hart BA, Laman JD. Multiple sclerosis is linked to MAPK ERK overactivity in microglia. J Mol Med (Berl) 2021; 99:1033-1042. [PMID: 33948692 PMCID: PMC8313465 DOI: 10.1007/s00109-021-02080-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/31/2021] [Accepted: 04/19/2021] [Indexed: 12/18/2022]
Abstract
Reassessment of published observations in patients with multiple sclerosis (MS) suggests a microglial malfunction due to inappropriate (over)activity of the mitogen-activated protein kinase pathway ERK (MAPKERK). These observations regard biochemistry as well as epigenetics, and all indicate involvement of this pathway. Recent preclinical research on neurodegeneration already pointed towards a role of MAPK pathways, in particular MAPKERK. This is important as microglia with overactive MAPK have been identified to disturb local oligodendrocytes which can lead to locoregional demyelination, hallmark of MS. This constitutes a new concept on pathophysiology of MS, besides the prevailing view, i.e., autoimmunity. Acknowledged risk factors for MS, such as EBV infection, hypovitaminosis D, and smoking, all downregulate MAPKERK negative feedback phosphatases that normally regulate MAPKERK activity. Consequently, these factors may contribute to inappropriate MAPKERK overactivity, and thereby to neurodegeneration. Also, MAPKERK overactivity in microglia, as a factor in the pathophysiology of MS, could explain ongoing neurodegeneration in MS patients despite optimized immunosuppressive or immunomodulatory treatment. Currently, for these patients with progressive disease, no effective treatment exists. In such refractory MS, targeting the cause of overactive MAPKERK in microglia merits further investigation as this phenomenon may imply a novel treatment approach.
Collapse
Affiliation(s)
- George J A Ten Bosch
- Department of Medical Oncology, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands.
| | - Jolande Bolk
- Department of Anesthesiology, Medisch Spectrum Twente, Enschede, The Netherlands
| | - Bert A 't Hart
- Department Anatomy and Neuroscience, Amsterdam University Medical Center (VUmc), Amsterdam, The Netherlands.,Department Biomedical Sciences of Cells & Systems, University Medical Center Groningen, Groningen, The Netherlands
| | - Jon D Laman
- Department Biomedical Sciences of Cells & Systems, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
37
|
Grieb P, Świątkiewicz M, Kamińska A, Jünemann A, Rejdak R, Rejdak K. Citicoline: A Candidate for Adjunct Treatment of Multiple Sclerosis. Pharmaceuticals (Basel) 2021; 14:ph14040326. [PMID: 33918331 PMCID: PMC8066453 DOI: 10.3390/ph14040326] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/24/2021] [Accepted: 03/29/2021] [Indexed: 12/13/2022] Open
Abstract
In remitting–relapsing multiple sclerosis (RR-MS), relapses are driven by autoreactive immune cells that enter the brain and spinal cord and damage myelin sheaths of axons in white and grey matter, whereas during remissions myelin is repaired by activated oligodendroglial cells. Disease-modifying therapies (DMTs) may either retard/attenuate myelin damage or promote/enhance/speed up myelin repair. Almost all currently approved DMTs inhibit myelin damage and are considerably toxic. Enhancement of myelin repair is considered an unmet medical need of MS patients. Citicoline, known for many years as a nootropic and neuroprotective drug and recently pronounced food supplement, has been found to be significantly efficacious in two complementary rodent models of MS, experimental autoimmune encephalomyelitis (EAE) and cuprizone-induced myelin toxicity. Moreover, citicoline treatment improves visual evoked potentials (VEPs) in glaucoma patients, which is relevant because VEP monitoring is frequently used as an indicator of remyelination in MS. Although over-the-counter availability of citicoline may impede its formal translation to the clinic of MS, evaluation of its efficacy for supporting remyelination in this disease is strongly indicated.
Collapse
Affiliation(s)
- Paweł Grieb
- Department of Experimental Pharmacology, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland;
| | - Maciej Świątkiewicz
- Department of Experimental Pharmacology, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland;
- Correspondence:
| | - Agnieszka Kamińska
- Faculty of Medical Sciences, Collegium Medicum, Cardinal Stefan Wyszynski University, 01-938 Warsaw, Poland;
| | - Anselm Jünemann
- Chair and Department of General and Pediatric Ophthalmology, Medical University of Lublin, 20-079 Lublin, Poland; (A.J.); (R.R.)
| | - Robert Rejdak
- Chair and Department of General and Pediatric Ophthalmology, Medical University of Lublin, 20-079 Lublin, Poland; (A.J.); (R.R.)
| | - Konrad Rejdak
- Department of Neurology, Medical University of Lublin, 20-954 Lublin, Poland;
| |
Collapse
|
38
|
Luchicchi A, Preziosa P, 't Hart B. Editorial: "Inside-Out" vs "Outside-In" Paradigms in Multiple Sclerosis Etiopathogenesis. Front Cell Neurosci 2021; 15:666529. [PMID: 33732113 PMCID: PMC7957074 DOI: 10.3389/fncel.2021.666529] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 02/11/2021] [Indexed: 11/24/2022] Open
Affiliation(s)
- Antonio Luchicchi
- Division Clinical Neurosciences, Department of Anatomy and Neurosciences, Amsterdam Universitair Medische Centra, Vrije Universiteit Medical Center, Amsterdam, Netherlands
| | - Paolo Preziosa
- Neuroimaging Research Unit, Division of Neuroscience, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy
| | - Bert 't Hart
- Division Clinical Neurosciences, Department of Anatomy and Neurosciences, Amsterdam Universitair Medische Centra, Vrije Universiteit Medical Center, Amsterdam, Netherlands
| |
Collapse
|